Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Viruses ; 13(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34960621

ABSTRACT

West Nile virus (WNV) and Usutu virus (USUV) are mosquito-borne flaviviruses that can cause neuroinvasive disease in humans. WNV and USUV circulate in both Africa and Europe and are closely related. Due to antigenic similarity, WNV-specific antibodies and USUV-specific antibodies have the potential to bind heterologous viruses; however, it is unclear whether this interaction may offer protection against infection. To investigate how prior WNV exposure would influence USUV infection, we used an attenuated WNV vaccine that contains the surface proteins of WNV in the backbone of a dengue virus 2 vaccine strain and protects against WNV disease. We hypothesized that vaccination with this attenuated WNV vaccine would protect against USUV infection. Neutralizing responses against WNV and USUV were measured in vitro using sera following vaccination. Sera from vaccinated CD-1 and Ifnar1-/- mice cross-neutralized with WNV and USUV. All mice were then subsequently challenged with an African or European USUV strain. In CD-1 mice, there was no difference in USUV titers between vaccinated and mock-vaccinated mice. However, in the Ifnar1-/- model, vaccinated mice had significantly higher survival rates and significantly lower USUV viremia compared to mock-vaccinated mice. Our results indicate that exposure to an attenuated form of WNV protects against severe USUV disease in mice and elicits a neutralizing response to both WNV and USUV. Future studies will investigate the immune mechanisms responsible for the protection against USUV infection induced by WNV vaccination, providing critical insight that will be essential for USUV and WNV vaccine development.


Subject(s)
Flavivirus Infections/prevention & control , Flavivirus/immunology , West Nile Virus Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Male , Mice , Mice, Knockout , Vaccination
2.
Front Immunol ; 12: 730346, 2021.
Article in English | MEDLINE | ID: mdl-34566991

ABSTRACT

Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cell Degranulation/drug effects , Immunity, Mucosal/drug effects , Mast Cells/drug effects , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile virus/pathogenicity , Administration, Intranasal , Animals , Cell Line , Disease Models, Animal , Drug Discovery , Female , High-Throughput Screening Assays , Host-Pathogen Interactions , Immunity, Mucosal/genetics , Immunization , Immunogenicity, Vaccine , Mast Cells/immunology , Mast Cells/virology , Mice, Inbred BALB C , Proof of Concept Study , West Nile Fever/genetics , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/immunology
3.
Viruses ; 11(9)2019 09 05.
Article in English | MEDLINE | ID: mdl-31491885

ABSTRACT

Although West Nile virus (WNV) has been a prominent mosquito-transmitted infection in North America for twenty years, no human vaccine has been licensed. With a cumulative number of 24,714 neurological disease cases and 2314 deaths in the U.S. since 1999, plus a large outbreak in Europe in 2018 involving over 2000 human cases in 15 countries, a vaccine is essential to prevent continued morbidity, mortality, and economic burden. Currently, four veterinary vaccines are licensed, and six vaccines have progressed into clinical trials in humans. All four veterinary vaccines require multiple primary doses and annual boosters, but for a human vaccine to be protective and cost effective in the most vulnerable older age population, it is ideal that the vaccine be strongly immunogenic with only a single dose and without subsequent annual boosters. Of six human vaccine candidates, the two live, attenuated vaccines were the only ones that elicited strong immunity after a single dose. As none of these candidates have yet progressed beyond phase II clinical trials, development of new candidate vaccines and improvement of vaccination strategies remains an important area of research.


Subject(s)
West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Clinical Trials, Phase II as Topic/history , Drug Development/history , History, 21st Century , Humans , West Nile Fever/immunology , West Nile Fever/virology , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile Virus Vaccines/history , West Nile virus/genetics
4.
Math Biosci ; 305: 60-70, 2018 11.
Article in English | MEDLINE | ID: mdl-30171883

ABSTRACT

A discrete-time model describing the west nile virus transmission among the mosquito, wild bird, and domestic bird populations is constructed. The expressions for the basic reproduction number R0 and the disease-free fixed point of the model are computed. The local stability of the disease-free fixed point is established based on R0. Optimal control theory is used to devise the most effective administration profile of mosquito larvicide, mosquito adulticide and domestic bird-protection in controlling the virus transmission among the mosquito - wild bird - domestic bird community.


Subject(s)
Models, Biological , West Nile Fever/transmission , Animals , Birds/immunology , Birds/virology , Computer Simulation , Culex/virology , Culicidae/virology , Disease Vectors , Female , Humans , Insect Bites and Stings/virology , Insecticides/administration & dosage , Mathematical Concepts , Mosquito Vectors/virology , Time Factors , West Nile Fever/prevention & control , West Nile Fever/virology , West Nile Virus Vaccines/administration & dosage , West Nile virus/immunology , West Nile virus/pathogenicity
5.
Vaccine ; 35(23): 3143-3151, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28456529

ABSTRACT

BACKGROUND: West Nile virus (WNV) is the leading cause of domestically-acquired arboviral disease in the United States. Several WNV vaccines are in various stages of development. We estimate the cost-effectiveness of WNV vaccination programs targeting groups at increased risk for severe WNV disease. METHODS: We used a mathematical model to estimate costs and health outcomes of vaccination with WNV vaccine compared to no vaccination among seven cohorts, spaced at 10year intervals from ages 10 to 70years, each followed until 90-years-old. U.S. surveillance data were used to estimate WNV neuroinvasive disease incidence. Data for WNV seroprevalence, acute and long-term care costs of WNV disease patients, quality-adjusted life-years (QALYs), and vaccine characteristics were obtained from published reports. We assumed vaccine efficacy to either last lifelong or for 10years with booster doses given every 10years. RESULTS: There was a statistically significant difference in cost-effectiveness ratios across cohorts in both models and all outcomes assessed (Kruskal-Wallis test p<0.0001). The 60-year-cohort had a mean cost per neuroinvasive disease case prevented of $664,000 and disability averted of $1,421,000 in lifelong model and $882,000 and $1,887,000, respectively in 10-year immunity model; these costs were statistically significantly lower than costs for other cohorts (p<0.0001). Vaccinating 70-year-olds had the lowest cost per death averted in both models at around $4.7 million (95%CI $2-$8 million). Cost per disease case averted was lowest among 40- and 50-year-old cohorts and cost per QALY saved lowest among 60-year cohorts in lifelong immunity model. The models were most sensitive to disease incidence, vaccine cost, and proportion of persons developing disease among infected. CONCLUSIONS: Age-based WNV vaccination program targeting those at higher risk for severe disease is more cost-effective than universal vaccination. Annual variation in WNV disease incidence, QALY weights, and vaccine costs impact the cost effectiveness ratios.


Subject(s)
Immunization Programs/economics , West Nile Virus Vaccines/economics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Cost-Benefit Analysis , Female , Humans , Immunization, Secondary/economics , Male , Markov Chains , Middle Aged , Monte Carlo Method , Quality-Adjusted Life Years , Risk Factors , Seroepidemiologic Studies , United States/epidemiology , Vaccination/economics , West Nile Fever/epidemiology , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , Young Adult
6.
J Infect Dis ; 215(1): 52-55, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-28077583

ABSTRACT

West Nile virus (WNV) is a major cause of mosquito-borne illness in the United States. Human disease ranges from mild febrile illness to severe fatal neurologic infection. Adults aged >60 years are more susceptible to neuroinvasive disease accompanied by a high mortality rate or long-lasting neurologic sequelae. A chimeric live attenuated West Nile virus vaccine, rWN/DEN4Δ30, was shown to be safe and immunogenic in healthy adults aged 18-50 years. This study evaluated rWN/DEN4Δ30 in flavivirus-naive adults aged 50-65 years and found it to be safe and immunogenic. Outbreaks of WNV infection tend to be unpredictable, and a safe and effective vaccine will be an important public health tool.


Subject(s)
West Nile Virus Vaccines/adverse effects , West Nile Virus Vaccines/immunology , Adult , Age Factors , Aged , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Outbreaks , Female , Humans , Male , Middle Aged , Seroconversion , United States , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Viremia , West Nile Fever/epidemiology , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile virus/immunology
7.
Vaccine ; 35(2): 283-292, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27919629

ABSTRACT

West Nile virus (WNV) is a mosquito-transmitted pathogen with a wide geographical range that can lead to long-term disability and death in some cases. Despite the public health risk posed by WNV, including an estimated 3 million infections in the United States alone, no vaccine is available for use in humans. Here, we present a scaled manufacturing approach for production of a hydrogen peroxide-inactivated whole virion WNV vaccine, termed HydroVax-001WNV. Vaccination resulted in robust virus-specific neutralizing antibody responses and protection against WNV-associated mortality in mice or viremia in rhesus macaques (RM). A GLP-compliant toxicology study performed in rats demonstrated an excellent safety profile with clinical findings limited to minor and transient irritation at the injection site. An in vitro relative potency (IVRP) assay was developed and shown to correlate with in vivo responses following forced degradation studies. Long-term in vivo potency comparisons between the intended storage condition (2-8°C) and a thermally stressed condition (40±2°C) demonstrated no loss in vaccine efficacy or protective immunity over a 6-month span of time. Together, the positive pre-clinical findings regarding immunogenicity, safety, and stability indicate that HydroVax-001WNV is a promising vaccine candidate.


Subject(s)
West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , Animals , Anti-Infective Agents, Local/metabolism , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Stability , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Hot Temperature , Hydrogen Peroxide/metabolism , Macaca mulatta , Male , Mice, Inbred BALB C , Rats, Sprague-Dawley , Survival Analysis , United States , Vaccine Potency , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/immunology , Vaccines, Inactivated/isolation & purification , Viremia/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/adverse effects , West Nile Virus Vaccines/isolation & purification
8.
Vaccine ; 34(46): 5479-5482, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27670075

ABSTRACT

A crucial issue in vaccine development is to balance safety with immunogenicity. The low immunogenicity of most subunit antigens warrants a search for adjuvants able to stimulate both cell-mediated and humoral immunity. In recent years, successful applications of nanotechnology and bioengineering in the field of vaccine development have enabled the production of novel adjuvant technologies. In this work, we investigated totally synthetic and supramolecular peptide hydrogels as novel vaccine adjuvants in conjunction with the immunoprotective envelope protein domain III (EIII) of West Nile virus as an immunogen in a mouse model. Our results indicate that, compared to the clinically approved adjuvant alum, peptide hydrogel adjuvanted antigen elicited stronger antibody responses and conferred significant protection against mortality after virus challenge. The high chemical definition and biocompatibility of self-assembling peptide hydrogels makes them attractive as immune adjuvants for the production of subunit vaccines against viral and bacterial infections where antibody-mediated protection is desirable.


Subject(s)
Adjuvants, Immunologic , Antibodies, Viral/immunology , Hydrogels , Peptides/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Immunity, Cellular , Immunity, Humoral , Mice , Protein Domains/immunology , Th1 Cells/immunology , Vaccines, Subunit/immunology , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/chemistry , West Nile virus/chemistry
9.
Vaccine ; 34(44): 5366-5375, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27637937

ABSTRACT

West Nile virus (WNV) is an important agent of human encephalitis that has quickly become endemic across much of the United States since its identification in North America in 1999. While the majority (∼75%) of infections are subclinical, neurologic disease can occur in a subset of cases, with outcomes including permanent neurologic damage and death. Currently, there are no WNV vaccines approved for use in humans. This study introduces a novel vaccine platform for WNV to reduce viral replication in the central nervous system while maintaining peripheral replication to elicit strong neutralizing antibody titers. Vaccine candidates were engineered to incorporate microRNA (miRNA) target sequences for a cognate miRNA expressed only in neurons, allowing the host miRNAs to target viral transcription through endogenous RNA silencing. To maintain stability, these targets were incorporated in multiple locations within the 3'-untranslated region, flanking sequences essential for viral replication without affecting the viral open reading frame. All candidates replicated comparably to wild type WNV in vitro within cells that did not express the cognate miRNA. Insertional control viruses were also capable of neuroinvasion and neurovirulence in vivo in CD-1 mice. Vaccine viruses were safe at all doses tested and did not demonstrate mutations associated with a reversion to virulence when serially passaged in mice. All vaccine constructs were protective from lethal challenge in mice, producing 93-100% protection at the highest dose tested. Overall, this is a safe and effective attenuation strategy with broad potential application for vaccine development.


Subject(s)
MicroRNAs , Virus Replication , West Nile Virus Vaccines/genetics , West Nile Virus Vaccines/immunology , West Nile virus/physiology , 3' Untranslated Regions , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Brain/virology , DNA Replication , Disease Models, Animal , Mice , Mutagenesis, Insertional , Neurons/physiology , Neurons/virology , Open Reading Frames , RNA Interference , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile virus/genetics , West Nile virus/pathogenicity
10.
Vaccine ; 33(43): 5764-5771, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26424604

ABSTRACT

Hypersensitivity reactions, such as hives or fatal anaphylactic shock, in response to vaccination constitute a health hazard for horses that develop allergies to vaccine components. In such horses vaccination with viral vaccines stimulates an IgE response to non-target antigens. Viral vaccines share contaminating non-target proteins, such as bovine serum albumin (BSA); these antigens can stimulate IgE production with each exposure. We hypothesized that the addition of a CpG oligodeoxynucleotide (ODN) administered in conjunction with a West Nile virus vaccine would decrease the IgE response; through up-regulation of T regulatory cells and T helper 1 cells thus decreasing the potential to induce a type 1 hypersensitivity response. Thirty adult horses were injected with either CpG ODN or control GpC ODN with a killed WNV vaccine. T regulatory cell numbers and BSA specific IgE concentrations were determined pre and post vaccination. Multicolor flow cytometry was used to evaluate expression of CD4, CD25, and intracellular Foxp3 on PBMCs. Serum concentrations of BSA specific IgE were determined by ELISA. Cell culture supernatants from BSA re-stimulated lymphocytes were evaluated for concentrations of IL-2, IL-4, IL-10, and IFN-γ. The inclusion of the CpG ODN significantly increased the differentiation of T regulatory cells in response to antigen in vitro and in vivo. A significant inverse correlation was found between T regulatory cell numbers and serum BSA specific IgE concentrations. These results suggest that we can provide a safer alternate vaccination strategy, particularly for horses that have demonstrated a pro-allergic phenotype.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Allergens/immunology , Immunoglobulin E/blood , Oligodeoxyribonucleotides/administration & dosage , T-Lymphocytes, Regulatory/immunology , West Nile Virus Vaccines/immunology , Allergens/administration & dosage , Animals , Drug Contamination , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Horses , Immunophenotyping , Male , Th1 Cells/immunology , West Nile Virus Vaccines/administration & dosage
11.
Clin Vaccine Immunol ; 22(9): 1040-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26178384

ABSTRACT

Although experimental data regarding cross-protection of horse West Nile virus (WNV) vaccines against lineage 2 infections exist, the cross-protective efficacy of these vaccines under field conditions has not been demonstrated. This study was conducted to evaluate the capability of an inactivated lineage 1 vaccine (Equip WNV) to protect against natural infections from the Nea Santa-Greece-2010 lineage 2 strain. In total, 185 WNV-seronegative horses in Thessaloniki, Greece, were selected during 2 consecutive years (2011 and 2012); 140 were immunized, and 45 were used as controls. Horses were examined for signs compatible with WNV infection. Neutralizing antibody titers against the Greek strain and the PaAn001/France lineage 1 strain were determined in immunized horses. WNV circulation was detected during both years in the study area. It was estimated that 37% and 27% of the horses were infected during 2011 and 2012, respectively. Three control animals developed clinical signs, and the WNV diagnosis was confirmed. Signs related to WNV infection were not observed in the vaccinated animals. The nonvaccinated animals had a 7.58% ± 1.82% higher chance of exhibiting signs than immunized animals (P < 0.05). Neutralizing antibodies raised against both strains in all immunized horses were detectable 1 month after the initial vaccination course. The cross-protective capacity of the lowest titer (1:40) was evident in 19 animals which were subsequently infected and did not exhibit signs. Neutralizing antibodies were detectable until the annual booster, when strong anamnestic responses were observed (geometrical mean titer ratio [GMTR] for lineage 1 of 30.2; GMTR for lineage 2 of 27.5). The results indicate that Equip WNV is capable of inducing cross-protection against natural infections from a virulent lineage 2 WNV strain in horses.


Subject(s)
Cross Protection , Horse Diseases/prevention & control , West Nile Fever/veterinary , West Nile Virus Vaccines/immunology , West Nile virus/immunology , West Nile virus/pathogenicity , Amino Acid Sequence , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Greece , Horse Diseases/immunology , Horse Diseases/virology , Horses , Immunization Schedule , Sequence Alignment , Time Factors , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Fever/virology , West Nile Virus Vaccines/administration & dosage , West Nile virus/classification
12.
J Gen Virol ; 95(Pt 10): 2176-2191, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24958626

ABSTRACT

West Nile virus (WNV; genus Flavivirus, family Flaviviridae) is an emerging pathogenic arbovirus responsible for outbreaks of encephalitis around the world. Whilst no vaccines are currently available to prevent WNV infection of humans, the use of cDNA copies of flavivirus RNA genomes with large internal deletions within the capsid (C) appears promising. C-deleted vaccines are able to replicate and secrete large amounts of non-infectious immunogenic subviral particles (SVPs) from transfected cells. We have previously generated a WNV DNA vaccine candidate pKUNdC/C where C-deleted WNV cDNA was placed under the control of one copy of the cytomegalovirus (CMV) promoter and the C gene was placed under the control of a second copy of the CMV promoter in the same plasmid DNA. This DNA was shown to generate single-round infectious particles (SRIPs) capable of delivering self-replicating C-deleted RNA producing SVPs to surrounding cells, thus enhancing the vaccine potential. However, the amounts of both SRIPs and SVPs produced from pKUNdC/C DNA were relatively low. In this investigation, we aimed at increasing SRIP production by optimizing trans-C expression via incorporating different forms of C and the use of a more powerful promoter. The construct containing an elongation factor EF1α promoter encoding an extended form of C was demonstrated to produce the highest titres of SRIPs and was immunogenic in mice. Additionally, SRIP and SVP titres were further improved via incorporation of a glycosylation motif in the envelope protein. The optimized DNA yielded ~100-fold greater titres of SRIPs than the original construct, thus providing a promising candidate for further vaccine evaluation.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/immunology , Gene Expression , Vaccines, DNA/immunology , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Female , Mice , Promoter Regions, Genetic , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile virus/genetics
13.
Expert Rev Vaccines ; 13(5): 589-608, 2014 May.
Article in English | MEDLINE | ID: mdl-24689659

ABSTRACT

West Nile virus (WNV) is a mosquito-borne flavivirus that has become endemic in the United States. From 1999-2012, there have been 37088 reported cases of WNV and 1549 deaths, resulting in a 4.2% case-fatality rate. Despite development of effective WNV vaccines for horses, there is no vaccine to prevent human WNV infection. Several vaccines have been tested in preclinical studies and to date there have been eight clinical trials, with promising results in terms of safety and induction of antiviral immunity. Although mass vaccination is unlikely to be cost effective, implementation of a targeted vaccine program may be feasible if a safe and effective vaccine can be brought to market. Further evaluation of new and advanced vaccine candidates is strongly encouraged.


Subject(s)
Immunization Programs/trends , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile virus , Animals , Humans , Immunization Programs/methods , West Nile Fever/epidemiology , West Nile virus/drug effects
14.
Toxins (Basel) ; 6(4): 1397-418, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24759174

ABSTRACT

West Nile virus (WNV) causes potentially fatal neuroinvasive disease and persists at endemic levels in many parts of the world. Despite advances in our understanding of WNV pathogenesis, there remains a significant need for a human vaccine. The domain III (DIII) region of the WNV envelope protein contains epitopes that are the target of neutralizing antibodies. We have constructed a chimeric fusion of the non-toxic cholera toxin (CT) CTA2/B domains to DIII for investigation as a novel mucosally-delivered WNV vaccine. Purification and assembly of the chimera, as well as receptor-binding and antigen delivery, were verified by western blot, GM1 ELISA and confocal microscopy. Groups of BALB/c mice were immunized intranasally with DIII-CTA2/B, DIII, DIII mixed with CTA2/B, or CTA2/B control, and boosted at 10 days. Analysis of serum IgG after 14 and 45 days revealed that mucosal immunization with DIII-CTA2/B induced significant DIII-specific humoral immunity and drove isotype switching to IgG2a. The DIII-CTA2/B chimera also induced antigen-specific IgM and IgA responses. Bactericidal assays indicate that the DIII-CTA2/B immunized mice produced DIII-specific antibodies that can trigger complement-mediated killing. A dose escalation resulted in increased DIII-specific serum IgG titers on day 45. DIII antigen alone, in the absence of adjuvant, also induced significant systemic responses after intranasal delivery. Our results indicate that the DIII-CTA2/B chimera is immunogenic after intranasal delivery and merits further investigation as a novel WNV vaccine candidate.


Subject(s)
Antibodies, Viral/blood , Cholera Toxin/immunology , Immunoglobulin G/blood , Viral Envelope Proteins/immunology , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibody-Dependent Cell Cytotoxicity , Chlorocebus aethiops , Cholera Toxin/administration & dosage , Cholera Toxin/genetics , Complement Activation , Female , Immunization Schedule , Immunization, Secondary , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Time Factors , Vaccines, Synthetic/immunology , Vero Cells , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/genetics , West Nile virus/genetics
15.
Viruses ; 5(12): 3048-70, 2013 Dec 09.
Article in English | MEDLINE | ID: mdl-24351795

ABSTRACT

Substantial success has been achieved in the development and implementation of West Nile (WN) vaccines for horses; however, no human WN vaccines are approved. This review focuses on the construction, pre-clinical and clinical characterization of ChimeriVax-WN02 for humans, a live chimeric vaccine composed of a yellow fever (YF) 17D virus in which the prM-E envelope protein genes are replaced with the corresponding genes of the WN NY99 virus. Pre-clinical studies demonstrated that ChimeriVax-WN02 was significantly less neurovirulent than YF 17D in mice and rhesus and cynomolgus monkeys. The vaccine elicited neutralizing antibody titers after inoculation in hamsters and monkeys and protected immunized animals from lethal challenge including intracerebral inoculation of high dose of WN NY99 virus. Safety, viremia and immunogenicity of ChimeriVax-WN02 were assessed in one phase I study and in two phase II clinical trials. No safety signals were detected in the three clinical trials with no remarkable differences in incidence of adverse events (AEs) between vaccine and placebo recipients. Viremia was transient and the mean viremia levels were low. The vaccine elicited strong and durable neutralizing antibody and cytotoxic T cell responses. WN epidemiology impedes a classical licensure pathway; therefore, innovative licensure strategies should be explored.


Subject(s)
Drug Carriers , Genetic Vectors , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Yellow fever virus/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Clinical Trials as Topic , Cricetinae , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Macaca fascicularis , Mice , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/adverse effects , West Nile Virus Vaccines/genetics , West Nile virus/genetics
16.
Vaccine ; 31(48): 5772-7, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-23968769

ABSTRACT

WNV has become the leading vector-borne cause of meningoencephalitis in the United States. Although the majority of WNV infections result in asymptomatic illness, approximately 20% of infections result in West Nile fever and 1% in West Nile neuroinvasive disease (WNND), which causes encephalitis, meningitis, or flaccid paralysis. The elderly are at particular risk for WNND, with more than half the cases occurring in persons older than sixty years of age. There is no licensed treatment for WNND, nor is there any licensed vaccine for humans for the prevention of WNV infection. The Laboratory of Infectious Diseases at the National Institutes of Health has developed a recombinant live attenuated WNV vaccine based on chimerization of the wild-type WNV NY99 genome with that of the live attenuated DENV-4 candidate vaccine rDEN4Δ30. The genes encoding the prM and envelope proteins of DENV-4 were replaced with those of WNV NY99 and the resultant virus was designated rWN/DEN4Δ30. The vaccine was evaluated in healthy flavivirus-naïve adult volunteers age 18-50 years in two separate studies, both of which are reported here. The first study evaluated 10³ or 104 PFU of the vaccine given as a single dose; the second study evaluated 105 PFU of the vaccine given as two doses 6 months apart. The vaccine was well-tolerated and immunogenic at all three doses, inducing seroconversion to WNV NY99 in 74% (10³ PFU), 75% (104 PFU), and 55% (105 PFU) of subjects after a single dose. A second 105 PFU dose of rWN/DEN4Δ30 given 6 months after the first dose increased the seroconversion rate 89%. Based on the encouraging results from these studies, further evaluation of the candidate vaccine in adults older than 50 years of age is planned.


Subject(s)
West Nile Virus Vaccines/adverse effects , West Nile Virus Vaccines/immunology , Adolescent , Adult , Antibodies, Viral/blood , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , National Institutes of Health (U.S.) , Placebos/administration & dosage , United States , Vaccination/adverse effects , Vaccination/methods , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , West Nile Virus Vaccines/administration & dosage , Young Adult
17.
Vaccine ; 31(41): 4523-7, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23933372

ABSTRACT

West Nile virus (WNV) is maintained in nature in an enzootic transmission cycle between birds and mosquitoes, although it occasionally infects other vertebrates, including humans, in which it may result fatal. To date, no licensed vaccines against WNV infection are available for birds, but its availability would certainly benefit certain populations, as birds grown for restocking, hunting activities, or alimentary purposes, and those confined to wildlife reservations and recreation installations. We have tested the protective capability of WNV envelope recombinant (rE) protein in red-legged partridges (Alectoris rufa). Birds (n=28) were intramuscularly immunized three times at 2-weeks interval with rE and a control group (n=29) was sham-immunized. Except for 5 sham-immunized birds that were not infected and housed as contact controls, partridges were subcutaneously challenged with WNV. Oropharyngeal and cloacal swabs and feather pulps were collected at several days after infection and blood samples were taken during vaccination and after infection. All rE-vaccinated partridges elicited anti-WNV antibodies before challenge and survived to the infection, while 33.3% of the sham-immunized birds succumbed, as did 25% of the contact animals. Most (84%) unvaccinated birds showed viremia 3 d.p.i., but virus was only detected in 14% of the rE vaccinated birds. WNV-RNA was detected in feathers and swabs from sham-immunized partridges from 3 to 7 d.p.i., mainly in birds that succumbed to the infection, but not in rE vaccinated birds. Thus, rE vaccination fully protected partridges against WND and reduced the risk of virus spread.


Subject(s)
Bird Diseases/prevention & control , Viral Envelope Proteins/immunology , West Nile Fever/veterinary , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , Antibodies, Viral/blood , Bird Diseases/immunology , Blood/virology , Cloaca/virology , Galliformes , Injections, Intramuscular , Oropharynx/virology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Survival Analysis , Vaccination/methods , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Envelope Proteins/genetics , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage
18.
Vaccine ; 31(7): 1045-50, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23277093

ABSTRACT

West Nile virus (WNV) is an emergent pathogen in the Americas, first reported in New York during 1999, and has since spread across the USA, Central and South America causing neurological disease in humans, horses and some bird species, including domestic geese. No WNV vaccines are licensed in the USA for use in geese. This study reports the development of a domestic goose vaccine efficacy model, based on utilizing multiple parameters to determine protection. To test the model, 47 geese were divided in two experiments, testing five different vaccine groups and two sham groups (challenged and unchallenged). Based on the broad range of results for individual metrics between the Challenged-Sham and Unchallenged-Sham groups, the best parameters to measure protection were Clinical Pathogenicity Index (CPI), plasma virus positive geese on days 1-4 post-inoculation and plasma virus titers, and brain histological lesion rates and severity scores. Compared to the Challenged-Sham group, the fowlpox virus vectored vaccine with inserts of WNV prM and E proteins (vFP2000) provided the best protection with significant differences in all five metrics, followed by the canarypox virus vectored vaccine with inserts of WNV prM and E proteins (vCP2018) with four metrics of protection, recombinant vCP2017 with three metrics and WNV E protein with one. These data indicate that domestic geese can be used in an efficacy model for vaccine protection studies using clinical, plasma virological and brain histopathological parameters to evaluate protection against WNV challenge.


Subject(s)
Geese , Vaccination/methods , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/immunology , Animals , Brain/pathology , Brain/virology , Disease Models, Animal , Histocytochemistry , Viral Load , Viremia/prevention & control , West Nile Fever/pathology , West Nile Fever/virology
19.
J Virol ; 87(4): 1926-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23221549

ABSTRACT

West Nile virus (WNV) is an emerging pathogen that is now the leading cause of mosquito-borne and epidemic encephalitis in the United States. In humans, a small percentage of infected individuals develop severe neuroinvasive disease, with the greatest relative risk being in the elderly and immunocompromised, two populations that are difficult to immunize effectively with vaccines. While inactivated and subunit-based veterinary vaccines against WNV exist, currently there is no vaccine or therapy available to prevent or treat human disease. Here, we describe the generation and preclinical efficacy of a hydrogen peroxide (H(2)O(2))-inactivated WNV Kunjin strain (WNV-KUNV) vaccine as a candidate for further development. Both young and aged mice vaccinated with H(2)O(2)-inactivated WNV-KUNV produced robust adaptive B and T cell immune responses and were protected against stringent and lethal intracranial challenge with a heterologous virulent North American WNV strain. Our studies suggest that the H(2)O(2)-inactivated WNV-KUNV vaccine is safe and immunogenic and may be suitable for protection against WNV infection in vulnerable populations.


Subject(s)
Disinfectants/pharmacology , Hydrogen Peroxide/pharmacology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology , West Nile virus/immunology , Animals , B-Lymphocytes/immunology , Immunity, Cellular , Immunity, Humoral , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Survival Analysis , T-Lymphocytes/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Virus Inactivation , West Nile Fever/immunology , West Nile Virus Vaccines/administration & dosage
20.
Vaccine ; 30(52): 7615-20, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23088888

ABSTRACT

Vaccination of horses is performed annually or semi-annually with multiple viral antigens, either in a combination vaccine or as separate injections. While this practice undoubtedly prevents infection from such diseases as rabies, equine influenza, West Nile virus, and equine herpes virus, the procedure is not without repercussions. Hypersensitivity reactions, including fatal anaphylactic shock, after vaccination, although uncommon, have increased in incidence in recent years. Studies reported herein document the development of IgE antibodies against non-target antigen components of equine viral vaccines. We hypothesize that viral vaccines can induce an IgE response to non-target antigens, which could elicit an adverse response after vaccination with another viral vaccine containing the same component. In one study IgE responses to components of West Nile virus vaccine were evaluated by ELISA before and after vaccination in 30 horses. In a second five-year study 77 horses were similarly tested for IgE antibodies against bovine serum albumin (BSA), a component of most viral vaccines. Mast cell sensitization was evaluated in horses with high, moderate, and negative serum BSA specific IgE using an intradermal skin test with BSA. Over the five-year period high IgE responder horses showed gradually increasing BSA specific serum IgE levels and positive skin test reactivity, yet none had an adverse event. Sera from horses that had developed adverse vaccine reactions were also tested for IgE antibodies. Several of these horses had extremely high levels of BSA-specific IgE. These data suggest that non-essential protein components of vaccines may sensitize horses for future adverse responses to vaccination.


Subject(s)
Excipients/adverse effects , Immunoglobulin E/blood , Serum Albumin, Bovine/immunology , West Nile Virus Vaccines/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Excipients/administration & dosage , Horses , Serum Albumin, Bovine/administration & dosage , Serum Albumin, Bovine/adverse effects , West Nile Virus Vaccines/administration & dosage , West Nile Virus Vaccines/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...