Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Mol Pharm ; 18(2): 679-698, 2021 02 01.
Article En | MEDLINE | ID: mdl-32491861

Current influenza virus vaccines are focused on humoral immunity and are limited by the short duration of protection, narrow cross-strain efficacy, and suboptimal immunogenicity. Here, we combined two chemically and biologically distinct adjuvants, an oil-in-water nanoemulsion (NE) and RNA-based agonists of RIG-I, to determine whether the diverse mechanisms of these adjuvants could lead to improved immunogenicity and breadth of protection against the influenza virus. NE activates TLRs, stimulates immunogenic apoptosis, and enhances cellular antigen uptake, leading to a balanced TH1/TH2/TH17 response when administered intranasally. RIG-I agonists included RNAs derived from Sendai and influenza viral defective interfering RNAs (IVT DI, 3php, respectively) and RIG-I/TLR3 agonist, poly(I:C) (pIC), which induce IFN-Is and TH1-polarized responses. NE/RNA combined adjuvants potentially allow for costimulation of multiple innate immune receptor pathways, more closely mimicking patterns of activation occurring during natural viral infection. Mice intranasally immunized with inactivated A/Puerto Rico/8/1934 (H1N1) (PR/8) adjuvanted with NE/IVT DI or NE/3php (but not NE/pIC) showed synergistic enhancement of systemic PR/8-specific IgG with significantly greater avidity and virus neutralization activity than the individual adjuvants. Notably, NE/IVT DI induced protective neutralizing titers after a single immunization. Hemagglutinin stem-specific antibodies were also improved, allowing recognition of heterologous and heterosubtypic hemagglutinins. All NE/RNAs elicited substantial PR/8-specific sIgA. Finally, a unique cellular response with enhanced TH1/TH17 immunity was induced with the NE/RNAs. These results demonstrate that the enhanced immunogenicity of the adjuvant combinations was synergistic and not simply additive, highlighting the potential value of a combined adjuvant approach for improving the efficacy of vaccination against the influenza virus.


DEAD Box Protein 58/metabolism , Drug Carriers/chemistry , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , RNA, Small Interfering/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Dogs , Emulsions , Female , Humans , Immunity, Cellular , Immunity, Humoral , Immunity, Mucosal , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Nanoparticles/chemistry , Poly I-C/administration & dosage , Primary Cell Culture , RNA, Small Interfering/immunology , Vaccination/methods
2.
Lancet Infect Dis ; 20(1): 80-91, 2020 01.
Article En | MEDLINE | ID: mdl-31630990

BACKGROUND: Influenza viruses cause substantial annual morbidity and mortality globally. Current vaccines protect against influenza only when well matched to the circulating strains. However, antigenic drift can cause considerable mismatches between vaccine and circulating strains, substantially reducing vaccine effectiveness. Moreover, current seasonal vaccines are ineffective against pandemic influenza, and production of a vaccine matched to a newly emerging virus strain takes months. Therefore, there is an unmet medical need for a broadly protective influenza virus vaccine. We aimed to test the ability of chimeric H1 haemagglutinin-based universal influenza virus vaccine candidates to induce broadly cross-reactive antibodies targeting the stalk domain of group 1 haemagglutinin-expressing influenza viruses. METHODS: We did a randomised, observer-blinded, phase 1 study in healthy adults in two centres in the USA. Participants were randomly assigned to one of three prime-boost, chimeric haemagglutinin-based vaccine regimens or one of two placebo groups. The vaccine regimens included a chimeric H8/1, intranasal, live-attenuated vaccine on day 1 followed by a non-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine on day 85; the same regimen but with the inactivated vaccine being adjuvanted with AS03; and an AS03-adjuvanted, chimeric H8/1, intramuscular, inactivated vaccine followed by an AS03-adjuvanted, chimeric H5/1, intramuscular, inactivated vaccine. In this planned interim analysis, the primary endpoints of reactogenicity and safety were assessed by blinded study group. We also assessed anti-H1 haemagglutinin stalk, anti-H2, anti-H9, and anti-H18 IgG antibody titres and plasmablast and memory B-cell responses in peripheral blood. This trial is registered with ClinicalTrials.gov, number NCT03300050. FINDINGS: Between Oct 10, 2017, and Nov 27, 2017, 65 participants were enrolled and randomly assigned. The adjuvanted inactivated vaccine, but not the live-attenuated vaccine, induced a substantial serum IgG antibody response after the prime immunisation, with a seven times increase in anti-H1 stalk antibody titres on day 29. After boost immunisation, all vaccine regimens induced detectable anti-H1 stalk antibody (2·2-5·6 times induction over baseline), cross-reactive serum IgG antibody, and peripheral blood plasmablast responses. An unsolicited adverse event was reported for 29 (48%) of 61 participants. Solicited local adverse events were reported in 12 (48%) of 25 participants following prime vaccination with intramuscular study product or placebo, in 12 (33%) of 36 after prime immunisation with intranasal study product or placebo, and in 18 (32%) of 56 following booster doses of study product or placebo. Solicited systemic adverse events were reported in 14 (56%) of 25 after prime immunisation with intramuscular study product or placebo, in 22 (61%) of 36 after immunisation with intranasal study product or placebo, and in 21 (38%) of 56 after booster doses of study product or placebo. Disaggregated safety data were not available at the time of this interim analysis. INTERPRETATION: The tested chimeric haemagglutinin-based, universal influenza virus vaccine regimens elicited cross-reactive serum IgG antibodies that targeted the conserved haemagglutinin stalk domain. This is the first proof-of-principle study to show that high anti-stalk titres can be induced by a rationally designed vaccine in humans and opens up avenues for further development of universal influenza virus vaccines. On the basis of the blinded study group, the vaccine regimens were tolerable and no safety concerns were observed. FUNDING: Bill & Melinda Gates Foundation.


Adjuvants, Immunologic , Hemagglutinins , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Vaccination , Adjuvants, Immunologic/administration & dosage , Adult , Female , Healthy Volunteers , Humans , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Male , Vaccines, Attenuated/immunology , Vaccines, Inactivated/immunology
3.
J Virol ; 91(12)2017 06 15.
Article En | MEDLINE | ID: mdl-28356526

Seasonal influenza virus epidemics represent a significant public health burden. Approximately 25% of all influenza virus infections are caused by type B viruses, and these infections can be severe, especially in children. Current influenza virus vaccines are an effective prophylaxis against infection but are impacted by rapid antigenic drift, which can lead to mismatches between vaccine strains and circulating strains. Here, we describe a broadly protective vaccine candidate based on chimeric hemagglutinins, consisting of globular head domains from exotic influenza A viruses and stalk domains from influenza B viruses. Sequential vaccination with these constructs in mice leads to the induction of broadly reactive antibodies that bind to the conserved stalk domain of influenza B virus hemagglutinin. Vaccinated mice are protected from lethal challenge with diverse influenza B viruses. Results from serum transfer experiments and antibody-dependent cell-mediated cytotoxicity (ADCC) assays indicate that this protection is antibody mediated and based on Fc effector functions. The present data suggest that chimeric hemagglutinin-based vaccination is a viable strategy to broadly protect against influenza B virus infection.IMPORTANCE While current influenza virus vaccines are effective, they are affected by mismatches between vaccine strains and circulating strains. Furthermore, the antiviral drug oseltamivir is less effective for treating influenza B virus infections than for treating influenza A virus infections. A vaccine that induces broad and long-lasting protection against influenza B viruses is therefore urgently needed.


Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibody-Dependent Cell Cytotoxicity , Disease Models, Animal , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A virus/chemistry , Influenza A virus/genetics , Influenza A virus/immunology , Influenza B virus/chemistry , Mice , Orthomyxoviridae Infections/virology , Receptors, Fc/immunology , Vaccination
4.
mBio ; 8(2)2017 03 28.
Article En | MEDLINE | ID: mdl-28351919

An exacerbated immune response is one of the main causes of influenza-induced lung damage during infection. The molecular mechanisms regulating the fate of the initial immune response to infection, either as a protective response or as detrimental immunopathology, are not well understood. The purinergic receptor P2X7 is an ionotropic nucleotide-gated ion channel receptor expressed on immune cells that has been implicated in induction and maintenance of excessive inflammation. Here, we analyze the role of this receptor in a mouse model of influenza virus infection using a receptor knockout (KO) mouse strain. Our results demonstrate that the absence of the P2X7 receptor results in a better outcome to influenza virus infection characterized by reduced weight loss and increased survival upon experimental influenza challenge compared to wild-type mice. This effect was not virus strain specific. Overall lung pathology and apoptosis were reduced in virus-infected KO mice. Production of proinflammatory cytokines and chemokines such as interleukin-10 (IL-10), gamma interferon (IFN-γ), and CC chemokine ligand 2 (CCL2) was also reduced in the lungs of the infected KO mice. Infiltration of neutrophils and depletion of CD11b+ macrophages, characteristic of severe influenza virus infection in mice, were lower in the KO animals. Together, these results demonstrate that activation of the P2X7 receptor is involved in the exacerbated immune response observed during influenza virus infection.IMPORTANCE A hallmark of influenza virus infection is the development of lung pathology induced by an exacerbated immune response. The mechanisms shared by the antiviral host defense required for viral clearance and those required for development of immunopathology are not clearly understood. Purinergic receptors, and in particular the purinergic receptor P2X7 (P2X7r), are involved in activation of the immune response. We used mice lacking the P2X7r (P2X7r KO mice) to better understand the mechanisms that lead to development of lung pathology during influenza virus infection. In our studies, we observed that P2X7r KO mice developed less lung immunopathology and had better survival than the wild-type mice. These results implicate P2X7r in the induction of an exacerbated local immune response to influenza virus and help us to better understand the mechanisms leading to the lung immunopathology observed during severe viral infections.


Host-Pathogen Interactions , Lung/pathology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae/pathogenicity , Receptors, Purinergic P2X7/metabolism , Animals , Body Weight , Cytokines/analysis , Disease Models, Animal , Macrophages/immunology , Mice , Mice, Knockout , Neutrophils/immunology , Orthomyxoviridae Infections/virology , Survival Analysis
5.
mBio ; 8(2)2017 03 21.
Article En | MEDLINE | ID: mdl-28325769

Antibody responses to influenza virus hemagglutinin provide protection against infection and are well studied. Less is known about the human antibody responses to the second surface glycoprotein, neuraminidase. Here, we assessed human antibody reactivity to a panel of N1, N2, and influenza B virus neuraminidases in different age groups, including children, adults, and the elderly. Using enzyme-linked immunosorbent assays (ELISA), we determined the breadth, magnitude, and isotype distribution of neuraminidase antibody responses to historic, current, and avian strains, as well as to recent isolates to which these individuals have not been exposed. It appears that antibody levels against N1 neuraminidases were lower than those against N2 or B neuraminidases. The anti-neuraminidase antibody levels increased with age and were, in general, highest against strains that circulated during the childhood of the tested individuals, providing evidence for "original antigenic sin." Titers measured by ELISA correlated well with titers measured by the neuraminidase inhibition assays. However, in the case of the 2009 pandemic H1N1 virus, we found evidence of interference from antibodies binding to the conserved stalk domain of the hemagglutinin. In conclusion, we found that antibodies against the neuraminidase differ in magnitude and breadth between subtypes and age groups in the human population. (This study has been registered at ClinicalTrials.gov under registration no. NCT00336453, NCT00539981, and NCT00395174.)IMPORTANCE Anti-neuraminidase antibodies can afford broad protection from influenza virus infection in animal models and humans. However, little is known about the breadth and magnitude of the anti-neuraminidase response in the human population. Here we assessed antibody levels of children, adults, and the elderly against a panel of N1, N2, and type B influenza virus neuraminidases. We demonstrated that antibody levels measured by ELISA correlate well with functional neuraminidase inhibition titers. This is an important finding since ELISA is a simpler method than functional assays and can be implemented in high-throughput settings to analyze large numbers of samples. Furthermore, we showed that low titers of broadly cross-reactive antibodies against neuraminidase are prevalent in humans. By the use of an appropriate vaccination strategy, these titers could potentially be boosted to levels that might provide broad protection from influenza virus infection.


Antibodies, Viral/blood , Neuraminidase/immunology , Viral Proteins/immunology , Adolescent , Adult , Adult Children , Age Factors , Aged , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Humans , Infant , Middle Aged , Neuraminidase/analysis , Randomized Controlled Trials as Topic , Young Adult
6.
J Virol ; 90(7): 3789-93, 2016 Jan 13.
Article En | MEDLINE | ID: mdl-26764009

A panel of influenza A viruses expressing chimeric hemagglutinins (cHA) with intragroup or intergroup head/stalk combinations was generated. Viruses were characterized for growth kinetics and preservation of stalk epitopes. With a few notable exceptions, cHA viruses behaved similarly to wild-type viruses and maintained stalk epitopes, which indicated their potential as vaccine candidates to induce stalk-specific antibodies.


Hemagglutinin Glycoproteins, Influenza Virus/biosynthesis , Influenza A virus/genetics , Recombinant Proteins/biosynthesis , Cell Line , Epitopes/genetics , Gene Expression , Genomic Instability , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A virus/growth & development , Influenza A virus/physiology , Influenza Vaccines/genetics , Neutralization Tests , Recombinant Proteins/genetics , Viral Plaque Assay , Virus Replication
7.
Emerg Infect Dis ; 21(7): 1197-201, 2015 Jul.
Article En | MEDLINE | ID: mdl-26079843

Three cases of influenza A(H10N8) virus infection in humans have been reported; 2 of these infected persons died. Characterization of the receptor binding pattern of H10 hemagglutinin from avian and human isolates showed that both interact weakly with human-like receptors and maintain strong affinity for avian-like receptors.


Hemagglutinins/physiology , Influenza A Virus, H10N8 Subtype/physiology , Receptors, Virus/physiology , Animals , Cell Line, Tumor , Dogs , Hemagglutinins/chemistry , Humans , Madin Darby Canine Kidney Cells , Protein Binding , Receptors, Virus/chemistry , Virus Attachment , Virus Replication
8.
PLoS Negl Trop Dis ; 9(3): e0003584, 2015 Mar.
Article En | MEDLINE | ID: mdl-25756647

BACKGROUND: Multiple recent outbreaks of Rift Valley Fever (RVF) in Africa, Madagascar, and the Arabian Peninsula have resulted in significant morbidity, mortality, and financial loss due to related livestock epizootics. Presentation of human RVF varies from mild febrile illness to meningoencephalitis, hemorrhagic diathesis, and/or ophthalmitis with residual retinal scarring, but the determinants for severe disease are not understood. The aim of the present study was to identify human genes associated with RVF clinical disease in a high-risk population in Northeastern Province, Kenya. METHODOLOGY/PRINCIPAL FINDINGS: We conducted a cross-sectional survey among residents (N = 1,080; 1-85 yrs) in 6 villages in the Sangailu Division of Ijara District. Participants completed questionnaires on past symptoms and exposures, physical exam, vision testing, and blood collection. Single nucleotide polymorphism (SNP) genotyping was performed on a subset of individuals who reported past clinical symptoms consistent with RVF and unrelated subjects. Four symptom clusters were defined: meningoencephalitis, hemorrhagic fever, eye disease, and RVF-not otherwise specified. SNPs in 46 viral sensing and response genes were investigated. Association was analyzed between SNP genotype, serology and RVF symptom clusters. The meningoencephalitis symptom phenotype cluster among seropositive patients was associated with polymorphisms in DDX58/RIG-I and TLR8. Having three or more RVF-related symptoms was significantly associated with polymorphisms in TICAM1/TRIF, MAVS, IFNAR1 and DDX58/RIG-I. SNPs significantly associated with eye disease included three different polymorphisms TLR8 and hemorrhagic fever symptoms associated with TLR3, TLR7, TLR8 and MyD88. CONCLUSIONS/SIGNIFICANCE: Of the 46 SNPs tested, TLR3, TLR7, TLR8, MyD88, TRIF, MAVS, and RIG-I were repeatedly associated with severe symptomatology, suggesting that these genes may have a robust association with RVFV-associated clinical outcomes. Studies of these and related genetic polymorphisms are warranted to advance understanding of RVF pathogenesis.


Immunity, Innate , Polymorphism, Single Nucleotide , Rift Valley Fever/genetics , Rift Valley Fever/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child , Child, Preschool , Cross-Sectional Studies , Female , Genotype , Haplotypes , Humans , Infant , Male , Middle Aged , Rift Valley fever virus/genetics , Toll-Like Receptor 3/genetics , Young Adult
9.
Virology ; 449: 174-80, 2014 Jan 20.
Article En | MEDLINE | ID: mdl-24418550

Inflammasome activation is gaining recognition as an important mechanism for protection during viral infection. Here, we investigate whether Rift Valley fever virus, a negative-strand RNA virus, can induce inflammasome responses and IL-1ß processing in immune cells. We have determined that RVFV induces NLRP3 inflammasome activation in murine dendritic cells, and that this process is dependent upon ASC and caspase-1. Furthermore, absence of the cellular RNA helicase adaptor protein MAVS/IPS-1 significantly reduces extracellular IL-1ß during infection. Finally, direct imaging using confocal microscopy shows that the MAVS protein co-localizes with NLRP3 in the cytoplasm of RVFV infected cells.


Carrier Proteins/immunology , Inflammasomes/immunology , Rift Valley Fever/immunology , Rift Valley fever virus/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Humans , Inflammasomes/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Rift Valley Fever/genetics , Rift Valley Fever/virology , Rift Valley fever virus/genetics
10.
J Virol ; 87(9): 4846-60, 2013 May.
Article En | MEDLINE | ID: mdl-23408632

Rift Valley fever virus (RVFV) is an emerging RNA virus with devastating economic and social consequences. Clinically, RVFV induces a gamut of symptoms ranging from febrile illness to retinitis, hepatic necrosis, hemorrhagic fever, and death. It is known that type I interferon (IFN) responses can be protective against severe pathology; however, it is unknown which innate immune receptor pathways are crucial for mounting this response. Using both in vitro assays and in vivo mucosal mouse challenge, we demonstrate here that RNA helicases are critical for IFN production by immune cells and that signaling through the helicase adaptor molecule MAVS (mitochondrial antiviral signaling) is protective against mortality and more subtle pathology during RVFV infection. In addition, we demonstrate that Toll-like-receptor-mediated signaling is not involved in IFN production, further emphasizing the importance of the RNA cellular helicases in type I IFN responses to RVFV.


DEAD-box RNA Helicases/immunology , Interferon-beta/immunology , Mucous Membrane/virology , Rift Valley Fever/enzymology , Rift Valley Fever/immunology , Rift Valley fever virus/physiology , Animals , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Humans , Interferon-beta/genetics , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Mucous Membrane/immunology , Rift Valley Fever/prevention & control , Rift Valley Fever/virology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
...