Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Ther ; 29(9): 2782-2793, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34058388

ABSTRACT

We present a live-attenuated RNA hybrid vaccine technology that uses an RNA vaccine delivery vehicle to deliver in vitro-transcribed, full-length, live-attenuated viral genomes to the site of vaccination. This technology allows ready manufacturing in a cell-free environment, regardless of viral attenuation level, and it promises to avoid many safety and manufacturing challenges of traditional live-attenuated vaccines. We demonstrate this technology through development and testing of a live-attenuated RNA hybrid vaccine against Chikungunya virus (CHIKV), comprised of an in vitro-transcribed, highly attenuated CHIKV genome delivered by a highly stable nanostructured lipid carrier (NLC) formulation as an intramuscular injection. We demonstrate that single-dose immunization of immunocompetent C57BL/6 mice results in induction of high CHIKV-neutralizing antibody titers and protection against mortality and footpad swelling after lethal CHIKV challenge.


Subject(s)
Antibodies, Neutralizing/blood , Chikungunya Fever/prevention & control , Chikungunya virus/genetics , Lipids/chemistry , mRNA Vaccines/administration & dosage , Animals , Antibodies, Viral/blood , Chikungunya Fever/immunology , Chikungunya virus/immunology , Chlorocebus aethiops , Disease Models, Animal , Drug Compounding , Female , Genome, Viral , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Nanostructures , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/immunology , mRNA Vaccines/chemistry , mRNA Vaccines/immunology
2.
Eur J Immunol ; 49(2): 266-276, 2019 02.
Article in English | MEDLINE | ID: mdl-30548475

ABSTRACT

Influenza A annually infects 5-10% of the world's human population resulting in one million deaths. Influenza causes annual epidemics and reinfects previously exposed individuals because of antigenic drift in the glycoprotein hemagglutinin. Due to antigenic drift, the immune system is simultaneously exposed to novel and conserved parts of the influenza virus via vaccination and/or infection throughout life. Preexisting immunity has long been known to augment subsequent hemagglutination inhibitory antibody (hAb) responses. However, the preexisting immunological contributors that influence hAb responses are not understood. Therefore, we adapted and developed sequential infection and immunization mouse models using drifted influenza strains to show that MHC Class II haplotype and T-cell reactivity influences subsequent hAb responses. We found that CB6F1 mice infected with A/CA followed by immunization with A/PR8 have increased hAb responses to A/PR8 compared to C57BL/6 mice. Increased hAb responses in CB6F1 mice were CD4+  T-cell and B-cell dependent and corresponded to increased germinal center A/PR8-specific B and T-follicular helper cells. These results suggest conserved MHC Class II restricted epitopes within HA are essential for B cells to respond to drifting influenza and could be leveraged to boost hAb responses.


Subject(s)
Antibodies, Viral/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunization , Immunologic Memory , Influenza A virus/immunology , Animals , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Female , Mice
3.
Mol Ther ; 26(10): 2507-2522, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30078765

ABSTRACT

Since the first demonstration of in vivo gene expression from an injected RNA molecule almost two decades ago,1 the field of RNA-based therapeutics is now taking significant strides, with many cancer and infectious disease targets entering clinical trials.2 Critical to this success has been advances in the knowledge and application of delivery formulations. Currently, various lipid nanoparticle (LNP) platforms are at the forefront,3 but the encapsulation approach underpinning LNP formulations offsets the synthetic and rapid-response nature of RNA vaccines.4 Second, limited stability of LNP formulated RNA precludes stockpiling for pandemic readiness.5 Here, we show the development of a two-vialed approach wherein the delivery formulation, a highly stable nanostructured lipid carrier (NLC), can be manufactured and stockpiled separate from the target RNA, which is admixed prior to administration. Furthermore, specific physicochemical modifications to the NLC modulate immune responses, either enhancing or diminishing neutralizing antibody responses. We have combined this approach with a replicating viral RNA (rvRNA) encoding Zika virus (ZIKV) antigens and demonstrated a single dose as low as 10 ng can completely protect mice against a lethal ZIKV challenge, representing what might be the most potent approach to date of any Zika vaccine.


Subject(s)
Antigens, Viral/administration & dosage , Lipids/administration & dosage , Nanoparticles/administration & dosage , Zika Virus Infection/therapy , Animals , Antigens, Viral/genetics , Disease Models, Animal , Drug Delivery Systems , Humans , Lipids/chemistry , Mice , Nanoparticles/chemistry , RNA, Viral/genetics , RNA, Viral/immunology , Virus Replication/drug effects , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/genetics , Zika Virus Infection/virology
4.
Mol Ther ; 22(3): 575-587, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24419083

ABSTRACT

As sentinels of the immune system, dendritic cells (DCs) play an essential role in regulating cellular immune responses. One of the main challenges of developing DC-targeted therapies includes the delivery of antigen to DCs in order to promote the activation of antigen-specific effector CD8 T cells. With the goal of creating antigen-directed immunotherapeutics that can be safely administered directly to patients, Immune Design has developed a platform of novel integration-deficient lentiviral vectors that target and deliver antigen-encoding nucleic acids to human DCs. This platform, termed ID-VP02, utilizes a novel genetic variant of a Sindbis virus envelope glycoprotein with posttranslational carbohydrate modifications in combination with Vpx, a SIVmac viral accessory protein, to achieve efficient targeting and transduction of human DCs. In addition, ID-VP02 incorporates safety features in its design that include two redundant mechanisms to render ID-VP02 integration-deficient. Here, we describe the characteristics that allow ID-VP02 to specifically transduce human DCs, and the advances that ID-VP02 brings to conventional third-generation lentiviral vector design as well as demonstrate upstream production yields that will enable manufacturing feasibility studies to be conducted.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lentivirus/genetics , Sindbis Virus/genetics , Viral Envelope Proteins/genetics , Genetic Vectors/administration & dosage , HEK293 Cells , Humans , Immunity, Cellular/immunology , Tissue Distribution
5.
Proc Natl Acad Sci U S A ; 109(43): 17585-90, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23045649

ABSTRACT

Extensive preparation is underway to mitigate the next pandemic influenza outbreak. New vaccine technologies intended to supplant egg-based production methods are being developed, with recombinant hemagglutinin (rHA) as the most advanced program for preventing seasonal and avian H5N1 Influenza. Increased efforts are being focused on adjuvants that can broaden vaccine immunogenicity against emerging viruses and maximize vaccine supply on a worldwide scale. Here, we test protection against avian flu by using H5N1-derived rHA and GLA-SE, a two-part adjuvant system containing glucopyranosyl lipid adjuvant (GLA), a formulated synthetic Toll-like receptor 4 agonist, and a stable emulsion (SE) of oil in water, which is similar to the best-in-class adjuvants being developed for pandemic flu. Notably, a single submicrogram dose of rH5 adjuvanted with GLA-SE protects mice and ferrets against a high titer challenge with H5N1 virus. GLA-SE, relative to emulsion alone, accelerated induction of the primary immune response and broadened its durability against heterosubtypic H5N1 virus challenge. Mechanistically, GLA-SE augments protection via induction of a Th1-mediated antibody response. Innate signaling pathways that amplify priming of Th1 CD4 T cells will likely improve vaccine performance against future outbreaks of lethal pandemic flu.


Subject(s)
Adjuvants, Immunologic/chemistry , Influenza Vaccines/chemical synthesis , Influenza, Human/prevention & control , Animals , Antibodies, Viral/biosynthesis , Female , Ferrets , Humans , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred BALB C
6.
J Infect Dis ; 205(3): 474-85, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22158704

ABSTRACT

Host innate immunity is the first line of defense against invading pathogens, including influenza viruses. Ferrets are well recognized as the best model of influenza virus pathogenesis and transmission, but little is known about the innate immune response of ferrets after infection with this virus. The goal of this study was to investigate the contribution of localized host responses to influenza virus pathogenicity and transmissibility in this model by measuring the level of messenger RNA expression of 12 cytokines and chemokines in the upper and lower respiratory tracts of ferrets infected with H5N1, H1N1, or H3N2 influenza viruses that exhibit diverse virulence and transmissibility in ferrets. We found a strong temporal correlation between inflammatory mediators and the kinetics and frequency of transmission, clinical signs associated with transmission, peak virus shedding, and virulence. Our findings point to a link between localized innate immunity and influenza virus transmission and disease progression.


Subject(s)
Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae Infections/transmission , Animals , Cytokines/biosynthesis , Disease Models, Animal , Ferrets , Gene Expression Profiling , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Respiratory System/immunology , Respiratory System/virology , Virulence , Virus Shedding
7.
Proc Natl Acad Sci U S A ; 106(9): 3366-71, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19211790

ABSTRACT

The influenza virus genes that confer efficient transmission of epidemic and pandemic strains in humans have not been identified. The rapid spread and severe disease caused by the 1918 influenza pandemic virus makes it an ideal virus to study the transmissibility of potentially pandemic influenza strains. Here, we used a series of human 1918-avian H1N1 influenza reassortant viruses to identify the genetic determinants that govern airborne transmission of avian influenza viruses. We have demonstrated that the 1918 HA gene was necessary for efficient direct contact transmission, but did not allow respiratory droplet transmission between ferrets of an avian influenza virus possessing an avian polymerase subunit PB2. The 1918 PB2 protein was found to be both necessary and sufficient for airborne transmission of a virus expressing the 1918 HA and neuraminidase. Also, it was found that influenza viruses that were able to transmit efficiently in ferrets were able to replicate efficiently at the lower temperature (33 degrees C) found in the environment of mammalian airway. These findings demonstrate that the adaptation of the HA and PB2 proteins are critical for the development of pandemic influenza strains from avian influenza viruses.


Subject(s)
Air Microbiology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H1N1 Subtype/metabolism , Influenza in Birds/transmission , Influenza in Birds/virology , Influenza, Human/virology , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Animals , Birds/virology , Cell Line , Dogs , Ferrets/virology , Glycoproteins/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/transmission , Male , Mutation/genetics , Protein Subunits/metabolism , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics
8.
J Infect Dis ; 204(10): 1491-9, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21957153

ABSTRACT

Continued H5N1 virus infection in humans highlights the need for vaccine strategies that provide cross-clade protection against this rapidly evolving virus. We report a comparative evaluation in ferrets of the immunogenicity and cross-protective efficacy of isogenic mammalian cell-grown, live attenuated influenza vaccine (LAIV) and adjuvanted, whole-virus, inactivated influenza vaccine (IIV), produced from a clade 1 H5N1 6:2 reassortant vaccine candidate (caVN1203-Len17rg) based on the cold-adapted A/Leningrad/134/17/57 (H2N2) master donor virus. Two doses of LAIV or IIV provided complete protection against lethal homologous H5N1 virus challenge and a reduction in virus shedding and disease severity after heterologous clade 2.2.1 H5N1 virus challenge and increased virus-specific serum and nasal wash antibody levels. Although both vaccines demonstrated cross-protective efficacy, LAIV induced higher levels of nasal wash IgA and reduction of heterologous virus shedding, compared with IIV. Thus, enhanced respiratory tract antibody responses elicited by LAIV were associated with improved cross-clade protection.


Subject(s)
Cross Protection/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Animals , Antibodies, Viral/analysis , Ferrets , Influenza A Virus, H5N1 Subtype/genetics , Male , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology , Virus Cultivation/methods , Virus Shedding
9.
Mol Ther Methods Clin Dev ; 25: 205-214, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35308783

ABSTRACT

Current RNA vaccines against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) are limited by instability of both the RNA and the lipid nanoparticle delivery system, requiring storage at -20°C or -70°C and compromising universally accessible vaccine distribution. This study demonstrates the thermostability and adaptability of a nanostructured lipid carrier (NLC) delivery system for RNA vaccines that has the potential to address these concerns. Liquid NLC alone is stable at refrigerated temperatures for ≥1 year, enabling stockpiling and rapid deployment by point-of-care mixing with any vaccine RNA. Alternatively, NLC complexed with RNA may be readily lyophilized and stored at room temperature for ≥8 months or refrigerated temperature for ≥21 months while still retaining the ability to express protein in vivo. The thermostability of this NLC/RNA vaccine delivery platform could significantly improve distribution of current and future pandemic response vaccines, particularly in low-resource settings.

10.
Proc Natl Acad Sci U S A ; 105(21): 7558-63, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18508975

ABSTRACT

Avian H7 influenza viruses from both the Eurasian and North American lineage have caused outbreaks in poultry since 2002, with confirmed human infection occurring during outbreaks in The Netherlands, British Columbia, and the United Kingdom. The majority of H7 infections have resulted in self-limiting conjunctivitis, whereas probable human-to-human transmission has been rare. Here, we used glycan microarray technology to determine the receptor-binding preference of Eurasian and North American lineage H7 influenza viruses and their transmissibility in the ferret model. We found that highly pathogenic H7N7 viruses from The Netherlands in 2003 maintained the classic avian-binding preference for alpha2-3-linked sialic acids (SA) and are not readily transmissible in ferrets, as observed previously for highly pathogenic H5N1 viruses. However, H7N3 viruses isolated from Canada in 2004 and H7N2 viruses from the northeastern United States isolated in 2002-2003 possessed an HA with increased affinity toward alpha2-6-linked SA, the linkage type found prominently on human tracheal epithelial cells. We identified a low pathogenic H7N2 virus isolated from a man in New York in 2003, A/NY/107/03, which replicated efficiently in the upper respiratory tract of ferrets and was capable of transmission in this species by direct contact. These results indicate that H7 influenza viruses from the North American lineage have acquired sialic acid-binding properties that more closely resemble those of human influenza viruses and have the potential to spread to naïve animals.


Subject(s)
Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza in Birds/transmission , Influenza, Human/transmission , Receptors, Virus/physiology , Virus Attachment , Animals , Disease Models, Animal , Ferrets/virology , Hemagglutination Tests , Humans , Influenza A Virus, H7N7 Subtype/chemistry , Influenza A Virus, H7N7 Subtype/physiology , Male , Microarray Analysis , N-Acetylneuraminic Acid/analysis , Polysaccharides/analysis , Poultry/virology , Virus Replication
11.
J Virol ; 83(16): 8131-40, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19493997

ABSTRACT

This study investigated whether transmissible H5 subtype human-avian reassortant viruses could be generated in vivo. To this end, ferrets were coinfected with recent avian H5N1 (A/Thailand/16/04) and human H3N2 (A/Wyoming/3/03) viruses. Genotype analyses of plaque-purified viruses from nasal secretions of coinfected ferrets revealed that approximately 9% of recovered viruses contained genes from both progenitor viruses. H5 and H3 subtype viruses, including reassortants, were found in airways extending toward and in the upper respiratory tract of ferrets. However, only parental H5N1 genotype viruses were found in lung tissue. Approximately 34% of the recovered reassortant viruses possessed the H5 hemagglutinin (HA) gene, with five unique H5 subtypes recovered. These H5 reassortants were selected for further studies to examine their growth and transmissibility characteristics. Five H5 viruses with representative reassortant genotypes showed reduced titers in nasal secretions of infected ferrets compared to the parental H5N1 virus. No transmission by direct contact between infected and naïve ferrets was observed. These studies indicate that reassortment between H5N1 avian influenza and H3N2 human viruses occurred readily in vivo and furthermore that reassortment between these two viral subtypes is likely to occur in ferret upper airways. Given the relatively high incidence of reassortant viruses from tissues of the ferret upper airway, it is reasonable to conclude that continued exposure of humans and animals to H5N1 alongside seasonal influenza viruses increases the risk of generating H5 subtype reassortant viruses that may be shed from upper airway secretions.


Subject(s)
Disease Models, Animal , Ferrets , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/transmission , Reassortant Viruses/physiology , Animals , Cell Line , Dogs , Genotype , Humans , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/physiology , Influenza, Human/virology , Male , Public Health , Reassortant Viruses/genetics , Respiratory System/virology , Virus Replication
12.
J Virol ; 83(7): 2851-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144714

ABSTRACT

Although highly pathogenic avian influenza H5N1 viruses have yet to acquire the ability to transmit efficiently among humans, the increasing genetic diversity among these viruses and continued outbreaks in avian species underscore the need for more effective measures for the control and prevention of human H5N1 virus infection. Additional small animal models with which therapeutic approaches against virulent influenza viruses can be evaluated are needed. In this study, we used the guinea pig model to evaluate the relative virulence of selected avian and human influenza A viruses. We demonstrate that guinea pigs can be infected with avian and human influenza viruses, resulting in high titers of virus shedding in nasal washes for up to 5 days postinoculation (p.i.) and in lung tissue of inoculated animals. However, other physiologic indicators typically associated with virulent influenza virus strains were absent in this species. We evaluated the ability of intranasal treatment with human alpha interferon (alpha-IFN) to reduce lung and nasal wash titers in guinea pigs challenged with the reconstructed 1918 pandemic H1N1 virus or a contemporary H5N1 virus. IFN treatment initiated 1 day prior to challenge significantly reduced or prevented infection of guinea pigs by both viruses, as measured by virus titer determination and seroconversion. The expression of the antiviral Mx protein in lung tissue correlated with the reduction of virus titers. We propose that the guinea pig may serve as a useful small animal model for testing the efficacy of antiviral compounds and that alpha-IFN treatment may be a useful antiviral strategy against highly virulent strains with pandemic potential.


Subject(s)
Immunologic Factors/therapeutic use , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Interferon-alpha/therapeutic use , Orthomyxoviridae Infections/prevention & control , Virus Shedding/immunology , Administration, Intranasal , Animals , Antibodies, Viral/blood , Disease Models, Animal , Guinea Pigs , Immunologic Factors/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Interferon-alpha/administration & dosage , Lung/virology , Nasal Cavity/virology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Virulence
13.
Mol Ther Methods Clin Dev ; 18: 402-414, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32695842

ABSTRACT

Monoclonal antibody (mAb) therapeutics are an effective modality for the treatment of infectious, autoimmune, and cancer-related diseases. However, the discovery, development, and manufacturing processes are complex, resource-consuming activities that preclude the rapid deployment of mAbs in outbreaks of emerging infectious diseases. Given recent advances in nucleic acid delivery technology, it is now possible to deliver exogenous mRNA encoding mAbs for in situ expression following intravenous (i.v.) infusion of lipid nanoparticle-encapsulated mRNA. However, the requirement for i.v. administration limits the application to settings where infusion is an option, increasing the cost of treatment. As an alternative strategy, and to enable intramuscular (IM) administration of mRNA-encoded mAbs, we describe a nanostructured lipid carrier for delivery of an alphavirus replicon encoding a previously described highly neutralizing human mAb, ZIKV-117. Using a lethal Zika virus challenge model in mice, our studies show robust protection following alphavirus-driven expression of ZIKV-117 mRNA when given by IM administration as pre-exposure prophylaxis or post-exposure therapy.

14.
Nat Biomed Eng ; 4(11): 1030-1043, 2020 11.
Article in English | MEDLINE | ID: mdl-32747832

ABSTRACT

The emergence and re-emergence of highly virulent viral pathogens with the potential to cause a pandemic creates an urgent need for the accelerated discovery of antiviral therapeutics. Antiviral human monoclonal antibodies (mAbs) are promising candidates for the prevention and treatment of severe viral diseases, but their long development timeframes limit their rapid deployment and use. Here, we report the development of an integrated sequence of technologies, including single-cell mRNA-sequence analysis, bioinformatics, synthetic biology and high-throughput functional analysis, that enables the rapid discovery of highly potent antiviral human mAbs, the activity of which we validated in vivo. In a 78-d study modelling the deployment of a rapid response to an outbreak, we isolated more than 100 human mAbs that are specific to Zika virus, assessed their function, identified that 29 of these mAbs have broadly neutralizing activity, and verified the therapeutic potency of the lead candidates in mice and non-human primate models of infection through the delivery of an antibody-encoding mRNA formulation and of the respective IgG antibody. The pipeline provides a roadmap for rapid antibody-discovery programmes against viral pathogens of global concern.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antiviral Agents/therapeutic use , Drug Discovery/methods , Zika Virus/immunology , Animals , Cells, Cultured , Computational Biology , Humans , Macaca mulatta , Mice , RNA, Messenger/immunology , Sequence Analysis, RNA
15.
Phytomedicine ; 64: 152927, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31465981

ABSTRACT

BACKGROUND: Next to aluminum salts, squalene nanoemulsions comprise the most widely employed class of adjuvants in approved vaccines. Despite their importance, the mechanisms of action of squalene nanoemulsions are not completely understood, nor are the structure/function requirements of the oil composition. PURPOSE: In this study, we build on previous work that compared the adjuvant properties of nanoemulsions made with different classes of oil structures to squalene nanoemulsion. Here, we introduce nanoemulsions made with polyprenols derived from species of the Pinaceae family as novel vaccine adjuvant compositions. In contrast with long-chain triglycerides that do not efficiently enhance an immune response, both polyprenols and squalene are comprised of multimeric isoprene units, which may represent an important structural property of oils in nanoemulsions with adjuvant properties. STUDY DESIGN: Oils derived from species of the Pinaceae family were formulated in nanoemulsions, with or without a synthetic Toll-like receptor 4 (TLR4) ligand, and characterized regarding physicochemical and biological activity properties in comparison to squalene nanoemulsions. METHODS: Oils were extracted from species of the Pinaceae family and used to prepare oil-in-water nanoemulsions by microfluidization. Emulsion droplet diameter stability was characterized by dynamic light scattering. Nanoemulsions were evaluated for in vitro biological activity using human whole blood, and in vivo biological activity in mouse, pig, and ferret models when combined with pandemic influenza vaccine antigens. RESULTS: Nanoemulsions comprised of Pinaceae-derived polyprenol oils demonstrated long-term physical stability, stimulated cytokine production from human cells in vitro, and promoted antigen-specific immune responses in various animal models, particularly when formulated with the TLR4 ligand glucopyranosyl lipid adjuvant (GLA). CONCLUSION: Pinaceae-derived nanoemulsions are compatible with inclusion of a synthetic TLR4 ligand and promote antigen-specific immune responses to pandemic influenza antigens in mouse, pig, and ferret models.


Subject(s)
Adjuvants, Immunologic/pharmacology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Pinaceae/chemistry , Plant Oils/pharmacology , Polyprenols/pharmacology , Squalene/pharmacology , Adjuvants, Immunologic/chemistry , Animals , Emulsions , Female , Ferrets , Humans , Influenza, Human/virology , Male , Mice , Mice, Inbred C57BL , Plant Oils/chemistry , Polyprenols/chemistry , Specific Pathogen-Free Organisms , Squalene/chemistry , Swine , Toll-Like Receptor 4/immunology
16.
Vaccine ; 37(29): 3902-3910, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31174937

ABSTRACT

The identification of adjuvants that promote lasting antigen-specific immunity and augment vaccine efficacy are integral to the development of new protein-based vaccines. The Ebola virus-like particle (VLP) vaccine expressing Ebola virus glycoprotein (GP) and matrix protein (VP40) was used in this study to evaluate the ability of TLR4 agonist glucopyranosyl lipid adjuvant (GLA) formulated in a stable emulsion (SE) to enhance immunogenicity and promote durable protection against mouse-adapted Ebola virus (ma-EBOV). Antibody responses and Ebola-specific T cell responses were evaluated post vaccination. Survival analysis after lethal ma-EBOV challenge was performed 4 weeks and 22 weeks following final vaccination. GLA-SE enhanced EBOV-specific immunity and resulted in long-term protection against challenge with ma-EBOV infection in a mouse model. Specifically, GLA-SE elicited Th1-skewed antibodies and promoted the generation of EBOV GP-specific polyfunctional T cells. These results provide further support for the utility of TLR4 activating GLA-SE-adjuvanted vaccines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Ebola Vaccines/immunology , Glycosides/immunology , Lipids/immunology , Vaccines, Virus-Like Particle/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Ebola Vaccines/administration & dosage , Ebolavirus , Female , Glycosides/administration & dosage , Glycosides/chemistry , Hemorrhagic Fever, Ebola/prevention & control , Lipids/administration & dosage , Mice , Vaccines, Virus-Like Particle/immunology
17.
Front Immunol ; 9: 2420, 2018.
Article in English | MEDLINE | ID: mdl-30386348

ABSTRACT

The rapid generation of strong T cell responses is highly desirable and viral vectors can have potent CD8+ T cell-inducing activity. Immunity to leishmaniasis requires selective T cell responses, with immunization schemes that raise either CD4 or CD8 T cell responses being protective in small animal models. We have defined the leishmaniasis vaccine candidate recombinant fusion antigens, LEISH-F2 and LEISH-F3+, that when formulated in a stable emulsion with a Toll-like receptor (TLR) 4 agonist, induce protective CD4+ T cell responses in animal models as well as providing therapeutic efficacy in canine leishmaniasis and in clinical trials in leishmaniasis patients. We used the genetic sequences of these validated vaccine antigens to design RNA vaccine constructs. Immunization of mice with the RNA replicons induced potent, local innate responses that were surprisingly independent of TLR7 and activated antigen-presenting cells (APC) to prime for extremely potent antigen-specific T helper 1 type responses upon heterologous boosting with either of the subunit vaccines (recombinant antigen with second generation glucopyranosyl lipid A in stable oil-in-water emulsion; SLA-SE). Inclusion of RNA in the immunization schedule also generated MHCI-restricted T cell responses. Immunization with LEISH-F2-expressing RNA vaccine followed later by subunit vaccine afforded protection against challenge with Leishmania donovani. Together, these data indicate the utility of heterologous prime-boost immunization schemes for the induction of potent antigen-specific CD4 and CD8 T cell responses for protection against intracellular pathogens.


Subject(s)
Immunity, Heterologous , Leishmania donovani/immunology , Leishmaniasis Vaccines/immunology , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , T-Lymphocytes/immunology , Vaccines, Subunit/immunology , Animals , Cell Line , Cytokines/metabolism , Female , Humans , Immunization, Secondary , Leishmania donovani/genetics , Leishmaniasis Vaccines/genetics , Leishmaniasis, Visceral/prevention & control , Lymphocyte Activation/immunology , Mice , NF-kappa B/metabolism , Protein Transport , T-Lymphocytes/metabolism , Toll-Like Receptor 7/metabolism , Vaccines, Subunit/genetics , Vaccines, Synthetic/immunology
18.
Front Immunol ; 9: 295, 2018.
Article in English | MEDLINE | ID: mdl-29515589

ABSTRACT

Elderly people are at high risk for influenza-related morbidity and mortality due to progressive immunosenescence. While toll-like receptor (TLR) agonist containing adjuvants, and other adjuvants, have been shown to enhance influenza vaccine-induced protective responses, the mechanisms underlying how these adjuvanted vaccines could benefit the elderly remain elusive. Here, we show that a split H1N1 influenza vaccine (sH1N1) combined with a TLR4 agonist, glucopyranosyl lipid adjuvant formulated in a stable oil-in-water emulsion (GLA-SE), boosts IgG2c:IgG1 ratios, enhances hemagglutination inhibition (HAI) titers, and increases protection in aged mice. We find that all adjuvanted sH1N1 vaccines tested were able to protect both young and aged mice from lethal A/H1N1/California/4/2009 virus challenge after two immunizations compared to vaccine alone. We show that GLA-SE combined with sH1N1, however, also provides enhanced protection from morbidity in aged mice given one immunization (based on change in weight percentage). While the GLA-SE-adjuvanted sH1N1 vaccine promotes the generation of cytokine-producing T helper 1 cells, germinal center B cells, and long-lived bone marrow plasma cells in young mice, these responses were muted in aged mice. Differential in vitro responses, dependent on age, were also observed from mouse-derived bone marrow-derived dendritic cells and lung homogenates following stimulation with adjuvants, including GLA-SE. Besides enhanced HAI titers, additional protective factors elicited with sH1N1 + GLA-SE in young mice were observed, including (a) rapid reduction of viral titers in the lung, (b) prevention of excessive lung inflammation, and (c) homeostatic maintenance of alveolar macrophages (AMs) following H1N1 infection. Collectively, our results provide insight into mechanisms of adjuvant-mediated immune protection in the young and elderly.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Aged , Animals , Antibodies, Viral/blood , Cells, Cultured , Dendritic Cells/virology , Female , Glucosides/pharmacology , Glucosides/therapeutic use , Humans , Immunity , Immunization , Lipid A/pharmacology , Lipid A/therapeutic use , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists
19.
Sci Adv ; 4(9): eaas9930, 2018 09.
Article in English | MEDLINE | ID: mdl-30221194

ABSTRACT

Adjuvants are key to shaping the immune response to vaccination, but to date, no adjuvant suitable for human use has been developed for intradermal vaccines. These vaccines could be self-administered and sent through the mail as they do not require long needles or technical expertise in immunization. In the event of a pandemic outbreak, this approach could alleviate the congregation of patients in health centers and thus reduce the potential of these centers to enhance the spread of lethal infection. A reliable and potent vaccine system for self-administration would provide an effective countermeasure for delivery through existing product distribution infrastructure. We report results from preclinical and clinical trials that demonstrate the feasibility of an adjuvanted, intradermal vaccine that induced single shot protection in ferrets and seroprotection in humans against one of the more lethal strains of pandemic flu, Indonesia H5N1. In the human trial, the vaccine was safe and clinical responses were above approvable endpoints for a protective flu vaccine. Inclusion of a modern TLR4 (Toll-like receptor 4) agonist-based adjuvant was critical to the development of the response in the intradermal groups. In humans, this is the first report of a safe and effective intradermal adjuvant, GLA-AF (aqueous formulation of glucopyranosyl lipid adjuvant), and provides a future path for developing a vaccine-device combination for distribution by mail and self-administration in case of a pandemic.


Subject(s)
1,2-Dipalmitoylphosphatidylcholine/immunology , Adjuvants, Immunologic/pharmacology , Influenza Vaccines/pharmacology , Lipid A/analogs & derivatives , 1,2-Dipalmitoylphosphatidylcholine/adverse effects , 1,2-Dipalmitoylphosphatidylcholine/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adult , Animals , Drug Combinations , Female , Ferrets , Guinea Pigs , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Injections, Intradermal , Lipid A/adverse effects , Lipid A/immunology , Lipid A/pharmacology , Male , Mice, Inbred C57BL , Toll-Like Receptor 4/agonists
20.
NPJ Vaccines ; 3: 39, 2018.
Article in English | MEDLINE | ID: mdl-30302281

ABSTRACT

Members of the Flaviviridae family are the leading causes of mosquito-borne viral disease worldwide. While dengue virus is the most prevalent, the recent Zika virus outbreak in the Americas triggered a WHO public health emergency, and yellow fever and West Nile viruses (WNV) continue to cause regional epidemics. Given the sporadic nature of flaviviral epidemics both temporally and geographically, there is an urgent need for vaccines that can rapidly provide effective immunity. Protection from flaviviral infection is correlated with antibodies to the viral envelope (E) protein, which encodes receptor binding and fusion functions. TLR agonist adjuvants represent a promising tool to enhance the protective capacity of flavivirus vaccines through dose and dosage reduction and broadening of antiviral antibody responses. This study investigates the ability to improve the immunogenicity and protective capacity of a promising clinical-stage WNV recombinant E-protein vaccine (WN-80E) using a novel combination adjuvant, which contains a potent TLR-4 agonist and the saponin QS21 in a liposomal formulation (SLA-LSQ). Here, we show that, in combination with WN-80E, optimized SLA-LSQ is capable of inducing long-lasting immune responses in preclinical models that provide sterilizing protection from WNV challenge, reducing viral titers following WNV challenge to undetectable levels in Syrian hamsters. We have investigated potential mechanisms of action by examining the antibody repertoire generated post-immunization. SLA-LSQ induced a more diverse antibody response to WNV recombinant E-protein antigen than less protective adjuvants. Collectively, these studies identify an adjuvant formulation that enhances the protective capacity of recombinant flavivirus vaccines.

SELECTION OF CITATIONS
SEARCH DETAIL