Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
mSphere ; 6(2)2021 03 10.
Article in English | MEDLINE | ID: mdl-33692193

ABSTRACT

Influenza vaccines have traditionally been tested in naive mice and ferrets. However, humans are first exposed to influenza viruses within the first few years of their lives. Therefore, there is a pressing need to test influenza virus vaccines in animal models that have been previously exposed to influenza viruses before being vaccinated. In this study, previously described H2 computationally optimized broadly reactive antigen (COBRA) hemagglutinin (HA) vaccines (Z1 and Z5) were tested in influenza virus "preimmune" ferret models. Ferrets were infected with historical, seasonal influenza viruses to establish preimmunity. These preimmune ferrets were then vaccinated with either COBRA H2 HA recombinant proteins or wild-type H2 HA recombinant proteins in a prime-boost regimen. A set of naive preimmune or nonpreimmune ferrets were also vaccinated to control for the effects of the multiple different preimmunities. All of the ferrets were then challenged with a swine H2N3 influenza virus. Ferrets with preexisting immune responses influenced recombinant H2 HA-elicited antibodies following vaccination, as measured by hemagglutination inhibition (HAI) and classical neutralization assays. Having both H3N2 and H1N1 immunological memory regardless of the order of exposure significantly decreased viral nasal wash titers and completely protected all ferrets from both morbidity and mortality, including the mock-vaccinated ferrets in the group. While the vast majority of the preimmune ferrets were protected from both morbidity and mortality across all of the different preimmunities, the Z1 COBRA HA-vaccinated ferrets had significantly higher antibody titers and recognized the highest number of H2 influenza viruses in a classical neutralization assay compared to the other H2 HA vaccines.IMPORTANCE H1N1 and H3N2 influenza viruses have cocirculated in the human population since 1977. Nearly every human alive today has antibodies and memory B and T cells against these two subtypes of influenza viruses. H2N2 influenza viruses caused the 1957 global pandemic and people born after 1968 have never been exposed to H2 influenza viruses. It is quite likely that a future H2 influenza virus could transmit within the human population and start a new global pandemic, since the majority of people alive today are immunologically naive to viruses of this subtype. Therefore, an effective vaccine for H2 influenza viruses should be tested in an animal model with previous exposure to influenza viruses that have circulated in humans. Ferrets were infected with historical influenza A viruses to more accurately mimic the immune responses in people who have preexisting immune responses to seasonal influenza viruses. In this study, preimmune ferrets were vaccinated with wild-type (WT) and COBRA H2 recombinant HA proteins in order to examine the effects that preexisting immunity to seasonal human influenza viruses have on the elicitation of broadly cross-reactive antibodies from heterologous vaccination.


Subject(s)
Antibodies, Viral/blood , Cross Reactions/immunology , Hemagglutinins, Viral/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Female , Ferrets/immunology , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/chemistry , Influenza A virus/classification , Influenza A virus/genetics , Orthomyxoviridae Infections/immunology , Vaccination
2.
Virulence ; 12(1): 12-19, 2021 12.
Article in English | MEDLINE | ID: mdl-33372841

ABSTRACT

Most of the current approaches to influenza vaccine design focus on antibodies against influenza (HA). However, these influenza vaccines typically provide strain-specific protection against mostly homologous subtypes. There is an urgent need to develop a universal vaccine that confers cross-protection against influenza viruses. Of note, the HA stalk domain (HAsd) is a promising target for such an influenza vaccine. In this study, we generated recombinant Lactococcus lactis (L. lactis)/pNZ8150-phosphatidylglycerophosphate synthetase A (pgsA)-HAsd, in which pgsA was used as an anchor protein, and investigated the immunogenicity of HAsd in a mouse model by oral administration without the use of a mucosal adjuvant. Compared with L. lactis/pNZ8150-pgsA, mice were orally vaccinated with L. lactis/pNZ8150-pgsA-HAsd and then produced strong humoral and mucosal immune responses. Importantly, L. lactis/pNZ8150-pgsA-HAsd provided cross-protection against H5N1, H3N2 and H1N1 virus infections. Our data support the hypothesis that HAsd presented on the surface of L. lactis can provide cross-protective immunity against divergent influenza A viruses. Taken together, these findings suggest that L. lactis/pNZ8150-pgsA-HAsd can be considered an alternative approach to developing a novel universal vaccine during an influenza A pandemic. Abbreviations: HA, HAsd, HA stalk domain; L. lactis, Lactococcus lactis; SDS-PAGE, sodium dodecyl sulfate-polyacrylamide gel electrophoresis; IFA, immunofluorescence assay; PBS, phosphate-buffered saline; pgsA, phosphatidylglycerophosphate synthetase A; SPF, specific pathogen-free; CFU, colony-forming unit; BSL-3, biosafety level-3 laboratory; TCID50, 50% tissue culture infective dose; ELISA, enzyme-linked immunosorbent assay; OD, optical density; LTB, liable enterotoxin B subunit; CTB, cholera toxin B subunit.


Subject(s)
Cross Protection/immunology , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Influenza A virus/immunology , Lactococcus lactis/genetics , Orthomyxoviridae Infections/immunology , Administration, Oral , Animals , Antibodies, Viral/blood , Female , Hemagglutinins, Viral/administration & dosage , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Influenza A virus/classification , Influenza A virus/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Lactococcus lactis/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
3.
Int J Pharm ; 532(1): 374-383, 2017 Oct 30.
Article in English | MEDLINE | ID: mdl-28855138

ABSTRACT

Microneedle (MN) patches have great potential as transcutaneous vaccine delivery devices because MNs can effectively deliver vaccine antigen into the skin through the micropores formed in the stratum corneum by low-invasive and painless skin puncturing. This study aims to develop novel double-decker MN patches which have not only high safety and efficacy but also broad applicability to various vaccine antigens. We developed two types of MN patches (PGA-MN and Nylon-MN) that are made from polyglycolic acid and Nylon-6. In pre-clinical studies, both MN patches could demonstrably deliver antigens into resected human dermal tissue, prolong antigen deposition and increase antigen-specific IgG levels after vaccination compared with conventional injections. We demonstrated both MN patches could be safely applied to human skin because no broken MNs or significant skin irritation were observed after applications in the clinical research. PGA-MN was suggested to be superior to Nylon-MN regarding human skin puncturability based on measurements of transepidermal water loss and needle failure force. A high content of tetravalent influenza hemagglutinin antigens loaded on PGA-MN could stably maintain HA titers at 35°C for 1year. Overall, double-decker MN patches can reliably and safely puncture human skin and are promising as effective transcutaneous vaccine delivery devices.


Subject(s)
Drug Delivery Systems , Microinjections , Needles , Transdermal Patch , Vaccination/instrumentation , Administration, Cutaneous , Adult , Animals , Antigens/administration & dosage , Antigens/immunology , Drug Delivery Systems/adverse effects , Female , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/immunology , Humans , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Microinjections/adverse effects , Middle Aged , Needles/adverse effects , Rats, Wistar , Skin/metabolism , Skin Irritancy Tests , Transdermal Patch/adverse effects , Young Adult
4.
J Immunol Methods ; 448: 112-115, 2017 09.
Article in English | MEDLINE | ID: mdl-28576653

ABSTRACT

Capripoxviruses (CaPVs) have been shown to be ideal viral vectors for the development of recombinant multivalent vaccines to enable delivery of immunogenic genes from ruminant pathogens. So far, the viral thymidine kinase (TK) gene is the only gene used to generate recombinants. A putative non-essential gene encoding a G-protein-coupled chemokine receptor subfamily homologue (GPCR) was targeted as an additional insertion site. Peste des petits ruminants (PPR) was chosen as a disease model. A new recombinant CaPV expressing the viral attachment hemagglutinin (H) of the PPR virus (PPRV) in the GPCR insertion site (rKS1-HPPR-GPCR) was generated in the backbone North African isolate KS1 strain of lumpy skin disease virus (LSDV). Comparison with the recombinant CaPV expressing the H of PPRV in the TK gene (rKS1-HPPR-TK) shown to induce protection against both PPR and LSD in both sheep and goats was assessed. The suitability of the GPCR gene to be a putative additional insertion site in the CaPV genome is evaluated and discussed.


Subject(s)
Capripoxvirus/genetics , Genetic Vectors , Lumpy Skin Disease/prevention & control , Lumpy skin disease virus/genetics , Mutagenesis, Insertional , Peste-des-Petits-Ruminants/prevention & control , Peste-des-petits-ruminants virus/genetics , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/genetics , Viral Vaccines/genetics , Animals , Antibodies, Viral/blood , Cattle , Chlorocebus aethiops , Goats , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Injections, Subcutaneous , Lumpy Skin Disease/genetics , Lumpy Skin Disease/immunology , Lumpy Skin Disease/virology , Lumpy skin disease virus/immunology , Peste-des-Petits-Ruminants/genetics , Peste-des-Petits-Ruminants/immunology , Peste-des-Petits-Ruminants/virology , Peste-des-petits-ruminants virus/immunology , Receptors, Chemokine/administration & dosage , Receptors, Chemokine/immunology , Receptors, G-Protein-Coupled/administration & dosage , Receptors, G-Protein-Coupled/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Virus Cultivation
5.
Virus Res ; 236: 30-43, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28465158

ABSTRACT

Measles virus (MV) remains a leading cause of vaccine-preventable deaths in children. Protection against MV is associated with neutralizing antibodies that preferentially recognize the viral hemagglutinin (MV-H), and to a lesser extent, the fusion protein (MV-F). Although MV is serologically monotypic, 24 genotypes have been identified. Here we report three neutralization epitopes conserved in the more prevalent circulating MV genotypes, two located in the MV-H receptor binding site (RBS) (antigenic site III) and a third in MV-H/MV-F interphase (antigenic site Ia) which are essential for MV multiplication. In contrast, two MV-H neutralization epitopes, showed a genotype-specific neutralization escape due to a single amino acid change, that we mapped in the "noose" antigenic site, or an enhanced neutralization epitope (antigenic site IIa). The monoclonal antibody (mAb) neutralization potency correlated with its binding affinity and was mainly driven by kinetic dissociation rate (koff). We developed an immunoassay for mAb binding to MV-H in its native hetero-oligomeric structure with MV-F on the surface of a MV productive steady-state persistently infected (p.i.) human cell lines, and a competitive-binding assay with serum from individuals with past infection by different MV genotypes. Binding assays revealed that a broad neutralization epitope, in RBS antigenic site, a genotype specific neutralization epitopes, in noose and IIa sites, were immunogenic in natural infection and vaccination and may elicit long-lasting humoral immunity that might contribute to explain MV immunogenic stability. These results support the design of improved measles vaccines, broad-spectrum prophylactic or therapeutic antibodies and MV-used in oncolytic therapies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Hemagglutinins, Viral/immunology , Measles virus/immunology , Measles/immunology , Antibodies, Neutralizing/immunology , Epitopes/administration & dosage , Epitopes/immunology , Genotype , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Humans , Measles/prevention & control , Measles/virology , Measles Vaccine/administration & dosage , Measles Vaccine/immunology , Measles virus/classification , Measles virus/genetics , Measles virus/isolation & purification , Neutralization Tests , Vaccination
6.
J Control Release ; 233: 208-19, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27178810

ABSTRACT

Influenza infection typically initiates at respiratory mucosal surfaces. Induction of immune responses at the sites where pathogens initiate replication is crucial for the prevention of infection. We studied the adjuvanticity of GPI-anchored CCL28 co-incorporated with influenza HA-antigens in chimeric virus-like particles (cVLPs), in boosting strong protective immune responses through an intranasal (i.n.) route in mice. We compared the immune responses to that from influenza VLPs without CCL28, or physically mixed with soluble CCL28 at systemic and various mucosal compartments. The cVLPs containing GPI-CCL28 showed in-vitro chemotactic activity towards spleen and lung cells expressing CCR3/CCR10 chemokine receptors. The cVLPs induced antigen specific endpoint titers and avidity indices of IgG in sera and IgA in tracheal, lung, and intestinal secretions, significantly higher (4-6 fold) than other formulations. Significantly higher (3-5 fold) hemagglutination inhibition titers and high serum neutralization against H3N2 viruses were also detected with CCL28-containing VLPs compared to other groups. The CCL28-containing VLPs showed complete and 80% protection, when vaccinated animals were challenged with A/Aichi/2/1968/H3N2 (homologous) and A/Philippines/2/1982/H3N2 (heterologous) viruses, respectively. Thus, GPI-anchored CCL28 in influenza VLPs act as a strong immunostimulator at both systemic and mucosal sites, boosting significant cross-protection in animals against heterologous viruses across a large distance.


Subject(s)
Antigens, Viral/administration & dosage , Chemokines, CC/administration & dosage , GPI-Linked Proteins/administration & dosage , Hemagglutinins, Viral/administration & dosage , Influenza A Virus, H3N2 Subtype/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Matrix Proteins/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cell Line , Dogs , Female , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Madin Darby Canine Kidney Cells , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Spodoptera
7.
J Control Release ; 232: 75-82, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27094605

ABSTRACT

The delivery of vaccines to the sublingual mucosa is an attractive prospect due to the ease and acceptability of such an approach. However, novel adjuvant and delivery approaches are required to optimally vaccinate at this site. We have previously shown that conjugation of protein antigen to the iron transport molecule, transferrin, can significantly enhance mucosal immune responses. We tested whether conjugating influenza haemagglutinin to transferrin could improve the immune response to sublingually delivered antigen. Transferrin conjugated haemagglutinin induced a significant antibody and T cell response in both naïve animals and previously immunized animals. The immune response generated was able to protect mice against influenza virus challenge. Sublingually administered antigen dispersed more widely through the gastro-intestinal tract than intranasally delivered antigen and transferrin conjugation had a more marked effect on sublingually delivered antigen than intranasal immunisation. From these studies we conclude that transferrin conjugation of antigen is effective at boosting immune responses to sublingually delivered antigen and may be an attractive approach for influenza vaccines, particularly when mass campaigns are required.


Subject(s)
Antigens, CD/administration & dosage , Hemagglutinins, Viral/administration & dosage , Influenza A Virus, H3N2 Subtype , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Receptors, Transferrin/administration & dosage , Administration, Intranasal , Administration, Sublingual , Animals , Antibodies, Viral/immunology , Antigens, CD/chemistry , Female , Hemagglutinins, Viral/chemistry , Immunoglobulin G/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/chemistry , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Receptors, Transferrin/chemistry , T-Lymphocytes/immunology
8.
Arch Virol ; 159(11): 3043-51, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25064513

ABSTRACT

Both porcine reproductive and respiratory syndrome and swine influenza are acute, highly contagious swine diseases. These diseases pose severe threats for the swine industry and cause heavy economic losses worldwide. In this study, we have developed a chimeric virus-like particle (VLP) vaccine candidate for porcine reproductive and respiratory syndrome virus (PRRSV) and H3N2 influenza virus and investigated its immunogenicity in mice. The HA and M1 proteins from the H3N2 influenza virus and the PRRSV GP5 protein fused to the cytoplasmic and transmembrane domains of the NA protein were both incorporated into the chimeric VLPs. Analysis of the immune responses showed that the chimeric VLPs elicited serum antibodies specific for both PRRSV GP5 and the H3N2 HA protein, and they stimulated cellular immune responses compared to the responses to equivalent amounts of inactivated viruses. Taken together, the results suggested that the chimeric VLP vaccine represents a potential strategy for the development of a safe and effective vaccine to control PRRSV and H3N2 influenza virus.


Subject(s)
Hemagglutinins, Viral/immunology , Influenza A Virus, H3N2 Subtype/immunology , Orthomyxoviridae Infections/veterinary , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Swine Diseases/prevention & control , Viral Envelope Proteins/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/immunology , Female , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Influenza A Virus, H3N2 Subtype/genetics , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Swine , Swine Diseases/immunology , Swine Diseases/virology , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/immunology
9.
Appl Biochem Biotechnol ; 172(7): 3635-45, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24562978

ABSTRACT

The conventional avian influenza vaccines rely on development of neutralizing antibodies against the HA and NA antigens. However, these antigens are highly variable, and hence there is a need for better vaccine candidates which would offer broader protection in animals. The M1 of avian influenza is another major structural protein that has conserved epitopes that are reported to induce CD8+ T cells and can contribute to protection against morbidity and mortality from influenza. Hence in an effort to study the immune response of rM1 either alone or in combination with rHA, the hemagglutinin (HA) and matrix protein (M1) of A/Hatay/2004/H5N1 strain of avian influenza were expressed in Pichia pastoris as his-tagged proteins and purified through Ni-NTA chromatography. The His-tag was removed using TEV protease cleavage site and the immunogenicity of purified rHA and rM1 either alone or in combination was determined in mice. One group of mice was immunized with 5 µg of purified rHA, the other group was immunized with rM1, and a third group of mice were immunized with 5 µg of rHA and rM1. All the animals were boosted twice, once on 28 days postimmunization (dpi) and the second on 42 dpi. The immune response was evaluated by enzyme-linked immunosorbent assay (ELISA) and hemagglutination inhibition (HI) assay. The group of mice immunized with rHA and rM1 together showed significantly higher immune response against rHA and rM1 than mice immunized with either HA or M1 antigens. The addition of rM1 with rHA resulted in increased HI titer in animals immunized with both the antigens. These results suggest that the HA and M1 expressed in P. pastoris can be utilized in combination for the development of faster and cost-effective vaccines for circulating and newer strains of avian influenza and would aid in combating the disease in a pandemic situation, in which production time matters greatly.


Subject(s)
Hemagglutinins, Viral/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/immunology , Female , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Humans , Influenza A Virus, H5N1 Subtype/genetics , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Pichia/genetics , Pichia/metabolism , Viral Matrix Proteins/administration & dosage , Viral Matrix Proteins/genetics
10.
Curr Comput Aided Drug Des ; 7(4): 255-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22050681

ABSTRACT

Recently WHO and NREVSS collaborating laboratories located in all 50 states, and Washington D.C reported that out of 3,588 specimens,164 were found positive for influenza type (i.e. 4.6%) and from these 164 specimens 162 (i.e. 98.8 %) were of influenza A H1N1 subtype. Comparative study of the past and current reports gives a general idea that the influenza activity deserves high attention from public health authorities in the U.S. In this connection, presently some groups are developing intensive computer-aided research in QSAR, Docking, Molecular Modeling and Drug Design, Sequence Analysis and Phylogenetic analysis of candidate compounds and/or targets; in order to advance in the treatment and/or prevention of this pandemic Flu. In this work, primarily we carry out a mini-review of the more important theoretical studies reported until now within this area, followed by the study of a specific type of target. Keeping in view the nature of this virus, we can conclude that there is always a need to find other target protein as inhibitor other than the existing one. So that this lethal pandemic flu can be treated and prevented further. Therefore, after Neuraminidase and M2 ion channels the surface protein that we can target in H1N1 strain is Hemagglutinins (HA). We use comparative modeling; which is one of the methods that can reliably generate a 3D model for HA protein. Multiple structures of this subtype of Influenza Virus are available at PDB, but we are focused on Influenza A (H1N1). Therefore, methodology of analysis mainly focuses on modeling the structure of this protein and, if possible, finding a probable active sites and inhibitors to it.


Subject(s)
Antiviral Agents/chemistry , Drug Delivery Systems/methods , Hemagglutinins, Viral/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Models, Molecular , Quantitative Structure-Activity Relationship , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/metabolism , Binding Sites/physiology , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/metabolism , Humans , Influenza A Virus, H1N1 Subtype/metabolism
11.
Article in Chinese | MEDLINE | ID: mdl-21863626

ABSTRACT

OBJECTIVE: To evaluate the seasonal influenza spilt vaccine's immunogenicity and the 50% effective dose (ED50a) of hemagglutin (HA) that can make 50% of the mice hemagglutination inhibition antibody (HI) titers to 40. METHODS: The 2008-2009 seasonal influenza spilt vaccine's two components, with HA from H1N1 and H3N2 influenza virus respectively, were used as a model. Mice were immunized once or twice with different doses, and the HI antibody titers were tested to determine the immunization procedure and to evaluate the immugenicity of seasonal influenza spilt vaccine in mice; Consequently, HI antibody response kinetics of the two components were observed to determine the time point when the HI antibody titer reached the peak point; Finally, mice were immunized with different doses of HA to evaluate the ED50a that can make 50% of mice HI titers reach 40. RESULTS: Immunization procedures study showed that one-dose of seasonal influenza vaccine induced the HI antibody titers ranged from 10 to 120, while two-dose of influenza vaccine improved the HI antibody titer 10-100 times as compared with one dose; antibody kinetics study suggested that the time point of HI antibody produced to peak is 28-35 days post one dose immunization; and the ED50a detection results indicated that one dose of 1.5 microg HA could make 50% of the mice HI antibody titer reach 40. CONCLUSION: Seasonal influenza spilt vaccine is very immunogenic in mouse; the time point of HI antibody produced to peak is 28-35 days post one dose immunization; and the ED50a of HA is 1.5 microg, which can make 50% of the mice HI titer reach 40. The experimental results provided foundation for the establishment of influenza vaccine evaluation system based on seasonal influenza vaccine.


Subject(s)
Hemagglutinins, Viral/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hemagglutination Inhibition Tests , Hemagglutinins, Viral/immunology , Humans , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/virology , Male , Mice , Mice, Inbred BALB C , Seasons
13.
Clin Vaccine Immunol ; 17(9): 1381-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20631330

ABSTRACT

A simple method suitable for self-administration of vaccine would improve mass immunization, particularly during a pandemic outbreak. Influenza virus-like particles (VLPs) have been suggested as promising vaccine candidates against potentially pandemic influenza viruses, as they confer long-lasting immunity but are not infectious. We investigated the immunogenicity and protective efficacy of influenza H5 VLPs containing the hemagglutinin (HA) of A/Vietnam/1203/04 (H5N1) virus delivered into the skin of mice using metal microneedle patches and also studied the response of Langerhans cells in a human skin model. Prime-boost microneedle vaccinations with H5 VLPs elicited higher levels of virus-specific IgG1 and IgG2a antibodies, virus-specific antibody-secreting cells, and cytokine-producing cells up to 8 months after vaccination compared to the same antigen delivered intramuscularly. Both prime-boost microneedle and intramuscular vaccinations with H5 VLPs induced similar hemagglutination inhibition titers and conferred 100% protection against lethal challenge with the wild-type A/Vietnam/1203/04 virus 16 weeks after vaccination. Microneedle delivery of influenza VLPs to viable human skin using microneedles induced the movement of CD207(+) Langerhans cells toward the basement membrane. Microneedle vaccination in the skin with H5 VLPs represents a promising approach for a self-administered vaccine against viruses with pandemic potential.


Subject(s)
B-Lymphocytes/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Viral/blood , Cytokines/metabolism , Female , Hemagglutination Inhibition Tests , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/immunology , Humans , Immunization, Secondary/methods , Immunoglobulin G/blood , In Vitro Techniques , Injections, Intradermal , Injections, Intramuscular , Langerhans Cells/immunology , Mice , Mice, Inbred BALB C , Needles , Survival Analysis , Time Factors , Vaccination/methods , Vaccines, Virosome/administration & dosage , Vaccines, Virosome/immunology
14.
MMWR Morb Mortal Wkly Rep ; 59(16): 485-6, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20431524

ABSTRACT

Persons aged >or=65 years are at greater risk for hospitalization and death from seasonal influenza compared with other age groups, and they respond to vaccination with lower antibody titers to influenza hemagglutinin (an established correlate of protection against influenza) compared with younger adults. On December 23, 2009, the Food and Drug Administration (FDA) licensed an injectable inactivated trivalent influenza vaccine (Fluzone High-Dose, Sanofi-Pasteur) that contains an increased amount of influenza virus hemagglutinin antigen compared with other inactivated influenza vaccines such as Fluzone. Fluzone High-Dose is licensed as a single dose for use among persons aged >or=65 years and will be available beginning with the 2010-11 influenza season. The Advisory Committee on Immunization Practices (ACIP) reviewed data from prelicensure clinical trials on the safety and immunogenicity of Fluzone High-Dose and expressed no preference for the new vaccine over other inactivated trivalent influenza vaccines. This report summarizes the FDA-approved indications for Fluzone High-Dose and provides guidance from ACIP for its use.


Subject(s)
Hemagglutinins, Viral/administration & dosage , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Licensure , Aged , Antibody Formation , Clinical Trials as Topic , Guidelines as Topic , Hemagglutinins, Viral/adverse effects , Humans , Influenza Vaccines/adverse effects , Injections, Intramuscular , Retrospective Studies , United States , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/adverse effects
15.
Drug Dev Ind Pharm ; 36(5): 581-93, 2010 May.
Article in English | MEDLINE | ID: mdl-19954407

ABSTRACT

PURPOSE: The objective of this study was to investigate the potential of rapidly gelling nasal inserts as vaccine delivery system. METHODS: Nasal inserts were prepared by freeze-drying hydrophilic polymer solutions containing influenza split vaccine. In vitro vaccine release from polymer solutions and inserts and the vaccine hemagglutination activity were determined. In vivo immunization studies in mice and rats were performed with nasal solutions and nasal inserts. RESULTS: The in vitro release of proteins (vaccine) from polymeric solutions and inserts was incomplete because of the high molecular weight of the proteins. The release rate was controlled by the polymer (Lutrol F68 > PVP 90 > HPMC K15M > Carbopol > chitosan > or = carrageenan = xanthan gum) because of differences in solution viscosity and possible polymer-protein interactions. Xanthan gum, a negatively charged polymer with intrinsic adjuvanticity, enhanced the serum IgG as well as the nasal IgA response in in vivo studies with nasal vaccine solutions. Poly-l-arginine and cationic lipid were the best performing adjuvants. Solutions containing vaccine with xanthan gum and cationic lipid were effectively stabilized with 0.4 M NaCl. DISCUSSION: The specific activity of the major vaccine protein, hemagglutinin, was not significantly affected by the addition of polymers and the freeze-drying process during insert preparation. The addition of cationic lipid as adjuvant decreased the hemagglutination activity, which strongly indicated inhibition of the protein binding site to erythrocytes. Inserts prepared from xanthan gum and cationic lipid stabilized with NaCl showed a reduced protein activity but were superior to the cationic lipid alone. CONCLUSION: Rat immunization with solid nasal inserts based on xanthan gum containing the influenza vaccine, with or without an additional cationic lipid adjuvant, resulted in similar IgG levels as the pure nasal liquid vaccine formulation.


Subject(s)
Drug Carriers/administration & dosage , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/administration & dosage , Administration, Intranasal , Animals , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Female , Gels , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/pharmacokinetics , Mice , Polysaccharides, Bacterial/administration & dosage , Polysaccharides, Bacterial/metabolism , Rats , Rats, Sprague-Dawley
17.
Avian Dis ; 52(2): 260-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18646455

ABSTRACT

The potency of inactivated Newcastle disease virus (NDV) vaccines in the United States is currently determined using vaccination and challenge of experimental animals against a velogenic strain of NDV. Because velogenic strains of NDV are now classified as select agents in the United States, all vaccine potency testing must be performed in live animals under biosafety level 3 agriculture conditions. If the minimum amount of inactivated viral antigen required for clinical protection can be determined using other methods, vaccines meeting these criteria might be considered of adequate potency. The linearity of correlation between the hemagglutination (HA) assay measurement and the 50% embryo infectious dose titer ofNDV Hitchner B1 vaccine virus was determined. Correlation between hemagglutinin units (HAU) per vaccine dose, clinical protection, and antibody response was then determined using a vaccinate-and-challenge model similar to Chapter 9 of the U.S. code of federal regulations approved method for vaccine potency testing. The dose providing 50% protection of an in-house water-in-oil emulsion vaccine formulated with inactivated NDV B1 was determined to be between 400 and 600 HAU from two separate trials. A positive correlation (R2 = 0.97) was observed between antibody response and HAU per vaccine dose. Serum antibody responses from vaccinated birds indicate HA inhibition titers >2(5) log2 would provide 100% protection from morbidity and mortality and require a minimum protective dose of 1000 HAU per bird. These are the first studies to examine establishing both a minimum protective HAU content for inactivated ND vaccines and a minimum serologic response necessary to ensure potency.


Subject(s)
Hemagglutinins, Viral/administration & dosage , Newcastle Disease/prevention & control , Newcastle disease virus/immunology , Viral Vaccines/immunology , Animals , Chick Embryo , Chickens , Dose-Response Relationship, Immunologic , Hemagglutination Inhibition Tests/veterinary , Hemagglutinins, Viral/analysis , Newcastle Disease/immunology , Newcastle Disease/virology , Newcastle disease virus/isolation & purification , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/analysis , Vaccines, Inactivated/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/analysis
18.
J Immunol ; 169(7): 3622-9, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12244153

ABSTRACT

It has long been established that exposure of naive T cells to specific Ag in the absence of adjuvant leads to tolerization. Nonetheless, the potential of effector CD4 cells to be tolerized has been less well characterized. To address this issue, we have used an adoptive transfer system in which naive TCR transgenic hemagglutinin (HA)-specific CD4(+) T cells are initially primed to express effector function upon exposure to an immunogenic recombinant vaccinia virus expressing HA, and then exposed to forms of HA that are tolerogenic for naive CD4 cells. HA-specific effector CD4 cells residing in both the spleen as well as in two separate nonlymphoid tissues were tolerized upon exposure to high doses of exogenous soluble HA peptide. Additionally, tolerance could also be induced by bone marrow-derived APCs that cross-present parenchymally derived self-HA. Thus, effector CD4 cells are susceptible to similar tolerogenic stimuli as are naive CD4 cells.


Subject(s)
Autoantigens/immunology , CD4-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/biosynthesis , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Immune Tolerance/genetics , Interphase/immunology , Liver/cytology , Liver/immunology , Liver/virology , Lung/cytology , Lung/immunology , Lung/virology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphoid Tissue/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligopeptides/administration & dosage , Oligopeptides/immunology , Solubility , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/virology , Vaccinia virus/genetics , Vaccinia virus/immunology
19.
Vaccine ; 19(15-16): 2163-71, 2001 Feb 28.
Article in English | MEDLINE | ID: mdl-11228389

ABSTRACT

Measles remains a significant problem in both the developed and developing world, and new measles vaccination strategies need to be developed. This paper examines the strategy of utilizing transgenic plants expressing a measles antigen for the development of an oral sub-unit measles vaccine. A 1.8 kb fragment encompassing the coding region of the measles virus hemagglutinin (H) protein was cloned into a plant expression cassette. Three different expression constructs were tested: pBinH (H gene alone), pBinH/KDEL (addition of a C-terminal endoplasmic reticulum-retention sequence SEKDEL) and pBinSP/H/KDEL (further addition of an authentic N-terminal plant signal peptide). The highest levels of recombinant H protein production were observed in plants transformed with pBinH/KDEL. Mice inoculated intraperitoneally with transgenic plant derived recombinant H protein produced serum anti-H protein antibodies that neutralized the measles virus (MV) in vitro. Mice gavaged with transgenic tobacco leaf extracts also developed serum H protein-specific antibodies with neutralizing activity against MV in vitro. These results indicate that the plant-derived measles H protein is immunogenic when administered orally and that, with further development, oral vaccination utilizing transgenic plants may become a viable approach to measles vaccine development.


Subject(s)
Hemagglutinins, Viral/immunology , Measles virus/immunology , Administration, Oral , Animals , Antibodies, Viral/blood , Base Sequence , Female , Genetic Vectors , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/genetics , Immunoglobulin G/blood , Injections, Intraperitoneal , Measles Vaccine/administration & dosage , Measles Vaccine/genetics , Measles Vaccine/isolation & purification , Measles virus/genetics , Mice , Mice, Inbred BALB C , Neutralization Tests , Plants, Genetically Modified , Plants, Toxic , Plasmids/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Nicotiana/genetics
20.
J Biomater Sci Polym Ed ; 10(8): 805-25, 1999.
Article in English | MEDLINE | ID: mdl-10487316

ABSTRACT

The aim of this study was to evaluate the potential of poly(D,L-lactide-co-glycolide) nano-and microspheres, with a mean diameter of 220 nm and 8 microm, respectively, to enhance the nasal and systemic immune responses against influenza virus antigen. High encapsulation levels of antigen were achieved in all cases. Neither the molecular weight nor the antigenicity of the entrapped antigen were affected by the encapsulation procedure. Following nasal immunization, the nasal washes IgA and the serum IgG responses were evaluated. With the soluble antigen, relatively high immune responses were observed. With nanospheres, nasal washes IgA levels were significantly lower (p<0.01) and serum IgG levels were not significantly different (p>0.05) from those obtained with the soluble antigen. With microspheres, both nasal washes IgA and serum IgG levels were significantly lower (p<0.01 and <0.05, respectively) as compared to the levels found for the soluble antigen. In addition, fluorescent microspheres administered intranasally failed to reach the nasal-associated lymphoid tissue (NALT). This lack of particle uptake by NALT and the high immunogenicity of the antigen used in this study, could explain the absence of enhancement of the immune responses by the polymeric particles.


Subject(s)
Orthomyxoviridae/immunology , Vaccination/methods , Administration, Intranasal , Animals , Antibody Formation , Antigens, Viral/immunology , Antigens, Viral/pharmacology , Biocompatible Materials , Drug Compounding/methods , Epitopes , Female , Hemagglutinins, Viral/administration & dosage , Hemagglutinins, Viral/pharmacology , Immunoglobulin A, Secretory/drug effects , Immunoglobulin A, Secretory/metabolism , Immunoglobulin G/blood , Immunoglobulin G/drug effects , Lactic Acid , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Microspheres , Molecular Weight , Particle Size , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...