Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Vaccine ; 34(46): 5629-5635, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27670072

ABSTRACT

In order for vaccines to induce efficacious immune responses against mucosally transmitted pathogens, such as HIV-1, activated lymphocytes must efficiently migrate to and enter targeted mucosal sites. We have previously shown that all-trans retinoic acid (ATRA) can be used as a vaccine adjuvant to enhance mucosal CD8+ T cell responses during vaccination and improve protection against mucosal viral challenge. However, the ATRA formulation is incompatible with most recombinant vaccines, and the teratogenic potential of ATRA at high doses limits its usage in many clinical settings. We hypothesized that increasing in vivo production of retinoic acid (RA) during vaccination with a DNA vector expressing retinaldehyde dehydrogenase 2 (RALDH2), the rate-limiting enzyme in RA biosynthesis, could similarly provide enhanced programming of mucosal homing to T cell responses while avoiding teratogenic effects. Administration of a RALDH2- expressing plasmid during immunization with a HIVgag DNA vaccine resulted in increased systemic and mucosal CD8+ T cell numbers with an increase in both effector and central memory T cells. Moreover, mice that received RALDH2 plasmid during DNA vaccination were more resistant to intravaginal challenge with a recombinant vaccinia virus expressing the same HIVgag antigen (VACVgag). Thus, RALDH2 can be used as an alternative adjuvant to ATRA during DNA vaccination leading to an increase in both systemic and mucosal T cell immunity and better protection from viral infection at mucosal sites.


Subject(s)
AIDS Vaccines/immunology , Adjuvants, Immunologic , Immunity, Mucosal , Retinal Dehydrogenase/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Aldehyde Dehydrogenase 1 Family , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Human Immunodeficiency Virus Proteins/administration & dosage , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/immunology , Immunization/methods , Immunologic Memory , Mice , Plasmids , Retinal Dehydrogenase/administration & dosage , Retinal Dehydrogenase/genetics , Tretinoin/immunology , Tretinoin/metabolism , Vaccines, DNA/administration & dosage , Vaccinia/immunology , Vaccinia/prevention & control , Vaccinia virus/genetics
2.
Methods Mol Biol ; 890: 161-76, 2012.
Article in English | MEDLINE | ID: mdl-22688767

ABSTRACT

Preclinical evaluation of novel anti-smallpox vaccines and antiviral treatments often rely on mouse -challenge models using pathogenic vaccinia virus, such as Western Reserve (WR) strain or other orthopoxviruses. Traditionally, efficacy of treatment is evaluated using various readouts, such as lethality (rare), measurements of body weight loss, pox lesion scoring, and determination of viral loads in internal organs by enumerating plaques in sensitive cell lines. These methodologies provide valuable information about the contribution of the treatment to protection from infection, yet all have similar limitations: they do not evaluate dissemination of the virus within the same animal and require large numbers of animals. These two problems prompted us to turn to a recently developed whole body imaging technology, where replication of recombinant vaccinia virus expressing luciferase enzyme (WRvFire) is sensed by detecting light emitted by the enzyme in the presence of D: -luciferin substrate administered to infected animal. Bioluminescence signals from infected organs in live animals are registered by the charge-coupled device camera in IVIS instrument developed by Caliper, and are converted into numerical values. This chapter describes whole body bioimaging methodology used to determine viral loads in normal live BALB/c mice infected with recombinant WRvFire vaccinia virus. Using Dryvax vaccination as a model, we show how bioluminescence data can be used to determine efficacy of treatment. In addition, we illustrate how bioluminescence and survival outcome can be combined in Receiver Operating Characteristic curve -analysis to develop predictive models of lethality that can be applied for testing of new therapeutics and second-generation vaccines.


Subject(s)
Antiviral Agents/therapeutic use , Smallpox Vaccine/administration & dosage , Vaccinia virus/physiology , Vaccinia/pathology , Animals , Antigens, Viral/administration & dosage , Antiviral Agents/pharmacology , Area Under Curve , Drug Evaluation, Preclinical/methods , Firefly Luciferin/administration & dosage , Genes, Reporter , Liver/virology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Luminescent Measurements , Lung/virology , Mice , Mice, Inbred BALB C , Nasal Cavity/virology , ROC Curve , Spleen/virology , Vaccination , Vaccinia/drug therapy , Vaccinia/prevention & control , Vaccinia virus/immunology , Vaccinia virus/metabolism , Whole Body Imaging
3.
J Virol ; 83(2): 1115-25, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19004937

ABSTRACT

While the smallpox vaccine, Dryvax or Dryvax-derived ACAM2000, holds potential for public immunization against the spread of smallpox by bioterror, there is serious concern about Dryvax-mediated side effects. Here, we report that a single-dose vaccination regimen comprised of Dryvax and an antiviral agent, cidofovir, could reduce vaccinia viral loads after vaccination and significantly control Dryvax vaccination side effects. However, coadministration of cidofovir and Dryvax also reduced vaccine-elicited immune responses of antibody and T effector cells despite the fact that the reduced priming could be boosted as a recall response after monkeypox virus challenge. Evaluations of four different aspects of vaccine efficacy showed that coadministration of cidofovir and Dryvax compromised the Dryvax-induced immunity against monkeypox, although the covaccinated monkeys exhibited measurable protection against monkeypox compared to that of naïve controls. Thus, the single-dose coadministration of cidofovir and Dryvax effectively controlled vaccination side effects but significantly compromised vaccine-elicited immune responses and vaccine-induced immunity to monkeypox.


Subject(s)
Antiviral Agents/administration & dosage , Cytosine/analogs & derivatives , Mpox (monkeypox)/prevention & control , Organophosphonates/administration & dosage , Smallpox Vaccine/adverse effects , Smallpox Vaccine/immunology , Vaccinia/prevention & control , Animals , Antibodies, Viral/immunology , Cidofovir , Cytosine/administration & dosage , Drug Incompatibility , Macaca fascicularis , Smallpox Vaccine/administration & dosage , T-Lymphocytes/immunology
4.
J Virol ; 81(24): 13392-402, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928345

ABSTRACT

The bioterror threat of a smallpox outbreak in an unvaccinated population has mobilized efforts to develop new antipoxviral agents. By screening a library of known drugs, we identified 13 compounds that inhibited vaccinia virus replication at noncytotoxic doses. The anticancer drug mitoxantrone is unique among the inhibitors identified in that it has no apparent impact on viral gene expression. Rather, it blocks processing of viral structural proteins and assembly of mature progeny virions. The isolation of mitoxantrone-resistant vaccinia strains underscores that a viral protein is the likely target of the drug. Whole-genome sequencing of mitoxantrone-resistant viruses pinpointed missense mutations in the N-terminal domain of vaccinia DNA ligase. Despite its favorable activity in cell culture, mitoxantrone administered intraperitoneally at the maximum tolerated dose failed to protect mice against a lethal intranasal infection with vaccinia virus.


Subject(s)
Antineoplastic Agents , Antiviral Agents , Mitoxantrone , Vaccinia virus/drug effects , Virion/metabolism , Virus Assembly/drug effects , Virus Replication/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cell Line , Drug Evaluation, Preclinical , Drug Resistance, Viral , Female , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mitoxantrone/administration & dosage , Mitoxantrone/pharmacology , Mitoxantrone/therapeutic use , Specific Pathogen-Free Organisms , Vaccinia/prevention & control , Vaccinia/virology , Vaccinia virus/genetics
5.
Article in Russian | MEDLINE | ID: mdl-16279533

ABSTRACT

On the basis of comparative experimental evaluation of specific features in the course of the vaccinal process after the immunization of laboratory animals with live smallpox vaccines, intended for oral use (in tablets) and for skin scarification was proposed. In experiments on rabbits, made with the use of virological and immunological methods, the counteraction of the elements constituting the vaccinal process was analyzed, the integral evaluation of its course was given, the greater safety of the oral preparation in comparison with the traditional vaccine for immunization by skin-scarification method were established. The conclusion was made that oral immunization was the safest immunization method under modern conditions and promising one for using live vaccines with population immunity being at a low level or absent.


Subject(s)
Smallpox Vaccine/administration & dosage , Vaccinia/prevention & control , Administration, Cutaneous , Administration, Oral , Animals , Drug Evaluation, Preclinical/methods , Gastric Mucosa/immunology , Gastric Mucosa/virology , Lymph Nodes/virology , Rabbits , Skin/immunology , Skin/virology , Smallpox Vaccine/adverse effects , Tablets/administration & dosage , Vaccinia/virology , Vaccinia virus/isolation & purification , Viscera/virology
6.
J Virol ; 79(20): 13139-49, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16189015

ABSTRACT

ST-246 is a low-molecular-weight compound (molecular weight = 376), that is potent (concentration that inhibited virus replication by 50% = 0.010 microM), selective (concentration of compound that inhibited cell viability by 50% = >40 microM), and active against multiple orthopoxviruses, including vaccinia, monkeypox, camelpox, cowpox, ectromelia (mousepox), and variola viruses. Cowpox virus variants selected in cell culture for resistance to ST-246 were found to have a single amino acid change in the V061 gene. Reengineering this change back into the wild-type cowpox virus genome conferred resistance to ST-246, suggesting that V061 is the target of ST-246 antiviral activity. The cowpox virus V061 gene is homologous to vaccinia virus F13L, which encodes a major envelope protein (p37) required for production of extracellular virus. In cell culture, ST-246 inhibited plaque formation and virus-induced cytopathic effects. In single-cycle growth assays, ST-246 reduced extracellular virus formation by 10 fold relative to untreated controls, while having little effect on the production of intracellular virus. In vivo oral administration of ST-246 protected BALB/c mice from lethal infection, following intranasal inoculation with 10x 50% lethal dose (LD(50)) of vaccinia virus strain IHD-J. ST-246-treated mice that survived infection acquired protective immunity and were resistant to subsequent challenge with a lethal dose (10x LD(50)) of vaccinia virus. Orally administered ST-246 also protected A/NCr mice from lethal infection, following intranasal inoculation with 40,000x LD(50) of ectromelia virus. Infectious virus titers at day 8 postinfection in liver, spleen, and lung from ST-246-treated animals were below the limits of detection (<10 PFU/ml). In contrast, mean virus titers in liver, spleen, and lung tissues from placebo-treated mice were 6.2 x 10(7), 5.2 x 10(7), and 1.8 x 10(5) PFU/ml, respectively. Finally, oral administration of ST-246 inhibited vaccinia virus-induced tail lesions in Naval Medical Research Institute mice inoculated via the tail vein. Taken together, these results validate F13L as an antiviral target and demonstrate that an inhibitor of extracellular virus formation can protect mice from orthopoxvirus-induced disease.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Indoles/pharmacology , Orthopoxvirus/drug effects , Poxviridae Infections/prevention & control , Administration, Oral , Amino Acid Sequence , Animals , Antiviral Agents/adverse effects , Antiviral Agents/chemistry , Benzamides/adverse effects , Benzamides/chemistry , Cytopathogenic Effect, Viral/drug effects , Drug Evaluation, Preclinical , Ectromelia virus/isolation & purification , Ectromelia, Infectious/prevention & control , Female , Indoles/adverse effects , Indoles/chemistry , Isoindoles , Liver/virology , Lung/virology , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molecular Weight , Orthopoxvirus/isolation & purification , Orthopoxvirus/physiology , Poxviridae Infections/virology , Sequence Alignment , Spleen/virology , Vaccinia/prevention & control , Viral Envelope Proteins/drug effects , Viral Envelope Proteins/genetics , Viral Plaque Assay , Virus Assembly/drug effects
7.
J Immunol ; 171(8): 4028-39, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530323

ABSTRACT

We evaluated immunogenicity of a novel Th-CTL fusion peptide composed of the pan DR Th epitope and a CTL epitope derived from HIV-pol in two transgenic HLA-A*0201/K(b) mouse models. The immunogenicity of peptides of this structure is highly dependent on coadministered cytosine-phosphate-guanine DNA. Initial evaluations of peptide-specific immunity are based on results of chromium release assay, intracellular cytokine, and tetramer staining. Significant cytotoxic T cell responses are found upon a single immunization with as low as 0.1 nmol both peptide and cytosine-phosphate-guanine DNA. Splenocytes from immunized mice recognize naturally processed HIV-pol expressed from vaccinia virus (pol-VV). Translation of immunologic criteria into more relevant assays was pursued using systemic challenge of immunized mice with pol-VV. Only mice receiving both peptide and DNA together successfully cleared upward of 6 logs of virus from ovaries, compared with controls. Challenge with pol-VV by intranasal route of intranasal immunized mice showed a significant reduction in the levels of VV in lung compared with naive mice. A convincing demonstration of the relevance of these vaccines is the robust lysis of HIV-infected Jurkat T cells (JA2/R7/Hyg) by immune splenocytes from peptide- and DNA-immunized mice. This surprisingly effective immunization merits consideration for clinical evaluation, because it succeeded in causing immune recognition and lysis of cells infected with its target virus and reduction in titer of highly pathogenic VV.


Subject(s)
AIDS Vaccines/immunology , Antigen Presentation , CpG Islands/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/genetics , Recombinant Fusion Proteins/immunology , Vaccines, DNA/immunology , Vaccinia/prevention & control , AIDS Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Animals , Antigen Presentation/genetics , Cells, Cultured , Coculture Techniques , Epitopes, T-Lymphocyte/administration & dosage , HIV Core Protein p24/biosynthesis , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HLA-A2 Antigen/immunology , Humans , Immunity, Mucosal/genetics , Injections, Intraperitoneal , Interferon-gamma/biosynthesis , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Jurkat Cells , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Nasal Mucosa/immunology , Nasal Mucosa/virology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Recombinant Fusion Proteins/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/virology , Vaccines, DNA/administration & dosage , Vaccinia/immunology
8.
J Immunol ; 168(6): 2880-6, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11884458

ABSTRACT

Virus-like particles (VLPs) are known to induce strong Ab responses in the absence of adjuvants. In addition, VLPs are able to prime CTL responses in vivo. To study the efficiency of this latter process, we fused peptide p33 derived from lymphocytic choriomeningitis virus to the hepatitis B core Ag, which spontaneously assembles into VLPs (p33-VLPs). These p33-VLPs were efficiently processed in vitro and in vivo for MHC class I presentation. Nevertheless, p33-VLPs induced weak CTL responses that failed to mediate effective protection from viral challenge. However, if APCs were activated concomitantly in vivo using either anti-CD40 Abs or CpG oligonucleotides, the CTL responses induced were fully protective against infection with lymphocytic choriomeningitis virus or recombinant vaccinia virus. Moreover, these CTL responses were comparable to responses generally induced by live vaccines, because they could be measured in primary ex vivo (51)Cr release assays. Thus, while VLPs alone are inefficient at inducing CTL responses, they become very powerful vaccines if applied together with substances that activate APCs.


Subject(s)
Antigen-Presenting Cells/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Vaccines/immunology , Virion/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antigen Presentation/genetics , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Antigens, Viral/immunology , CD40 Antigens/immunology , Chromium Radioisotopes , Cytotoxicity Tests, Immunologic , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Glycoproteins/administration & dosage , Glycoproteins/genetics , Glycoproteins/immunology , Hepatitis B Core Antigens/genetics , Hepatitis B Core Antigens/immunology , Injections, Intradermal , Injections, Subcutaneous , L Cells , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/virology , Tumor Cells, Cultured , Vaccinia/prevention & control , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virion/genetics
9.
J Clin Lab Immunol ; 32(4): 177-81, 1990 Aug.
Article in English | MEDLINE | ID: mdl-1966934

ABSTRACT

Non-specific protective activities against vaccinia virus (VV) and Pseudomonas aeruginosa infections as well as interferon (IFN)-inducing, natural killer (NK) cell and macrophage activation activities of chemically synthesized lipid A-subunit analogs were investigated. The analogs are 4-O-phosphono-D-glucosamine derivatives carrying different 2-N- and 3-O-linked acyl substituents such as (R)-3-tetradecanoyloxytetradecanoyl (C14-O-(C14)), (R)-3-hydroxytetradecanoyl (C14-OH) and tetradecanoyl (C14) groups. Compounds GLA-59 and GLA-60, which possess C14-OH and C14-O-(C14) groups as their acyl substituents, showed stronger IFN-inducing and anti-vaccinia virus activities than GLA-27 and GLA-68, which possess a C14 group instead of the C14-OH group in GLA-59 and GLA-60, although NK cell activation activity was similarly high in all of these compounds. In protective activity against P. aeruginosa infection and macrophage activation activity, GLA-60 and GLA-68, which carry a C14-O-(C14) group at the 3-O-position, expressed higher activities than GLA-27 and GLA-59, which carry the acyloxyacyl group at the 2-N-position. These results indicate that the acyl substituent (whether the counterpart of the C14-O-(C14) group is a C14 or a C14-OH group) and the binding position of the acyloxyacyl group at the 2-N- or the 3-O-position strongly influence the manifestation of antimicrobial and immunomodulating activities in different ways depending on the activity. Among the compounds, GLA-60 satisfied the structure requirements for protection against both VV and P. aeruginosa infections. This compound is a hopeful immunomodulator for prevention against broad microbial infections.


Subject(s)
Glucosamine/analogs & derivatives , Lipid A/analogs & derivatives , Pseudomonas Infections/prevention & control , Vaccinia/prevention & control , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/physiology , Animals , Drug Resistance , Glucosamine/pharmacology , Lipid A/pharmacology , Mice , Mice, Inbred ICR , Structure-Activity Relationship
10.
Acta Virol ; 23(6): 500-3, 1979 Nov.
Article in English | MEDLINE | ID: mdl-44666

ABSTRACT

The potential in vivo antiviral activity of 3-[bis-(2-hydroxyethyl)-amino]-acetophenone-[4,5-diphenyl-oxazolyl-(2)]-hydrazone (IMET 98/69) was evaluated on model infections in mice. Animals treated subcutaneously (s.c.) with 1 mmole of the drug per kg body weight once daily for five days were significantly protected against a lethal infection with cardioviruses, Semliki forest virus and vaccinia virus. In influenza A and B virus models no antiviral activity was observed either after s.c. or oral treatment.


Subject(s)
Antiviral Agents/therapeutic use , Virus Diseases/prevention & control , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Arbovirus Infections/prevention & control , Disease Models, Animal , Drug Evaluation, Preclinical , Encephalomyocarditis virus , Enterovirus Infections/prevention & control , Female , Injections, Subcutaneous , Male , Mice , Orthomyxoviridae Infections/prevention & control , Oxazoles/analogs & derivatives , Oxazoles/therapeutic use , RNA Viruses , Semliki forest virus , Vaccinia/prevention & control
11.
Acta Virol ; 20(5): 402-10, 1976 Oct.
Article in English | MEDLINE | ID: mdl-11668

ABSTRACT

A new mathematical method based on 3-, 4- or 6-point parallel-line assays was applied for working out a computer programme used to evaluate the activity of some new isatin-beta-thiosemicarbazone derivatives against vaccinia virus, studied in vivo.


Subject(s)
Antiviral Agents , Drug Evaluation, Preclinical/methods , Thiosemicarbazones/pharmacology , Vaccinia virus/drug effects , Animals , Antiviral Agents/administration & dosage , Chemical Phenomena , Chemistry , Computers , Female , Male , Methisazone/pharmacology , Mice , Thiosemicarbazones/administration & dosage , Vaccinia/prevention & control
12.
Arch Virol ; 52(1-2): 151-8, 1976.
Article in English | MEDLINE | ID: mdl-187146

ABSTRACT

Carbopol, a polymer of acrylic acid cross-linked with allylsucrose, was found to impart resistance to virus infection in mice which received an intraperitoneal injection of the polycarboxylate 1 or 4 days before either intravenous vaccinia virus challenge or intranasal herpes simplex virus challenge. An interferon-like substance was detected in the blood stream 20-40 hours after the intraperitoneal injection of carbopol. The antiviral activity of this cross-linked polycarboxylate could be due to activation of peritoneal macrophages and/or interferon production.


Subject(s)
Antiviral Agents/therapeutic use , Encephalitis/prevention & control , Herpes Simplex/prevention & control , Polyvinyls/therapeutic use , Vaccinia/prevention & control , Animals , Ascitic Fluid/cytology , Carboxylic Acids/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Injections, Intraperitoneal , Interferons/biosynthesis , Mice , Simplexvirus/drug effects , Tail , Vaccinia virus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL