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










Publication year range
1.
Clin Vaccine Immunol ; 19(7): 991-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22623651

ABSTRACT

Antivector immunity has been recognized as a potential caveat of using virus-based vaccines. In the present study, an alphavirus-based replicon particle vaccine platform, which has demonstrated robust immunogenicity in animal models, was tested for effects of antivector immunity on immunogenicity against hemagglutinin of influenza virus as a target antigen and efficacy for protection against lethal challenge with the virus. Chimeric alphavirus-based replicon particles, comprising Venezuelan equine encephalitis virus nonstructural and Sindbis virus structural components, induced efficient protective antibody responses, which were not adversely influenced after multiple immunizations with the same vector expressing various antigens.


Subject(s)
Alphavirus/immunology , Genetic Vectors/immunology , Influenza Vaccines/immunology , Vaccination/methods , Alphavirus/genetics , Animals , Antibodies, Viral/blood , Drug Carriers , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Survival Analysis
2.
PLoS One ; 5(4): e10297, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421972

ABSTRACT

A variety of vaccine platforms are under study for development of new vaccines for measles. Problems with past measles vaccines are incompletely understood and underscore the need to understand the types of immune responses induced by different types of vaccines. Detailed immune response evaluation is most easily performed in mice. Although mice are not susceptible to infection with wild type or vaccine strains of measles virus, they can be used for comparative evaluation of the immune responses to measles vaccines of other types. In this study we compared the immune responses in mice to a new protective alphavirus replicon particle vaccine expressing the measles virus hemagglutinin (VEE/SIN-H) with a non-protective formalin-inactivated, alum-precipitated measles vaccine (FI-MV). MV-specific IgG levels were similar, but VEE/SIN-H antibody was high avidity IgG2a with neutralizing activity while FI-MV antibody was low-avidity IgG1 without neutralizing activity. FI-MV antibody was primarily against the nucleoprotein with no priming to H. Germinal centers appeared, peaked and resolved later for FI-MV. Lymph node MV antibody-secreting cells were more numerous after FI-MV than VEE/SIN-H, but were similar in the bone marrow. VEE/SIN-H-induced T cells produced IFN-gamma and IL-4 both spontaneously ex vivo and after stimulation, while FI-MV-induced T cells produced IL-4 only after stimulation. In summary, VEE/SIN-H induced a balanced T cell response and high avidity neutralizing IgG2a while FI-MV induced a type 2 T cell response, abundant plasmablasts, late germinal centers and low avidity non-neutralizing IgG1 against the nucleoprotein.


Subject(s)
Hemagglutinins/genetics , Immunity, Humoral , Measles Vaccine/pharmacology , Vaccines, DNA/pharmacology , Vaccines, Inactivated/pharmacology , Alphavirus/genetics , Alum Compounds/pharmacology , Animals , Antibody Affinity , Formaldehyde/pharmacology , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Genetic Vectors/therapeutic use , Hemagglutinins/administration & dosage , Hemagglutinins/therapeutic use , Immunoglobulin G/blood , Measles Vaccine/immunology , Measles Vaccine/therapeutic use , Mice , Neutralization Tests , T-Lymphocytes/immunology , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/therapeutic use
3.
J Virol ; 84(8): 3798-807, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20130066

ABSTRACT

Measles remains a major cause of child mortality, in part due to an inability to vaccinate young infants with the current live attenuated virus vaccine (LAV). To explore new approaches to infant vaccination, chimeric Venezuelan equine encephalitis/Sindbis virus (VEE/SIN) replicon particles were used to express the hemagglutinin (H) and fusion (F) proteins of measles virus (MV). Juvenile rhesus macaques vaccinated intradermally with a single dose of VEE/SIN expressing H or H and F proteins (VEE/SIN-H or VEE/SIN-H+F, respectively) developed high titers of MV-specific neutralizing antibody and gamma-interferon (IFN-gamma)-producing T cells. Infant macaques vaccinated with two doses of VEE/SIN-H+F also developed neutralizing antibody and IFN-gamma-producing T cells. Control animals were vaccinated with LAV or with a formalin-inactivated measles vaccine (FIMV). Neutralizing antibody remained above the protective level for more than 1 year after vaccination with VEE/SIN-H, VEE/SIN-H+F, or LAV. When challenged with wild-type MV 12 to 17 months after vaccination, all vaccinated juvenile and infant monkeys vaccinated with VEE/SIN-H, VEE/SIN-H+F, and LAV were protected from rash and viremia, while FIMV-vaccinated monkeys were not. Antibody was boosted by challenge in all groups. T-cell responses to challenge were biphasic, with peaks at 7 to 25 days and at 90 to 110 days in all groups, except for the LAV group. Recrudescent T-cell activity coincided with the presence of MV RNA in peripheral blood mononuclear cells. We conclude that VEE/SIN expressing H or H and F induces durable immune responses that protect from measles and offers a promising new approach for measles vaccination. The viral and immunological factors associated with long-term control of MV replication require further investigation.


Subject(s)
Alphavirus/genetics , Genetic Vectors , Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles virus/immunology , Measles/prevention & control , Viral Fusion Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Hemagglutinins, Viral/genetics , Humans , Injections, Intradermal , Interferon-gamma/metabolism , Macaca mulatta , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Measles virus/genetics , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Fusion Proteins/genetics
4.
Vaccine ; 25(3): 481-9, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17052811

ABSTRACT

Parainfluenza virus type 3 (PIV3) infections continue to be a significant health risk for infants, young children, and immunocompromised adults. We describe a gene-based vaccine strategy against PIV3 using replication-defective alphavirus vectors. These RNA replicon vectors, delivered as virus-like particles and expressing the PIV3 hemagglutinin-neuraminidase glycoprotein, were shown to be highly immunogenic in mice and hamsters, inducing PIV3-specific neutralizing antibody responses. Importantly, the replicon particle-based vaccine administered intramuscularly or intranasally protected against mucosal PIV3 challenge in hamsters, preventing virus replication in both nasal turbinates and lungs. These data suggest that the alphavirus replicon platform can be useful for a PIV3 vaccine and possibly other respiratory viruses.


Subject(s)
Alphavirus/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Virus 3, Human/immunology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/prevention & control , RNA, Viral/genetics , RNA, Viral/immunology , Replicon/genetics , Replicon/immunology , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cricetinae , Encephalitis Virus, Venezuelan Equine/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hemagglutination Inhibition Tests , Mesocricetus , Mice , Mice, Inbred BALB C , Neutralization Tests , Parainfluenza Virus 3, Human/growth & development , Sindbis Virus/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
5.
AIDS Res Hum Retroviruses ; 22(10): 993-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17067269

ABSTRACT

Mucosal and systemic transmission of HIV is prevalent. Therefore, mucosal followed by parenteral immunizations with chimeric vs. complete alphavirus-based replicon particles, encoding an HIV envelope glycoprotein, were tested. Female rhesus macaques were immunized intranasally and then intramuscularly. Following the immunizations, enhanced mucosal and systemic antibody responses were detected with the chimeric compared to the complete replicon particles. Although similar proportions of the same peripheral blood monocyte lineage target cells were infected with the chimeric vs. the complete replicon particles, the latter resulted in enhanced expression of the gene of interest, suggesting a possible mechanism of the enhanced immunogenicity.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Gene Products, env/immunology , HIV Antibodies/biosynthesis , Immunization/methods , Macaca mulatta/immunology , Replicon/immunology , Sindbis Virus/immunology , Administration, Intranasal , Animals , Chimera/immunology , Female , Immunity, Mucosal , Injections, Intramuscular , env Gene Products, Human Immunodeficiency Virus
6.
AIDS Res Hum Retroviruses ; 22(10): 1022-30, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17067273

ABSTRACT

In the present study, macaques were coimmunized with VEErep/SINenv chimeric alphavirus replicon particles expressing SIVp55Gag and HIVDeltaV2gp140Env or only with replicon particles expressing HIVDeltaV2gp140Env. All animals were subsequently immunized with recombinant trimeric HIVDeltaV2gp140Env protein. During alphavirus immunization, anti-SIVGag and anti-HIVEnv-specific interferon (IFN)-gamma responses, as well as high titers of anti-HIVEnv binding (gp120 but not gp41 specific) and anti-HIV neutralizing antibodies, were generated. The subsequent immunization with recombinant HIVDeltaV2gp140 enhanced the neutralizing antibody titers and Env-specific IFN-gamma responses. Following intravenous challenge with the R5- tropic SHIV(SF162P4) virus, significantly lower primary plasma viremia levels were recorded in the immunized animals, as compared to control animals immunized with replicon particles expressing influenza virus HA. Our results show that this method of immunization elicits both strong cellular immunity and neutralizing antibodies in primates and, thus, merits further investigation.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Viral/biosynthesis , Gene Products, env/immunology , Gene Products, gag/immunology , HIV Antibodies/biosynthesis , Replicon , Animals , Encephalitis Virus, Venezuelan Equine/genetics , Genetic Vectors , Macaca mulatta , Recombinant Proteins/immunology , Sindbis Virus , Vaccines, Synthetic/immunology , env Gene Products, Human Immunodeficiency Virus
7.
J Immunol ; 176(9): 5671-81, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16622037

ABSTRACT

Infants younger than age 9 mo do not respond reliably to the live attenuated measles vaccine due the immaturity of their immune system and the presence of maternal Abs that interfere with successful immunization. We evaluated the immune responses elicited by Sindbis virus replicon-based DNA vaccines encoding measles virus (MV) hemagglutinin (H, pMSIN-H) or both hemagglutinin and fusion (F, pMSINH-FdU) glycoproteins in neonatal mice born to naive and measles-immune mothers. Despite the presence of high levels of maternal Abs, neonatal immunization with pMSIN-H induced long-lasting, high-avidity MV plaque reduction neutralization (PRN) Abs, mainly IgG2a, that also inhibited syncytium formation in CD150(+) B95-8 cells. IgG secreting plasma cells were detected in spleen and bone marrow. Newborns vaccinated with pMSINH-FdU elicited PRN titers that surpassed the protective level (200 mIU/ml) but were short-lived, had low syncytium inhibition capacity, and lacked avidity maturation. This vaccine failed to induce significant PRN titers in the presence of placentally transferred Abs. Both pMSIN-H and pMSINH-FdU elicited strong Th1 type cell-mediated immunity, measured by T cell proliferation and IFN-gamma production, that was unaffected by maternal Abs. Newborns responded to measles DNA vaccines with similar or even higher PRN titers and cell-mediated immunity than adult mice. This study is the first demonstration that a Sindbis virus-based measles DNA vaccine can elicit robust MV immunity in neonates bypassing maternal Abs. Such a vaccine could be followed by the current live attenuated MV vaccine in a heterologous prime-boost to protect against measles early in life.


Subject(s)
Aging/immunology , Animals, Newborn/immunology , Antibodies, Viral/immunology , Immunity, Maternally-Acquired/immunology , Measles Vaccine/immunology , Sindbis Virus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/classification , Antibody Affinity/immunology , Antigens, Viral/immunology , Bone Marrow/immunology , Cell Line , Female , Hemagglutinins, Viral/immunology , Kinetics , Mice , Neutralization Tests , Spleen/immunology
8.
Vaccine ; 24(15): 2755-63, 2006 Apr 05.
Article in English | MEDLINE | ID: mdl-16460840

ABSTRACT

The worldwide HIV-1 vaccine research endeavor is focused increasingly on subtype C, which is now the predominant strain of the present HIV/AIDS epidemic. Expression cassettes of HIV-1 subtype C gag, pol and versions of gagpol fusion cassettes were constructed and evaluated for their relative abilities to induce cellular immune responses in mice. Animals were vaccinated with DNA or alphavirus replicon particle-based vaccines and cellular immune responses were measured by flow cytometry. Five new major histocompatibility complex (MHC) class I-restricted T cell epitopes in subtype C Gag and Pol were identified. Although two CD8(+) T cell epitopes within Gag were immunodominant in BALB/c and CB6F1 mice, the overall breadth of the T cell responses in mice immunized with plasmids or recombinant alphavirus replicon particles encoding gagpol fusion genes was improved over single antigen genes (i.e. gag or pol alone). The patterns of epitope dominance were consistent among mice although there were variations observed between different animals in the relative contributions of the various epitopes to the total response. These data are consistent with observations in non-human primates (Otten GR, Schaefer M, Doe B, Liu H, Magede JZ, Donnelly J, et al. Potent immunogenicity of an HIV-1 gag-pol fusion DNA vaccine delivered by in vivo electroporation. Vaccine 2005, in press) and support a subtype C in-frame gagpol fusion gene vaccine.


Subject(s)
AIDS Vaccines/immunology , Gene Products, gag/genetics , Gene Products, pol/genetics , HIV Infections/immunology , HIV-1/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , Alphavirus/genetics , Alphavirus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Female , Flow Cytometry , Gene Products, gag/immunology , Gene Products, pol/immunology , HIV Antibodies/blood , HIV-1/genetics , Lymphocyte Subsets/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Replicon , Vaccines, DNA/administration & dosage , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
9.
J Virol ; 80(1): 360-71, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16352561

ABSTRACT

In order to establish nonlytic persistent infections (PI) of BHK cells, replicons derived from Sindbis (SIN) and Semliki Forest (SFV) viruses have mutations in nsP2. Five different nsP2 PI replicons were compared to wild-type (wt) SIN, SFV, and wt nsPs SIN replicons. Replicon PI BHK21 cells had viral RNA synthesis rates that were less than 5% of those of the wt virus and approximately 10% or less of those of SIN wt replicon-infected cells, and, in contrast to wt virus and replicons containing wt nsP2, all showed a phenotype of continuous minus-strand synthesis and of unstable, mature replication/transcription complexes (RC+) that are active in plus-strand synthesis. Minus-strand synthesis and incorporation of [3H]uridine into replicative intermediates differed among PI replicons, depending on the location of the mutation in nsP2. Minus-strand synthesis by PI cells appeared normal; it was dependent on continuous P123 and P1234 polyprotein synthesis and ceased when protein synthesis was inhibited. The failure by the PI replicons to shut off minus-strand synthesis was not due to some defect in the PI cells but rather was due to the loss of some function in the mutated nsP2. This was demonstrated by showing that superinfection of PI cells with wt SFV triggered the shutdown of minus-strand synthesis, which we believe is a host response to infection with alphaviruses. Together, the results indicate alphavirus nsP2 functions to engage the host response to infection and activate a switch from the early-to-late phase. The loss of this function leads to continuous viral minus-strand synthesis and the production of unstable RC+.


Subject(s)
Alphavirus/physiology , Cysteine Endopeptidases/metabolism , RNA, Viral/biosynthesis , Replicon , Viral Nonstructural Proteins/metabolism , Alphavirus/genetics , Animals , Chickens , Cysteine Endopeptidases/genetics , Fibroblasts/virology , Gene Expression Regulation, Viral , Viral Nonstructural Proteins/biosynthesis , Virus Replication
10.
Proc Natl Acad Sci U S A ; 102(33): 11581-8, 2005 Aug 16.
Article in English | MEDLINE | ID: mdl-16037211

ABSTRACT

Measles remains a major worldwide problem partly because of difficulties with vaccination of young infants. New vaccine strategies need to be safe and to provide sustained protective immunity. We have developed Sindbis virus replicon particles that express the measles virus (MV) hemagglutinin (SIN-H) or fusion (SIN-F) proteins. In mice, SIN-H induced high-titered, dose-dependent, MV-neutralizing antibody after a single vaccination. SIN-F, or SIN-H and SIN-F combined, induced somewhat lower responses. To assess protective efficacy, juvenile macaques were vaccinated with a single dose of 10(6) or 10(8) SIN-H particles and infant macaques with two doses of 10(8) particles. A dose of 10(8) particles induced sustained levels of high-titered, MV-neutralizing antibody and IFN-gamma-producing memory T cells, and most monkeys were protected from rash when challenged with wild-type MV 18 months later. After challenge, there was a biphasic appearance of H- and F-specific IFN-gamma-secreting CD4+ and CD8+ T cells in vaccinated monkeys, with peaks approximately 1 and 3-4 months after challenge. Viremia was cleared within 14 days, but MV RNA was detectable for 4-5 months. These studies suggest that complete clearance of MV after infection is a prolonged, phased, and complex process influenced by prior vaccination.


Subject(s)
Alphavirus/genetics , Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles virus/immunology , Measles/immunology , Measles/virology , T-Lymphocytes/immunology , Aging/physiology , Alphavirus/physiology , Animals , Antibodies, Viral/immunology , Female , Hemagglutinins, Viral/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Macaca/immunology , Macaca/virology , Measles Vaccine/chemistry , Measles Vaccine/genetics , Measles virus/physiology , Mice , Mice, Inbred BALB C , RNA, Viral/genetics , Vaccination , Virion/genetics , Virion/immunology , Virus Replication
11.
J Virol ; 79(15): 9854-61, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16014946

ABSTRACT

Measles virus (MV) hemagglutinin (MV-H) and fusion (MV-F) proteins induce plaque reduction neutralizing (PRN) antibodies and cell-mediated immune responses that protect against clinical measles. DNA vaccines that encode MV-H and MV-F are being investigated as a new generation of measles vaccine to protect infants too young to receive currently licensed attenuated measles vaccines. However, it is unclear whether DNA vaccines encoding both MV-H and MV-F act synergistically to induce stronger immunity than immunization with plasmids encoding MV-H or MV-F alone. To address this question, we generated Sindbis virus-based pSINCP DNA vaccines that encode either MV-H or MV-F alone or bicistronic or fusion system vectors that encode both MV-H and MV-F (to mimic MV infection where both MV-H and MV-F proteins are expressed by the same mammalian cell). Mice immunized with DNA vaccine encoding MV-H alone developed significantly greater PRN titers than mice immunized with bicistronic constructs. Interestingly, the presence of MV-F in the bicistronic constructs stimulated serum MV-specific immunoglobulin G of reduced avidity. By contrast, mice immunized with bicistronic constructs induced equivalent or higher levels of MV-specific gamma interferon responses than mice immunized with DNA vaccine encoding MV-H alone. These data will help guide the design of DNA-based MV vaccines to be used early in life in a heterologous prime-boost strategy.


Subject(s)
Hemagglutinins, Viral/immunology , Measles Vaccine/immunology , Measles virus/immunology , Measles/immunology , Vaccination , Viral Fusion Proteins/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Affinity , Drug Evaluation, Preclinical , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Intramuscular , Interferon-gamma/biosynthesis , Measles Vaccine/administration & dosage , Mice , Mice, Inbred BALB C , Vaccines, DNA/immunology
12.
J Virol ; 79(13): 8189-200, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15956564

ABSTRACT

DNA vaccines have been used widely in experimental primate models of human immunodeficiency virus (HIV), but their effectiveness has been limited. In this study, we evaluated three technologies for increasing the potency of DNA vaccines in rhesus macaques. These included DNA encoding Sindbis virus RNA replicons (pSINCP), cationic poly(lactide-co-glycolide) (PLG) microparticles for DNA delivery, and recombinant protein boosting. The DNA-based pSINCP replicon vaccines encoding HIV Gag and Env were approximately equal in potency to human cytomegalovirus (CMV) promoter-driven conventional DNA vaccines (pCMV). The PLG microparticle DNA delivery system was particularly effective at enhancing antibody responses induced by both pCMV and pSINCP vaccines and had less effect on T cells. Recombinant Gag and Env protein boosting elicited rapid and strong recall responses, in some cases to levels exceeding those seen after DNA or DNA/PLG priming. Of note, Env protein boosting induced serum-neutralizing antibodies and increased frequencies of gamma interferon-producing CD4 T cells severalfold. Thus, PLG microparticles are an effective means of delivering DNA vaccines in nonhuman primates, as demonstrated for two different types of DNA vaccines encoding two different antigens, and are compatible for use with DNA prime-protein boost regimens.


Subject(s)
AIDS Vaccines , DNA, Viral/pharmacology , Plasmids/genetics , Vaccines, DNA , Animals , Gene Products, env/genetics , Gene Products, env/immunology , Immunization, Secondary , Lymphocyte Activation , Macaca mulatta , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/immunology
13.
J Virol ; 79(11): 7135-45, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15890953

ABSTRACT

A safe, replication-defective viral vector that can induce mucosal and systemic immune responses and confer protection against many infectious pathogens, such as human immunodeficiency virus type 1 (HIV-1), may be an ideal vaccine platform. Accordingly, we have generated and tested alphavirus replicon particles encoding HIV-1 Gag from Sindbis virus (SIN-Gag) and Venezuelan equine encephalitis virus (VEE-Gag), as well as chimeras between the two (VEE/SIN-Gag). Following intramuscular (i.m.), intranasal (i.n.), or intravaginal (IVAG) immunization with VEE/SIN-Gag and an IVAG challenge with vaccinia virus encoding HIV Gag (VV-Gag), a larger number of Gag-specific CD8+ intracellular gamma interferon-expressing cells (iIFNEC) were detected in iliac lymph nodes (ILN), which drain the vaginal/uterine mucosa (VUM), than were observed after immunizations with SIN-Gag. Moreover, a single i.n. or IVAG immunization with VEE/SIN-Gag induced a larger number of cells expressing HIV Gag in ILN, and immunizations with VEE/SIN-Gag through any route induced better protective responses than immunizations with SIN-Gag. In VUM, a larger percentage of iIFNEC expressed alpha4beta7 or alpha(Ebeta)7 integrin than expressed CD62L integrin. However, in spleens (SP), a larger percentage of iIFNEC expressed alpha4beta7 or CD62L than expressed alpha(Ebeta)7. Moreover, a larger percentage of iIFNEC expressed the chemokine receptor CCR5 in VUM and ILN than in SP. These results demonstrate a better induction of cellular and protective responses following immunizations with VEE/SIN-Gag than that following immunizations with SIN-Gag and also indicate a differential expression of homing and chemokine receptors on iIFNEC in mucosal effector and inductive sites versus systemic lymphoid tissues.


Subject(s)
Gene Products, gag/genetics , Gene Products, gag/immunology , HIV-1/genetics , HIV-1/immunology , Interferon-gamma/biosynthesis , Animals , CD8-Positive T-Lymphocytes/immunology , Chimera/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Female , Gene Products, gag/biosynthesis , Genes, gag , Genetic Vectors , HIV-1/pathogenicity , HIV-1/physiology , Immunity, Mucosal , Immunization , Mice , Mice, Inbred BALB C , Receptors, CCR5/biosynthesis , Receptors, Lymphocyte Homing/biosynthesis , Replicon , Sindbis Virus/genetics
14.
J Virol ; 78(19): 10328-35, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367599

ABSTRACT

We have expressed and characterized the severe acute respiratory syndrome coronavirus (SARS-CoV) spike protein in cDNA-transfected mammalian cells. The full-length spike protein (S) was newly synthesized as an endoglycosidase H (endo H)-sensitive glycoprotein (gp170) that is further modified into an endo H-resistant glycoprotein (gp180) in the Golgi apparatus. No substantial proteolytic cleavage of S was observed, suggesting that S is not processed into head (S1) and stalk (S2) domains as observed for certain other coronaviruses. While the expressed full-length S glycoprotein was exclusively cell associated, a truncation of S by excluding the C-terminal transmembrane and cytoplasmic tail domains resulted in the expression of an endoplasmic reticulum-localized glycoprotein (gp160) as well as a Golgi-specific form (gp170) which was ultimately secreted into the cell culture medium. Chemical cross-linking, thermal denaturation, and size fractionation analyses suggested that the full-length S glycoprotein of SARS-CoV forms a higher order structure of approximately 500 kDa, which is consistent with it being an S homotrimer. The latter was also observed in purified virions. The intracellular form of the C-terminally truncated S protein (but not the secreted form) also forms trimers, but with much less efficiency than full-length S. Deglycosylation of the full-length homotrimer with peptide N-glycosidase-F under native conditions abolished recognition of the protein by virus-neutralizing antisera raised against purified virions, suggesting the importance of the carbohydrate in the correct folding of the S protein. These data should aid in the design of recombinant vaccine antigens to prevent the spread of this emerging pathogen.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Animals , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Antigens, Viral/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Culture Media/chemistry , DNA, Complementary , DNA, Viral/genetics , DNA, Viral/metabolism , Endoplasmic Reticulum/chemistry , Glycoside Hydrolases/metabolism , Golgi Apparatus/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Molecular Weight , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Subunits/analysis , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
15.
Breast Cancer Res ; 6(4): R275-83, 2004.
Article in English | MEDLINE | ID: mdl-15217493

ABSTRACT

INTRODUCTION: The purpose of the present study was to determine whether cytotoxic chemotherapeutic agents administered prior to immunotherapy with gene vaccines could augment the efficacy of the vaccines. METHODS: Mice were injected in the mammary fat pad with an aggressive breast tumor cell line that expresses HER2/neu. The mice were treated 3 days later with a noncurative dose of either doxorubicin or paclitaxel, and the following day with a gene vaccine to HER2/neu. Two more doses of vaccine were given 14 days apart. Two types of gene vaccines were tested: a plasmid vaccine encoding a self-replicating RNA (replicon) of Sindbis virus (SINCP), in which the viral structural proteins were replaced by the gene for neu; and a viral replicon particle derived from an attenuated strain of Venezuelan equine encephalitis virus, containing a replicon RNA in which the Venezuelan equine encephalitis virus structural proteins were replaced by the gene for neu. RESULTS: Neither vaccination alone nor chemotherapy alone significantly reduced the growth of the mammary carcinoma. In contrast, chemotherapy followed by vaccination reduced tumor growth by a small, but significant amount. Antigen-specific CD8+ T lymphocytes were induced by the combined treatment, indicating that the control of tumor growth was most probably due to an immunological mechanism. The results demonstrated that doxorubicin and paclitaxel, commonly used chemotherapeutic agents for the treatment of breast cancer, when used at immunomodulating doses augmented the antitumor efficacy of gene vaccines directed against HER2/neu. CONCLUSIONS: The combination of chemotherapeutic agents plus vaccine immunotherapy may induce a tumor-specific immune response that could be beneficial for the adjuvant treatment of patients with minimal residual disease. The regimen warrants further evaluation in a clinical setting.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma/drug therapy , Carcinoma/therapy , Disease Models, Animal , Doxorubicin/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/therapy , Paclitaxel/therapeutic use , Receptor, ErbB-2/therapeutic use , Animals , Antigens, Neoplasm/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Carcinoma/immunology , Cell Line , Combined Modality Therapy/methods , Combined Modality Therapy/trends , Doxorubicin/administration & dosage , Drug Administration Schedule , Epitopes/physiology , Female , Humans , Interferon-gamma/metabolism , Lymphocyte Activation/physiology , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Paclitaxel/administration & dosage , T-Lymphocyte Subsets/physiology , T-Lymphocytes, Cytotoxic/physiology , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use
16.
J Virol ; 77(19): 10394-403, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970424

ABSTRACT

Alphavirus replicon particle-based vaccine vectors derived from Sindbis virus (SIN), Semliki Forest virus, and Venezuelan equine encephalitis virus (VEE) have been shown to induce robust antigen-specific cellular, humoral, and mucosal immune responses in many animal models of infectious disease and cancer. However, since little is known about the relative potencies among these different vectors, we compared the immunogenicity of replicon particle vectors derived from two very different parental alphaviruses, VEE and SIN, expressing a human immunodeficiency virus type 1 p55(Gag) antigen. Moreover, to explore the potential benefits of combining elements from different alphaviruses, we generated replicon particle chimeras of SIN and VEE. Two distinct strategies were used to produce particles with VEE-p55(gag) replicon RNA packaged within SIN envelope glycoproteins and SIN-p55(gag) replicon RNA within VEE envelope glycoproteins. Each replicon particle configuration induced Gag-specific CD8(+) T-cell responses in murine models when administered alone or after priming with DNA. However, Gag-specific responses varied dramatically, with the strongest responses to this particular antigen correlating with the VEE replicon RNA, irrespective of the source of envelope glycoproteins. Comparing the replicons with respect to heterologous gene expression levels and sensitivity to alpha/beta interferon in cultured cells indicated that each might contribute to potency differences. This work shows that combining desirable elements from VEE and SIN into a replicon particle chimera may be a valuable approach toward the goal of developing vaccine vectors with optimal in vivo potency, ease of production, and safety.


Subject(s)
AIDS Vaccines/immunology , Encephalitis Virus, Venezuelan Equine/genetics , Genetic Vectors , Replicon , Sindbis Virus/genetics , Vaccines, Synthetic/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Chimera , Cricetinae , Gene Products, gag/genetics , Gene Products, gag/immunology , Interferons/pharmacology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Precursors/genetics , Protein Precursors/immunology , RNA, Viral/biosynthesis , Virus Assembly
17.
J Virol ; 77(9): 5209-17, 2003 May.
Article in English | MEDLINE | ID: mdl-12692223

ABSTRACT

Measles remains a leading cause of child mortality in developing countries. Residual maternal measles antibodies and immunologic immaturity dampen immunogenicity of the current vaccine in young infants. Because cotton rat respiratory tract is susceptible to measles virus (MV) replication after intranasal (i.n.) challenge, this model can be used to assess the efficacy of MV vaccines. Pursuing a new measles vaccine strategy that might be effective in young infants, we used attenuated Salmonella enterica serovar Typhi CVD 908-htrA and Shigella flexneri 2a CVD 1208 vaccines to deliver mucosally to cotton rats eukaryotic expression plasmid pGA3-mH and Sindbis virus-based DNA replicon pMSIN-H encoding MV hemagglutinin (H). The initial i.n. dose-response with bacterial vectors alone identified a well-tolerated dosage (1 x 10(9) to 7 x 10(9) CFU) and a volume (20 micro l) that elicited strong antivector immune responses. Animals immunized i.n. on days 0, 28, and 76 with bacterial vectors carrying DNA plasmids encoding MV H or immunized parenterally with these naked DNA vaccine plasmids developed MV plaque reduction neutralizing antibodies and proliferative responses against MV antigens. In a subsequent experiment of identical design, cotton rats were challenged with wild-type MV 1 month after the third dose of vaccine or placebo. MV titers were significantly reduced in lung tissue of animals immunized with MV DNA vaccines delivered either via bacterial live vectors or parenterally. Since attenuated serovar Typhi and S. flexneri can deliver measles DNA vaccines mucosally in cotton rats, inducing measles immune responses (including neutralizing antibodies) and protection, boosting strategies can now be evaluated in animals primed with MV DNA vaccines.


Subject(s)
Measles Vaccine/immunology , Measles/prevention & control , Salmonella typhi/genetics , Shigella flexneri/genetics , Sigmodontinae , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/genetics , Antigens, Viral/immunology , Disease Models, Animal , Genetic Vectors , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immunity, Mucosal , Measles/immunology , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Neutralization Tests , Salmonella typhi/immunology , Shigella flexneri/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, DNA/administration & dosage , Viral Plaque Assay
18.
Infect Immun ; 71(1): 575-9, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12496215

ABSTRACT

The immunogenicity of a plasmid DNA vaccine incorporating Sindbis virus RNA replicase functions (pSINCP) and expressing antigen 85A (Ag85A) from Mycobacterium tuberculosis was compared with a conventional plasmid DNA vector encoding Ag85A. pSINCP-85A was highly immunogenic in mice and gave enhanced long-term protection against M. tuberculosis compared with the conventional vector.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , Plasmids , Replicon , Sindbis Virus , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Acyltransferases/genetics , Animals , Antigens, Bacterial/genetics , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Plasmids/genetics , Plasmids/immunology , RNA-Dependent RNA Polymerase/genetics , Replicon/genetics , Replicon/immunology , Sindbis Virus/genetics , Sindbis Virus/immunology , Tuberculosis Vaccines/administration & dosage , Vaccination , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
19.
Drug Discov Today ; 7(13): 719-27, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12110228

ABSTRACT

Vaccinology has experienced a dramatic resurgence recently, as traditional methodologies of using attenuated live pathogens or inactivated whole pathogens have been either ineffective or are not an acceptable risk for several disease targets, including HIV and Hepatitis C. Gene-based vaccines can stimulate potent humoral and cellular immune responses, and viral vectors might be an efficient strategy for both delivery of antigen-encoding genes, as well as facilitating and enhancing antigen presentation. Vectors derived from diverse viruses with distinct tropism and gene expression strategies have been developed, and are being evaluated in preclinical and clinical vaccine studies. Virus-based vaccines represent a promising approach for vaccines against infectious and malignant disease.


Subject(s)
Genetic Vectors , Vaccines/therapeutic use , Viruses , Humans , Poliovirus/genetics , Vaccines/genetics
20.
Hum Gene Ther ; 13(4): 553-68, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11874633

ABSTRACT

Alphavirus vectors have emerged as a strategy for the development of cancer vaccines and gene therapy applications. The availability of a new packaging cell line (PCL), which is capable of generating alphavirus replicon particles without contamination from replication-competent virus, has advanced the field of vaccine development. This replication-defective vaccine vector has potential advantages over naked nucleic acid vaccines, such as increased efficiency of gene delivery and large-scale production. We have developed a new strategy to enhance nucleic acid vaccine potency by linking VP22, a herpes simplex virus type 1 (HSV-1) tegument protein, to a model antigen. This strategy facilitated the spread of linked E7 antigen to neighboring cells. In this study, we created a recombinant Sindbis virus (SIN)-based replicon particle encoding VP22 linked to a model tumor antigen, human papillomavirus type 16 (HPV-16) E7, using a stable SIN PCL. The linkage of VP22 to E7 in these SIN replicon particles resulted in a significant increase in the number of E7-specific CD8(+) T cell precursors and a strong antitumor effect against E7-expressing tumors in vaccinated C57BL/6 mice relative to wild-type E7 SIN replicon particles. Furthermore, a head-to-head comparison of VP22-E7-containing naked DNA, naked RNA replicons, or RNA replicon particle vaccines indicated that SINrep5-VP22/E7 replicon particles generated the most potent therapeutic antitumor effect. Our results indicated that the VP22 strategy used in the context of SIN replicon particles may facilitate the generation of a highly effective vaccine for widespread immunization.


Subject(s)
Cancer Vaccines , Immunotherapy , Neoplasms/therapy , Oncogene Proteins, Viral/immunology , Replicon/immunology , Sindbis Virus/immunology , Viral Proteins/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Replicon/genetics , Sindbis Virus/physiology , Vaccines, DNA , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL