Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Vaccine ; 42(9): 2220-2228, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38582606

ABSTRACT

The yearly epidemics and unpredictable outbreaks of influenza have raisedserious concernsglobally and led to prioritizing the development of an effective vaccine toprotectagainst newly emerging variants. Previously, we demonstrated that monoglycosylated influenza virus vaccines derived from A/California/7/2009 or an updated A/Brisbane/02/2018 (IVR-190) vaccine strain recommended by WHO are superior to fully glycosylated vaccines and could broadly protect against past and new coming H1N1 variants. However, whether such a monoglycosylated virus vaccine can be mass-produced to meet clinical demands and stable enough to provide consistent efficacy against H1N1 viruses remains unclear. Herein, we developed a platform for the pilot-scale production of the monoglycosylated split virus vaccine from the IVR-190 strain (IVR-190mg) with a robust and cost-effective manufacturing process. The critical parameters of inoculum dose, concentration of kifunensine, and optimized Endo H treatment process were comprehensively investigated. Several aims for preclinical studies of IVR-190mg were achieved, including [i] the execution of three engineering batch runs to validate lot-to-lot consistency, [ii] the establishment of IVR-190mg specifications to meet the acceptance criteria of a conventional influenza vaccine, [iii] an investigation of the stability profile of IVR-190mg, and completion of a safety evaluation by conducting an animal toxicology study. The toxicology study under GLP guidance found no systemic toxicity after rabbits were vaccinated with IVR-190mg. The serological data showed that IVR-190mg is highly immunogenic and effective in inducing a cross-strain protective level of antibody immune responses, including hemagglutination-inhibition titers, viral neutralization activity, and broad HA- and NA-inhibiting antibody titers against past and new H1N1 viruses. In conclusion, this study provides efficacy and safety profiles of IVR-190mg for further clinical study and shows that this vaccine without a glycan shield has great potential to be safe and protective against H1N1 variants.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Rabbits , Humans , Influenza, Human/prevention & control , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype
2.
Antiviral Res ; 220: 105759, 2023 12.
Article in English | MEDLINE | ID: mdl-37984568

ABSTRACT

The SARS-CoV-2 and influenza pandemics have posed a devastating threat to global public health. The best strategy for preventing the further spread of these respiratory viruses worldwide is to administer a vaccine capable of targeting both viruses. Here, we show that a novel monoglycosylated vaccine designed based on the influenza virus HAstem conserved domain fused with the SARS-CoV-2 spike-RBD domain (HSSRmg) can present proper antigenicity that elicits sufficient neutralization efficacy against various SARS-CoV-2 variants while simultaneously providing broad protection against H1N1 viruses in mice. Compared with the fully glycosylated HSSR (HSSRfg), HSSRmg induced higher ELISA titers targeting HAstem and spike-RBD and exhibited significantly enhanced neutralization activity against the Wuhan pseudovirus. The enhanced immune responses raised by JR300-adjuvanted HSSRmg compared to HSSRmg alone include more anti-HAstem and anti-spike-RBD antibodies that provide cross-protection against H1N1 challenges and cross-neutralization of SARS-CoV-2 pseudoviruses. Furthermore, the enhanced immune response raised by JR300-adjuvanted-HSSRmg skews toward a more balanced Th1/Th2 response than that raised by HSSRmg alone. Notably, HSSRmg elicited more plasma B cells and memory B cells, and higher IL-4 and IFN-γ cytokine immune responses than spike (S-2P) in mice with preexisting influenza-specific immunity, suggesting that B-cell activation most likely occurs through CD4+ T-cell stimulation. This study demonstrated that HSSRmg produced using a monoglycosylation process and combined with the JR300 adjuvant elicits superior cross-strain immune responses against SARS-CoV-2 and influenza viruses in mice compared with S-2P. JR300-adjuvanted HSSRmg has great potential as a coronavirus-influenza vaccine that provides dual protection against SARS-CoV-2 and influenza infections.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Viral Vaccines , Animals , Mice , Humans , SARS-CoV-2 , COVID-19/prevention & control , Antibodies, Viral , Antibodies, Neutralizing , Spike Glycoprotein, Coronavirus
3.
Antiviral Res ; 216: 105640, 2023 08.
Article in English | MEDLINE | ID: mdl-37263355

ABSTRACT

Influenza epidemics and pandemics caused by newly emerging virus strains highlight an urgent need to develop a universal vaccine against viruses. Previously, a monoglycosylated X-181mg vaccine demonstrated that the HA possessing a single N-acetylglucosamine at each N-glycosylation site is superior to confer broader protection in mice than conventional vaccines. However, the greatest challenge in conducting clinical trials is the need to develop robust manufacturing processes capable of producing vaccines at the pilot scale with the desired stability, potency, and efficacy. Whether the monoglycosylated virus vaccine platform can be applied to the new vaccine strain in a timely manner and whether the mass-produced vaccine has the proper immunogenicity to induce cross-protective immunity remains unclear. Here, we show that a pilot-scale manufacturing process produced a monoglycosylated A/Brisbane/02/2018(H1N1) virus vaccine (IVR-190mg) with a single glycan at each glycosylation site of HA and NA. Compared with the fully glycosylated virus vaccine (IVR-190fg), the IVR-190mg provided broader cross-protection in mice against a wide range of H1N1 variants. The enhanced antibody responses induced by IVR-190mg immunization include higher hemagglutination-inhibition titers, higher neutralization activity, more anti-HA head domain, more anti-HA stem antibodies, higher neuraminidase activity inhibition titers, and notably, higher antibody-dependent cellular cytotoxicity. Additionally, the IVR-190mg also induced a more balanced Th1/Th2 response and elicited broader splenic CD4+ and CD8+ T-cell responses than IVR-190fg. This study demonstrated that IVR-190mg produced using a pilot-scale manufacturing process elicits comprehensive cross-strain immune responses that have great potential to substantially mitigate the need for yearly reformulation of strain-specific inactivated vaccines.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Mice , Humans , Antibodies, Viral , Vaccines, Inactivated , Hemagglutinin Glycoproteins, Influenza Virus
4.
PLoS Pathog ; 17(8): e1009724, 2021 08.
Article in English | MEDLINE | ID: mdl-34352041

ABSTRACT

Hemagglutinin (HA) is the immunodominant protein of the influenza virus. We previously showed that mice injected with a monoglycosylated influenza A HA (HAmg) produced cross-strain-reactive antibodies and were better protected than mice injected with a fully glycosylated HA (HAfg) during lethal dose challenge. We employed a single B-cell screening platform to isolate the cross-protective monoclonal antibody (mAb) 651 from mice immunized with the HAmg of A/Brisbane/59/2007 (H1N1) influenza virus (Bris/07). The mAb 651 recognized the head domain of a broad spectrum of HAs from groups 1 and 2 influenza A viruses and offered prophylactic and therapeutic efficacy against A/California/07/2009 (H1N1) (Cal/09) and Bris/07 infections in mice. The antibody did not possess neutralizing activity; however, antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis mediated by natural killer cells and alveolar macrophages were important in the protective efficacy of mAb 651. Together, this study highlighted the significance of effector functions for non-neutralizing antibodies to exhibit protection against influenza virus infection.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antibody-Dependent Cell Cytotoxicity , Influenza A virus/immunology , Killer Cells, Natural/immunology , Macrophages, Alveolar/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/virology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
5.
J Biomed Sci ; 27(1): 33, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32059697

ABSTRACT

Vaccination is the most effective measure at preventing influenza virus infections. However, current seasonal influenza vaccines are only protective against closely matched circulating strains. Even with extensive monitoring and annual reformulation our efforts remain one step behind the rapidly evolving virus, often resulting in mismatches and low vaccine effectiveness. Fortunately, many next-generation influenza vaccines are currently in development, utilizing an array of innovative techniques to shorten production time and increase the breadth of protection. This review summarizes the production methods of current vaccines, recent advances that have been made in influenza vaccine research, and highlights potential challenges that are yet to be overcome. Special emphasis is put on the potential role of glycoengineering in influenza vaccine development, and the advantages of removing the glycan shield on influenza surface antigens to increase vaccine immunogenicity. The potential for future development of these novel influenza vaccine candidates is discussed from an industry perspective.


Subject(s)
Glycoproteins/immunology , Immunogenicity, Vaccine , Influenza Vaccines/immunology , Protein Engineering , Viral Proteins/immunology , Glycoproteins/chemistry , Glycoproteins/pharmacology , Glycosylation , Humans , Influenza Vaccines/analysis , Influenza Vaccines/chemistry , Influenza Vaccines/pharmacology , Viral Proteins/chemistry , Viral Proteins/pharmacology
6.
Proc Natl Acad Sci U S A ; 116(10): 4200-4205, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782805

ABSTRACT

Each year influenza virus infections cause hundreds of thousands of deaths worldwide and a significant level of morbidity with major economic burden. At the present time, vaccination with inactivated virus vaccine produced from embryonated chicken eggs is the most prevalent method to prevent the infections. However, current influenza vaccines are only effective against closely matched circulating strains and must be updated and administered yearly. Therefore, generating a vaccine that can provide broad protection is greatly needed for influenza vaccine development. We have previously shown that vaccination of the major surface glycoprotein hemagglutinin (HA) of influenza virus with a single N-acetylglucosamine at each of the N-glycosylation sites [monoglycosylated HA (HAmg)] can elicit better cross-protection compared with the fully glycosylated HA (HAfg). In the current study, we produced monoglycosylated inactivated split H1N1 virus vaccine from chicken eggs by the N-glycosylation process inhibitor kifunensine and the endoglycosidase Endo H, and intramuscularly immunized mice to examine its efficacy. Compared with vaccination of the traditional influenza vaccine with complex glycosylations from eggs, the monoglycosylated split virus vaccine provided better cross-strain protection against a lethal dose of virus challenge in mice. The enhanced antibody responses induced by the monoglycosylated vaccine immunization include higher neutralization activity, higher hemagglutination inhibition, and more HA stem selectivity, as well as, interestingly, higher antibody-dependent cellular cytotoxicity. This study provides a simple and practical procedure to enhance the cross-strain protection of influenza vaccine by removing the outer part of glycans from the virus surface through modifications of the current egg-based process.


Subject(s)
Cross Protection/immunology , Eggs , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/prevention & control , Vaccination , Animals , Chickens/abnormalities , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinins/immunology , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/prevention & control , Injections, Intramuscular , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/immunology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
7.
Appl Opt ; 54(28): E241-8, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26479660

ABSTRACT

This paper proposes a capsule endoscope (CE), based on color multiplexing, to simultaneously record front and side images. Only one lens associated with an X-cube prism is employed to catch the front and side view profiles in the CE. Three color filters and polarizers are placed on three sides of an X-cube prism. When objects locate at one of the X-cube's three sides, front and side view profiles of different colors will be caught through the proposed lens and recorded at the color image sensor. The proposed color multiplexing CE (CMCE) is designed with a field of view of up to 210 deg and a 180 lp/mm resolution under f-number 2.8 and overall length 13.323 mm. A ray-tracing simulation in the CMCE with the color multiplexing mechanism verifies that the CMCE not only records the front and side view profiles at the same time, but also has great image quality at a small size.


Subject(s)
Capsule Endoscopes , Endoscopy/methods , Algorithms , Color , Colorimetry/methods , Computer Simulation , Endoscopy/instrumentation , Equipment Design , Humans , Image Enhancement/methods , Image Processing, Computer-Assisted/methods , Lenses , Liquid Crystals , Silicon/chemistry
8.
Proc Natl Acad Sci U S A ; 112(34): 10611-6, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26253764

ABSTRACT

Antibodies have been developed as therapeutic agents for the treatment of cancer, infection, and inflammation. In addition to binding activity toward the target, antibodies also exhibit effector-mediated activities through the interaction of the Fc glycan and the Fc receptors on immune cells. To identify the optimal glycan structures for individual antibodies with desired activity, we have developed an effective method to modify the Fc-glycan structures to a homogeneous glycoform. In this study, it was found that the biantennary N-glycan structure with two terminal alpha-2,6-linked sialic acids is a common and optimized structure for the enhancement of antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, and antiinflammatory activities.


Subject(s)
Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Polysaccharides/chemistry , Rituximab/chemistry , Acetylglucosamine/chemistry , Acetylglucosamine/immunology , Animals , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Bacterial Proteins/metabolism , Bacteroides fragilis/enzymology , Cell Line, Tumor , Female , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred BALB C , Neuraminidase/metabolism , Orthomyxoviridae Infections/prevention & control , Protein Engineering , Receptors, IgG/immunology , Rituximab/immunology , Sialic Acids/chemistry , Sialic Acids/immunology , Streptococcus pyogenes/enzymology , Structure-Activity Relationship , Trastuzumab/chemistry , Trastuzumab/immunology , alpha-L-Fucosidase/metabolism
9.
Proc Natl Acad Sci U S A ; 111(7): 2476-81, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24469815

ABSTRACT

The 2009 H1N1 pandemic and recent human cases of H5N1, H7N9, and H6N1 in Asia highlight the need for a universal influenza vaccine that can provide cross-strain or even cross-subtype protection. Here, we show that recombinant monoglycosylated hemagglutinin (HAmg) with an intact protein structure from either seasonal or pandemic H1N1 can be used as a vaccine for cross-strain protection against various H1N1 viruses in circulation from 1933 to 2009 in mice and ferrets. In the HAmg vaccine, highly conserved sequences that were originally covered by glycans in the fully glycosylated HA (HAfg) are exposed and thus, are better engulfed by dendritic cells (DCs), stimulated better DC maturation, and induced more CD8+ memory T cells and IgG-secreting plasma cells. Single B-cell RT-PCR followed by sequence analysis revealed that the HAmg vaccine activated more diverse B-cell repertoires than the HAfg vaccine and produced antibodies with cross-strain binding ability. In summary, the HAmg vaccine elicits cross-strain immune responses that may mitigate the current need for yearly reformulation of strain-specific inactivated vaccines. This strategy may also map a new direction for universal vaccine design.


Subject(s)
Drug Design , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Immunity, Cellular/immunology , Influenza Vaccines/pharmacology , Influenza, Human/prevention & control , Orthomyxoviridae/immunology , Animals , Base Sequence , Chromatography, Liquid , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Female , Ferrets , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Homology , Species Specificity , Tandem Mass Spectrometry
10.
Proc Natl Acad Sci U S A ; 106(43): 18137-42, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19822741

ABSTRACT

Recent cases of avian influenza H5N1 and the swine-origin 2009 H1N1 have caused a great concern that a global disaster like the 1918 influenza pandemic may occur again. Viral transmission begins with a critical interaction between hemagglutinin (HA) glycoprotein, which is on the viral coat of influenza, and sialic acid (SA) containing glycans, which are on the host cell surface. To elucidate the role of HA glycosylation in this important interaction, various defined HA glycoforms were prepared, and their binding affinity and specificity were studied by using a synthetic SA microarray. Truncation of the N-glycan structures on HA increased SA binding affinities while decreasing specificity toward disparate SA ligands. The contribution of each monosaccharide and sulfate group within SA ligand structures to HA binding energy was quantitatively dissected. It was found that the sulfate group adds nearly 100-fold (2.04 kcal/mol) in binding energy to fully glycosylated HA, and so does the biantennary glycan to the monoglycosylated HA glycoform. Antibodies raised against HA protein bearing only a single N-linked GlcNAc at each glycosylation site showed better binding affinity and neutralization activity against influenza subtypes than the fully glycosylated HAs elicited. Thus, removal of structurally nonessential glycans on viral surface glycoproteins may be a very effective and general approach for vaccine design against influenza and other human viruses.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines/immunology , Polysaccharides/chemistry , Polysaccharides/metabolism , Receptors, Virus/metabolism , Animals , Cell Line , Female , Glycomics , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/metabolism , Influenza, Human/immunology , Influenza, Human/prevention & control , Mice , Mice, Inbred BALB C , Microarray Analysis , Models, Molecular , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Polysaccharides/analysis , Protein Binding , Protein Structure, Tertiary , Receptors, Virus/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...