1
|
Heinimäki S, Lampinen V, Tamminen K, Hankaniemi MM, Malm M, Hytönen VP, Blazevic V. Antigenicity and immunogenicity of HA2 and M2e influenza virus antigens conjugated to norovirus-like, VP1 capsid-based particles by the SpyTag/SpyCatcher technology. Virology 2021; 566:89-97. [PMID: 34894525 DOI: 10.1016/j.virol.2021.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Virus-like particles (VLPs) modified through different molecular technologies are employed as delivery vehicles or platforms for heterologous antigen display. We have recently created a norovirus (NoV) VLP platform, where two influenza antigens, the extracellular domain of matrix protein M2 (M2e) or the stem domain of the major envelope glycoprotein hemagglutinin (HA2) are displayed on the surface of the NoV VLPs by SpyTag/SpyCatcher conjugation. To demonstrate the feasibility of the platform to deliver foreign antigens, this study examined potential interference of the conjugation with induction of antibodies against conjugated M2e peptide, HA2, and NoV VLP carrier. High antibody response was induced by HA2 but not M2e decorated VLPs. Furthermore, HA2-elicited antibodies did not neutralize the homologous influenza virus in vitro. Conjugated NoV VLPs retained intact receptor binding capacity and self-immunogenicity. The results demonstrate that NoV VLPs could be simultaneously used as a platform to deliver foreign antigens and a NoV vaccine.
Collapse
Affiliation(s)
- Suvi Heinimäki
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Vili Lampinen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi Tamminen
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna M Hankaniemi
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Malm
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vesa P Hytönen
- Protein Dynamics Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Vesna Blazevic
- Vaccine Development and Immunology/Vaccine Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
2
|
Motawi TK, Shahin NN, Awad K, Maghraby AS, Abd-Elshafy DN, Bahgat MM. Glycolytic and immunological alterations in human U937 monocytes in response to H1N1 infection. IUBMB Life 2020; 72:2481-2498. [PMID: 32941696 DOI: 10.1002/iub.2378] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 11/06/2022]
Abstract
We monitored changes that took place in glycolytic enzymes, the pyruvate end product of glycolysis, tumor necrosis factor α (TNFα), and toll-like receptors (TLRs) both at the transcriptional and translational levels upon direct interaction between PR8-H1N1 and the human monocytes U937 in vitro system. U937 were first treated with H1N1 infectious viral particles or phorbol-12-myristate-13-acetate (PMA) or left untreated and later infected with the H1N1 virus. Levels of phosphofructokinase 1 (PFK1) and pyruvate were biochemically quantified. In addition, levels of TNFα, TLR3, and TLR7 were measured by ELISA. The transcriptional profiles of PFKs, inflammatory cytokines, TLR3 and TLR7 were relatively quantified by qRT-PCR. The results generally revealed significant changes in both the transcriptional and translational profiles of the studied biochemical and immunological parameters upon influenza infection in a time-dependent manner. In conclusion, H1N1 infection triggers transcriptional and translational changes in immortalized human monocytes, which might serve as markers for infection subject for further validation for their specificities.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nancy Nabil Shahin
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Kareem Awad
- Research Group Immune- and Bio-markers for Infection, the Center of Excellence for Advanced Sciences, the National Research Center, Cairo, Egypt.,Department of Therapeutic Chemistry, Division of Pharmaceutical and Drug Industries Research, the National Research Center, Cairo, Egypt
| | - Amany Sayed Maghraby
- Research Group Immune- and Bio-markers for Infection, the Center of Excellence for Advanced Sciences, the National Research Center, Cairo, Egypt.,Department of Therapeutic Chemistry, Division of Pharmaceutical and Drug Industries Research, the National Research Center, Cairo, Egypt
| | - Dina Nadeem Abd-Elshafy
- Research Group Immune- and Bio-markers for Infection, the Center of Excellence for Advanced Sciences, the National Research Center, Cairo, Egypt.,Department of Water Pollution Research, Division of Environmental Research, the National Research Center, Cairo, Egypt
| | - Mahmoud Mohamed Bahgat
- Research Group Immune- and Bio-markers for Infection, the Center of Excellence for Advanced Sciences, the National Research Center, Cairo, Egypt.,Department of Therapeutic Chemistry, Division of Pharmaceutical and Drug Industries Research, the National Research Center, Cairo, Egypt
| |
Collapse
|
3
|
Loes AN, Gentles LE, Greaney AJ, Crawford KHD, Bloom JD. Attenuated Influenza Virions Expressing the SARS-CoV-2 Receptor-Binding Domain Induce Neutralizing Antibodies in Mice. Viruses 2020; 12:E987. [PMID: 32899480 PMCID: PMC7552029 DOI: 10.3390/v12090987] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 08/28/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
An effective vaccine is essential for controlling the spread of the SARS-CoV-2 virus. Here, we describe an influenza virus-based vaccine for SARS-CoV-2. We incorporated a membrane-anchored form of the SARS-CoV-2 spike receptor binding domain (RBD) in place of the neuraminidase (NA) coding sequence in an influenza virus also possessing a mutation that reduces the affinity of hemagglutinin for its sialic acid receptor. The resulting ΔNA(RBD)-Flu virus can be generated by reverse genetics and grown to high titers in cell culture. A single-dose intranasal inoculation of mice with ΔNA(RBD)-Flu elicits serum neutralizing antibody titers against SAR-CoV-2 comparable to those observed in humans following natural infection (~1:200). Furthermore, ΔNA(RBD)-Flu itself causes no apparent disease in mice. It might be possible to produce a vaccine similar to ΔNA(RBD)-Flu at scale by leveraging existing platforms for the production of influenza vaccines.
Collapse
Affiliation(s)
- Andrea N. Loes
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
| | - Lauren E. Gentles
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
| | - Allison J. Greaney
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Katharine H. D. Crawford
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Jesse D. Bloom
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; (A.N.L.); (L.E.G.); (A.J.G.); (K.H.D.C.)
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
4
|
Loes AN, Gentles LE, Greaney AJ, Crawford KHD, Bloom JD. Attenuated influenza virions expressing the SARS-CoV-2 receptor-binding domain induce neutralizing antibodies in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.08.12.248823. [PMID: 32817935 PMCID: PMC7430565 DOI: 10.1101/2020.08.12.248823] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
An effective vaccine is essential to controlling the spread of SARS-CoV-2 virus. Here, we describe an influenza-virus-based vaccine for SARS-CoV-2. We incorporated a membrane-anchored form of the SARS-CoV-2 Spike receptor binding domain (RBD) in place of the neuraminidase (NA) coding sequence in an influenza virus also possessing a mutation that reduces the affinity of hemagglutinin for its sialic acid receptor. The resulting ΔNA(RBD)-Flu virus can be generated by reverse genetics and grown to high titers in cell culture. A single-dose intranasal inoculation of mice with ΔNA(RBD)-Flu elicits serum neutralizing antibody titers against SAR-CoV-2 comparable to those observed in humans following natural infection (~1:200). Furthermore, ΔNA(RBD)-Flu itself causes no apparent disease in mice. It might be possible to produce a vaccine similar to ΔNA(RBD)-Flu at scale by leveraging existing platforms for production of influenza vaccines.
Collapse
Affiliation(s)
- Andrea N. Loes
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
| | - Lauren E. Gentles
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
| | - Allison J. Greaney
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Katharine H. D. Crawford
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA 98195, USA
| | - Jesse D. Bloom
- Division of Basic Sciences and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, Seattle, WA 98103, USA
- Department of Microbiology, University of Washington, Seattle, WA 98195-7735, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Motawi TK, Shahin NN, Maghraby AS, Kirschfink M, Nadeem Abd-Elshafy D, Awad K, Bahgat MM. H1N1 Infection Reduces Glucose Level in Human U937 Monocytes Culture. Viral Immunol 2020; 33:384-390. [PMID: 32202987 DOI: 10.1089/vim.2019.0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Infection with influenza A (H1N1) virus contributes significantly to the global burden of acute respiratory diseases. Glucose uptake and metabolic changes are reported in different cell types after infections with different virus types, including influenza A virus. Alteration of glucose metabolism specifically in immune cells has major health consequences. The aim of this study was to monitor glucose concentration in unstimulated and stimulated U937 human monocytes with infectious or heat inactivated H1N1 or Staphylococcus aureus or in nonpathogenically stimulated monocytes with phorbol-12-myristate-13-acetate. Stimulated or unstimulated U937 human monocytes were subjected to H1N1 infection for different time points and the glucose profile in the growth medium was measured post infection. Results showed that regardless to whether the initial stimuli on U937 cells were of pathogen or nonpathogen origins, challenge infection by H1N1 causes a significant reduction of glucose levels 36 h post infection. In conclusion, H1N1 infection has a direct effect on the glucose uptake of U937 cells in vitro. This effect can be related to either H1N1 infection or cell differentiation status that might occur due to the exerted stimuli.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nancy Nabil Shahin
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Amany Sayed Maghraby
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt.,Division of Pharmaceutical and Drug Industries Research, Department of Therapeutic Chemistry, National Research Center, Cairo, Egypt
| | - Michael Kirschfink
- Institute of Immunology, Ruprecht Karls University of Heidelberg, Heidelberg, Germany
| | - Dina Nadeem Abd-Elshafy
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt.,Division of Environmental Research, Department of Water Pollution Research, National Research Center, Cairo, Egypt
| | - Kareem Awad
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt.,Division of Pharmaceutical and Drug Industries Research, Department of Therapeutic Chemistry, National Research Center, Cairo, Egypt
| | - Mahmoud Mohamed Bahgat
- Research Group Immune- and Bio-Markers for Infection, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt.,Division of Pharmaceutical and Drug Industries Research, Department of Therapeutic Chemistry, National Research Center, Cairo, Egypt
| |
Collapse
|
6
|
Nichols JE, Niles JA, Fleming EH, Roberts NJ. The role of cell surface expression of influenza virus neuraminidase in induction of human lymphocyte apoptosis. Virology 2019; 534:80-86. [PMID: 31220651 DOI: 10.1016/j.virol.2019.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/08/2023]
Abstract
The immunopathological mechanisms as well as the role played by influenza A virus infection of human leukocytes and induction of apoptosis have not been fully elucidated. We confirm here that the percentage of cells that are infected is less than the percent of apoptotic cells. Depletion of monocytes/macrophages and depletion of cells expressing influenza neuraminidase from the cultures after exposure to virus decreased lymphocyte apoptosis. Treatment of virus-exposed leukocyte cultures with anti-neuraminidase antibodies but not with anti-hemagglutinin antibodies, reduced lymphocyte production of active caspase-3 and induction of apoptosis. Different strains of virus induced different levels of apoptosis. Variations in induction of apoptosis correlated with production and expression of viral neuraminidase by infected leukocytes. The data suggest that cell surface expression of neuraminidase plays an important role in the induction of apoptosis in human lymphocytes. The benefit, or cost, to the host of lymphocyte apoptosis warrants continued investigation.
Collapse
Affiliation(s)
- Joan E Nichols
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Texas, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Jean A Niles
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Texas, USA
| | - Elisa H Fleming
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Texas, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Texas, USA
| | - Norbert J Roberts
- Division of Infectious Diseases, Department of Internal Medicine, University of Texas Medical Branch at Galveston, Texas, USA; Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Texas, USA; Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Spies C, Luetz A, Lachmann G, Renius M, von Haefen C, Wernecke KD, Bahra M, Schiemann A, Paupers M, Meisel C. Influence of Granulocyte-Macrophage Colony-Stimulating Factor or Influenza Vaccination on HLA-DR, Infection and Delirium Days in Immunosuppressed Surgical Patients: Double Blind, Randomised Controlled Trial. PLoS One 2015; 10:e0144003. [PMID: 26641243 PMCID: PMC4671639 DOI: 10.1371/journal.pone.0144003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
PURPOSE Surgical patients are at high risk for developing infectious complications and postoperative delirium. Prolonged infections and delirium result in worse outcome. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and influenza vaccination are known to increase HLA-DR on monocytes and improve immune reactivity. This study aimed to investigate whether GM-CSF or vaccination reverses monocyte deactivation. Secondary aims were whether it decreases infection and delirium days after esophageal or pancreatic resection over time. METHODS In this prospective, randomized, placebo-controlled, double-blind, double dummy trial setting on an interdisciplinary ICU of a university hospital 61 patients with immunosuppression (monocytic HLA-DR [mHLA-DR] <10,000 monoclonal antibodies [mAb] per cell) on the first day after esophageal or pancreatic resection were treated with either GM-CSF (250 μg/m2/d), influenza vaccination (Mutagrip 0.5 ml/d) or placebo for a maximum of 3 consecutive days if mHLA-DR remained below 10,000 mAb per cell. HLA-DR on monocytes was measured daily until day 5 after surgery. Infections and delirium were followed up for 9 days after surgery. Primary outcome was HLA-DR on monocytes, and secondary outcomes were duration of infection and delirium. RESULTS mHLA-DR was significantly increased compared to placebo (p < 0.001) and influenza vaccination (p < 0.001) on the second postoperative day. Compared with placebo, GM-CSF-treated patients revealed shorter duration of infection (p < 0.001); the duration of delirium was increased after vaccination (p = 0.003). CONCLUSION Treatment with GM-CSF in patients with postoperative immune suppression was safe and effective in restoring monocytic immune competence. Furthermore, therapy with GM-CSF reduced duration of infection in immune compromised patients. However, influenza vaccination increased duration of delirium after major surgery. TRIAL REGISTRATION www.controlled-trials.com ISRCTN27114642.
Collapse
Affiliation(s)
- Claudia Spies
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
- * E-mail:
| | - Alawi Luetz
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Gunnar Lachmann
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Markus Renius
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | | | - Marcus Bahra
- Department of General, Abdominal and Transplantation Surgery, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Alexander Schiemann
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Marco Paupers
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow-Klinikum, Charité – Universitätsmedizin, Berlin, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité – Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| |
Collapse
|
8
|
Li J, Diaz-Arévalo D, Chen Y, Zeng M. Intranasal vaccination with an engineered influenza virus expressing the receptor binding subdomain of botulinum neurotoxin provides protective immunity against botulism and influenza. Front Immunol 2015; 6:170. [PMID: 25954272 PMCID: PMC4404949 DOI: 10.3389/fimmu.2015.00170] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/27/2015] [Indexed: 11/24/2022] Open
Abstract
Influenza virus is a negative segmented RNA virus without DNA intermediate. This makes it safer as a vaccine delivery vector than most DNA viruses that have potential to integrate their genetic elements into host genomes. In this study, we developed a universal influenza viral vector, expressing the receptor binding subdomain of botulinum neurotoxin A (BoNT/A). We tested the growth characters of the engineered influenza virus in chicken eggs and Madin–Darby canine kidney epithelial cells (MDCK), and showed that it can be produced to a titer of 5 × 106 plaque forming unites/ml in chicken eggs and MDCK cells. Subsequently, mice intranasally vaccinated with the engineered influenza virus conferred protection against challenge with lethal doses of active BoNT/A toxin and influenza virus. Our results demonstrated the feasibility to develop a dual purpose nasal vaccine against both botulism and influenza.
Collapse
Affiliation(s)
- Junwei Li
- Department of Biomedical Sciences, Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center , El Paso, TX , USA
| | - Diana Diaz-Arévalo
- Department of Biomedical Sciences, Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center , El Paso, TX , USA
| | - Yanping Chen
- Department of Biomedical Sciences, Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center , El Paso, TX , USA
| | - Mingtao Zeng
- Department of Biomedical Sciences, Center of Excellence for Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center , El Paso, TX , USA
| |
Collapse
|
9
|
Crowe SR, Merrill JT, Vista ES, Dedeke AB, Thompson DM, Stewart S, Guthridge JM, Niewold TB, Franek BS, Air GM, Thompson LF, James JA. Influenza vaccination responses in human systemic lupus erythematosus: impact of clinical and demographic features. ARTHRITIS AND RHEUMATISM 2011; 63:2396-406. [PMID: 21598235 PMCID: PMC3149742 DOI: 10.1002/art.30388] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Vaccination against common pathogens, such as influenza, is recommended for patients with systemic lupus erythematosus (SLE) to decrease infections and improve health. However, most reports describing the vaccination response are limited to evaluations of SLE patients with quiescent disease. This study focuses on understanding the clinical, serologic, therapeutic, and demographic factors that influence the response to influenza vaccination in SLE patients with a broad range of disease activity. METHODS Blood specimens and information on disease activity were collected from 72 patients with SLE, at baseline and at 2, 6, and 12 weeks after influenza vaccination. Influenza-specific antibody responses were assessed by determining the total serum antibody concentration (B(max)), relative affinity (K(a)), and level of hemagglutination inhibition in the plasma. Using a cumulative score, the patients were evenly divided into groups of high or low vaccine responders. Autoantibody levels were evaluated at each time point using immunofluorescence tests and standard enzyme-linked immunosorbent assays. RESULTS Compared to high responders, low responders to the vaccine were more likely to have hematologic criteria (P = 0.009), to have more American College of Rheumatology classification criteria for SLE (P = 0.05), and to be receiving concurrent prednisone treatment (P = 0.04). Interestingly, European American patients were more likely to be low responders than were African American patients (P = 0.03). Following vaccination, low responders were more likely to experience disease flares (P = 0.01) and to have increased titers of antinuclear antibodies (P = 0.04). Serum interferon-α activity at baseline was significantly higher in patients in whom a flare occurred after vaccination compared to a matched group of patients who did not experience a disease flare (P = 0.04). CONCLUSION Ancestral background, prednisone treatment, hematologic criteria, and evidence of increased likelihood of disease flares were associated with low antibody responses to influenza vaccination in SLE patients.
Collapse
Affiliation(s)
- Sherry R. Crowe
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - Joan T. Merrill
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - Evan S. Vista
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - Amy B. Dedeke
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - David M. Thompson
- University of Oklahoma Health Sciences Center, 1100 N. Lindsay, Oklahoma City, OK 73104
| | - Scott Stewart
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - Joel M. Guthridge
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
| | - Timothy B. Niewold
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, 5841 S Maryland Avenue, Chicago, IL 60637
| | - Beverly S. Franek
- Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, 5841 S Maryland Avenue, Chicago, IL 60637
| | - Gillian M. Air
- University of Oklahoma Health Sciences Center, 1100 N. Lindsay, Oklahoma City, OK 73104
| | - Linda F. Thompson
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
- University of Oklahoma Health Sciences Center, 1100 N. Lindsay, Oklahoma City, OK 73104
| | - Judith A. James
- Oklahoma Medical Research Foundation, 825 NE 13 Street, Oklahoma City, OK 73104
- University of Oklahoma Health Sciences Center, 1100 N. Lindsay, Oklahoma City, OK 73104
| |
Collapse
|
10
|
Langley WA, Bradley KC, Li ZN, Talekar GR, Galloway SE, Steinhauer DA. The effects of preexisting immunity to influenza on responses to influenza vectors in mice. Vaccine 2010; 28:6305-13. [PMID: 20656032 DOI: 10.1016/j.vaccine.2010.06.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 06/10/2010] [Accepted: 06/30/2010] [Indexed: 02/04/2023]
Abstract
The use of viral vectors as vaccine candidates has shown promise against a number of pathogens. However, preexisting immunity to these vectors is a concern that must be addressed when deciding which viruses are suitable for use. A number of properties, including the existence of antigenically distinct subtypes, make influenza viruses attractive candidates for use as viral vectors. Here, we evaluate the ability of influenza viral vectors containing inserts of foreign pathogens to elicit antibody and CD8(+) T cell responses against these foreign antigens in the presence of preexisting immunity to influenza virus in mice. Specifically, responses to an H3N1-based vector expressing a 90 amino acid polypeptide derived from the protective antigen (PA) of Bacillus anthracis or an H1N1-based vector containing a CD8(+) T cell epitope from the glycoprotein (GP) of lymphocytic choriomeningitis virus were evaluated following infections with either homosubtypic or heterosubtypic influenza viruses. We found that mice previously infected with influenza viruses, even those expressing HA and NA proteins of completely different subtypes, were severely compromised in their ability to mount an immune response against the inserted epitopes. This inhibition was demonstrated to be mediated by CD8(+) T cells, which recognize multiple strains of influenza viruses. These CD8(+) T cells were further shown to protect mice from a lethal challenge by a heterologous influenza subtype. The implication of these data for the use of influenza virus vectors and influenza vaccination in general are discussed.
Collapse
Affiliation(s)
- William A Langley
- Department of Microbiology and Immunology, Emory University Medical School, 1510 Clifton Road, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|
11
|
Induction of neutralizing antibody responses to anthrax protective antigen by using influenza virus vectors: implications for disparate immune system priming pathways. J Virol 2010; 84:8300-7. [PMID: 20504926 DOI: 10.1128/jvi.00183-10] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral vectors based on influenza virus, rabies virus (RV), and vaccinia virus (VV) were used to express large polypeptide segments derived from the Bacillus anthracis protective antigen (PA). For the infectious influenza virus vector and recombinant VV constructs, the receptor binding domain (RBD or domain 4) or the lethal and edema factor binding domain (LEF or domain 1') were engineered into functional chimeric hemagglutinin (HA) glycoproteins. In the case of the RV vector, the viral glycoprotein (G) was used as a carrier for RBD in an inactivated form of the vector. These constructs were examined by using multiple homologous and heterologous prime/boost immunization regimens in order to optimize the induction of alpha-PA antibody responses. Several immunization combinations were shown to induce high titers of antibody recognizing the anthrax RBD and LEF domains, as well as the full-length PA protein in mice. The heterologous prime/boost immunization regimens that involved an initial intranasal administration of a live influenza virus vector, followed by an intramuscular boost with either the killed RV vector or the VV vector, were particularly effective, inducing antigen-specific antibodies at levels severalfold higher than homologous or alternative heterologous protocols. Furthermore, sera from several groups of the immunized mice demonstrated neutralization activity in an in vitro anthrax toxin neutralization assay. In some cases, such toxin-neutralizing activity was notably high, indicating that the mechanisms by which immunity is primed by live influenza virus vectors may have beneficial properties.
Collapse
|
12
|
Furuya Y, Regner M, Lobigs M, Koskinen A, Müllbacher A, Alsharifi M. Effect of inactivation method on the cross-protective immunity induced by whole 'killed' influenza A viruses and commercial vaccine preparations. J Gen Virol 2010; 91:1450-60. [PMID: 20147516 DOI: 10.1099/vir.0.018168-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We have recently shown that intranasal (i.n.) administration of gamma-irradiated A/PR/8 [A/Puerto Rico/8/34 (H1N1)] protects mice against lethal avian influenza A/Vietnam/1203/2004 (H5N1) and other heterosubtypic influenza A infections. Here, we used gamma-irradiated, formalin- and UV-inactivated A/PC [A/Port Chalmers/1/73 (H3N2)] virus preparations and compared their ability to induce both homologous and heterosubtypic protective immunity. Our data show that, in contrast to i.n. vaccination with formalin- or UV-inactivated virus, or the present commercially available trivalent influenza vaccine, a single dose of gamma-ray-inactivated A/PC (gamma-A/PC) conferred significant protection in mice against both homologous and heterosubtypic virus challenges. A multiple immunization regime was required for formalin-inactivated virus preparations to induce protective immunity against a homotypic virus challenge, but did not induce influenza A strain cross-protective immunity. The highly immunogenic gamma-A/PC, but not formalin- or UV-inactivated A/PC, nor the currently available subvirion vaccine, elicited cytotoxic T-cell responses that are most likely responsible for the cross-protective and long-lasting immunity against highly lethal influenza A infections in mice. Finally, freeze-drying of gamma-A/PC did not affect the ability to induce cross-protective immunity.
Collapse
Affiliation(s)
- Yoichi Furuya
- Viral Immunology and Molecular Virology, The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | | | | | | |
Collapse
|
13
|
Kang SM, Compans RW. Host responses from innate to adaptive immunity after vaccination: molecular and cellular events. Mol Cells 2009; 27:5-14. [PMID: 19214429 PMCID: PMC6280669 DOI: 10.1007/s10059-009-0015-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Accepted: 12/24/2008] [Indexed: 12/13/2022] Open
Abstract
The availability of effective vaccines has had the most profound positive effect on improving the quality of public health by preventing infectious diseases. Despite many successful vaccines, there are still old and new emerging pathogens against which there is no vaccine available. A better understanding of how vaccines work for providing protection will help to improve current vaccines as well as to develop effective vaccines against pathogens for which we do not have a proper means to control. Recent studies have focused on innate immunity as the first line of host defense and its role in inducing adaptive immunity; such studies have been an intense area of research, which will reveal the immunological mechanisms how vaccines work for protection. Toll-like receptors (TLRs), a family of receptors for pathogen-associated molecular patterns on cells of the innate immune system, play a critical role in detecting and responding to microbial infections. Importantly, the innate immune system modulates the quantity and quality of longterm T and B cell memory and protective immune responses to pathogens. Limited studies suggest that vaccines which mimic natural infection and/or the structure of pathogens seem to be effective in inducing long-term protective immunity. A better understanding of the similarities and differences of the molecular and cellular events in host responses to vaccination and pathogen infection would enable the rationale for design of novel preventive measures against many challenging pathogens.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
14
|
Correlation of cellular immune responses with protection against culture-confirmed influenza virus in young children. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 15:1042-53. [PMID: 18448618 DOI: 10.1128/cvi.00397-07] [Citation(s) in RCA: 212] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The highly sensitive gamma interferon (IFN-gamma) enzyme-linked immunosorbent spot (ELISPOT) assay permits the investigation of the role of cell-mediated immunity (CMI) in the protection of young children against influenza. Preliminary studies of young children confirmed that the IFN-gamma ELISPOT assay was a more sensitive measure of influenza memory immune responses than serum antibody and that among seronegative children aged 6 to <36 months, an intranasal dose of 10(7) fluorescent focus units (FFU) of a live attenuated influenza virus vaccine (CAIV-T) elicited substantial CMI responses. A commercial inactivated influenza virus vaccine elicited CMI responses only in children with some previous exposure to related influenza viruses as determined by detectable antibody levels prevaccination. The role of CMI in actual protection against community-acquired, culture-confirmed clinical influenza by CAIV-T was investigated in a large randomized, double-blind, placebo-controlled dose-ranging efficacy trial with 2,172 children aged 6 to <36 months in the Philippines and Thailand. The estimated protection curve indicated that the majority of infants and young children with >or=100 spot-forming cells/10(6) peripheral blood mononuclear cells were protected against clinical influenza, establishing a possible target level of CMI for future influenza vaccine development. The ELISPOT assay for IFN-gamma is a sensitive and reproducible measure of CMI and memory immune responses and contributes to establishing requirements for the future development of vaccines against influenza, especially those used for children.
Collapse
|
15
|
Sawai T, Itoh Y, Ozaki H, Isoda N, Okamoto K, Kashima Y, Kawaoka Y, Takeuchi Y, Kida H, Ogasawara K. Induction of cytotoxic T-lymphocyte and antibody responses against highly pathogenic avian influenza virus infection in mice by inoculation of apathogenic H5N1 influenza virus particles inactivated with formalin. Immunology 2008; 124:155-65. [PMID: 18205793 DOI: 10.1111/j.1365-2567.2007.02745.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We investigated whether a vaccine derived from an apathogenic reassortant type A H5N1 influenza strain could induce immune responses in vivo that mediated protection from highly pathogenic avian influenza virus infection in mice. After two subcutaneous immunizations with formalin-inactivated H5N1 whole virus particles (whole particle vaccine), significant killing specific for cells presenting a nucleoprotein peptide from the vaccine strain of the virus was observed. Similar vaccination with viruses treated with ether and formalin, which are commonly used for humans as ether-split vaccines, induced little or no cytotoxic T-cell response. Furthermore, whole particle vaccines of the apathogenic H5N1 strain were more effective than ether-split vaccines at inducing antibody production able to neutralize a highly pathogenic H5N1 strain. Finally, whole particle vaccines of H5N1 protected mice against infection by an H5N1 highly pathogenic avian influenza virus more effectively than did ether-split vaccines. These results suggest that formalin-inactivated virus particles of apathogenic strains are effective for induction of both cytotoxic T-lymphocyte and antibody responses against highly pathogenic avian influenza viruses in vivo, resulting in protection from infection by a highly pathogenic H5N1 virus.
Collapse
Affiliation(s)
- Toshihiro Sawai
- Department of Pathology, Shiga University of Medical Sciences, Setatsukinowa, Otsu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kang SM, Guo L, Yao Q, Skountzou I, Compans RW. Intranasal immunization with inactivated influenza virus enhances immune responses to coadministered simian-human immunodeficiency virus-like particle antigens. J Virol 2004; 78:9624-32. [PMID: 15331695 PMCID: PMC514968 DOI: 10.1128/jvi.78.18.9624-9632.2004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Intranasal immunization with inactivated influenza virus vaccine can provide protective immunity, whereas many other antigens are less effective when used for mucosal immunization. To determine whether influenza virus could enhance immune responses to an antigen coadministered to a mucosal surface, we studied the intranasal immunization of mice with a mixture of simian-human immunodeficiency virus (SHIV) virus-like particles (VLPs) and inactivated influenza virus. Compared to mice immunized with SHIV VLPs alone, mice coimmunized with SHIV VLPs and inactivated influenza virus showed significant increases in serum immunoglobulin G (IgG) and mucosal IgA antibodies specific to the human immunodeficiency virus envelope protein, neutralizing activities, numbers of gamma interferon- and interleukin 4-secreting lymphocytes, and cytotoxic-T-lymphocyte activities. The levels of enhancement of immune response by coimmunization with inactivated influenza virus were equivalent to those induced by inclusion of immunostimulatory CpG oligodeoxynucleotides (CpG DNA). We also observed that SHIV VLPs bind to influenza virus virions, forming mixed aggregates. These results indicate that inactivated influenza virus can play a role as a mucosal adjuvant to coadministered antigens.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Department of Microbiology and Immunology, Emory University School of Medicine, 1510 Clifton Rd., Atlanta, GA 30322, USA
| | | | | | | | | |
Collapse
|
17
|
Bossios A, Xatzipsalti M, Manoussakis E, Psarros F, Saxoni-Papageorgiou P, Papadopoulos NG. Expression of costimulatory molecules in peripheral blood mononuclear cells of atopic asthmatic children during virus-induced asthma exacerbations. Int Arch Allergy Immunol 2004; 134:223-6. [PMID: 15178891 DOI: 10.1159/000078769] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2003] [Accepted: 03/22/2004] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Respiratory viruses are the most frequent triggers of acute asthma exacerbations. Herein we investigate costimulatory molecule expression on peripheral blood mononuclear cells (PBMC) during such exacerbations. METHODS Eleven children with atopic asthma were followed prospectively and respiratory symptoms were recorded on diary cards. A blood sample and nasopharyngeal wash (NPW) were obtained at baseline and subsequently during an exacerbation. PBMC were immunophenotyped using flow cytometry. NPW samples were examined for the presence of respiratory viruses by RT-PCR. RESULTS A virus was detected in 73% of exacerbations and none at baseline. A drop of NK cells and a marginal increase of monocytes were the only changes of cell count during the exacerbation. A significant downregulation of B7-2 on NK cells and of B7-1 on monocytes was also observed during exacerbations. CONCLUSIONS The above observations are in contrast to in vitro findings showing an upregulation of costimulatory molecules after exposure of blood cells to viruses or allergens. It is possible that activated immune cells leave the blood stream to migrate to the inflammation site during acute asthma exacerbations.
Collapse
Affiliation(s)
- A Bossios
- Research Laboratory, Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens, Greece
| | | | | | | | | | | |
Collapse
|
18
|
Wareing MD, Tannock GA. Route of administration is the prime determinant of IgA and IgG2a responses in the respiratory tract of mice to the cold-adapted live attenuated influenza A donor strain A/Leningrad/134/17/57. Vaccine 2003; 21:3097-100. [PMID: 12804835 DOI: 10.1016/s0264-410x(03)00262-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Serum antibody and antibody secretory cell (ASC) responses to the cold-adapted (CA) live attenuated influenza A donor strain A/Leningrad/134/17/57 in BALB/c mice were determined in the lungs and mediastinal lymph nodes after administration by the intranasal, subcutaneous and intramuscular routes. Both types of response were greatest when an inoculum consisting of 10(6.5) 50% egg infectious doses (EID(50)) was administered twice intranasally at an interval of 3 weeks. Serum responses by the intramuscular route were much higher than by the subcutaneous route but, at doses of 10(6.5-7.5) EID(50), were still lower than that obtained with two doses of an intranasal inoculum of 10(6.5) EID(50). Virus-specific ASC responses for IgA and IgG2a were obtained in the lungs and mediastinal lymph nodes of mice inoculated with 10(6.5) EID(50) by the intranasal route. However, ASC responses after inoculation by either the subcutaneous or intramuscular routes were barely detectable, even at doses as high as 10(7.5) EID(50). These results confirm that intranasal administration of live vaccines induces far higher virus-specific IgA and IgG2a responses in the respiratory tract of mice than can be achieved by parenteral administration and that serum antibody levels induced by parenteral vaccination are unrelated to the respiratory ASC response.
Collapse
Affiliation(s)
- M D Wareing
- Department of Biotechnology and Environmental Biology, RMIT University, P.O. Box 71, Vic. 3083, Bundoora, Australia
| | | |
Collapse
|
19
|
Becker S, Soukup J. Coarse(PM(2.5-10)), fine(PM(2.5)), and ultrafine air pollution particles induce/increase immune costimulatory receptors on human blood-derived monocytes but not on alveolar macrophages. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2003; 66:847-859. [PMID: 12746131 DOI: 10.1080/15287390306381] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Diesel particles have been shown to possess adjuvant activity and influence the development of allergic sensitization. Also, more heterogeneous mixtures of pollution particles have been shown to affect host defenses and development of immunity in animal models. In the present study it was determined whether freshly collected particulate matter (PM(10)) in the size ranges 2.5-10 micro m (PM(2.5-10), coarse), 0.1-2.5 micro m (PM(2.5), fine), and </=0.1 micro m (ultrafine) in diameter affected the development of antigen presenting cells by evaluating the expression of surface receptors involved in T-cell interaction on both human alveolar macrophages (AM) and blood-derived monocytes (Mo). A Mo-AM coculture was exposed to 50 micro g/ml of particles and expression of HLA-DR, CD40, CD80, and CD86 on each cell type was assessed by flow cytometry. Mo upregulated the expression of all four receptors in response to each of the particle fractions, while expression was unaffected in AM. The cells were also exposed to two model air pollution particles, diesel dust and volcanic ash, neither of which affected receptor expression. Furthermore, Mo and AM were separately exposed to the three PM size fractions and supernatants assessed for the T-helper (CD4(+)) lymphocyte chemoattractant interleukin-16 (IL-16). AM, but not Mo, produced IL-16, and this chemoattractant was released only in response to PM(2.5-10). These data suggest that a wide size range of pollution particles contain materials that may promote antigen presentation by Mo, while the capability to specifically recruit CD4(+) lymphocytes is contained in AM stimulated with the coarse PM fraction.
Collapse
Affiliation(s)
- Susanne Becker
- U.S. Environmental Protection Agency, National Health and Environmental Effects Research Laboratory, Research Triangle Park, North Carolina, USA.
| | | |
Collapse
|
20
|
Ferko B, Stasakova J, Sereinig S, Romanova J, Katinger D, Niebler B, Katinger H, Egorov A. Hyperattenuated recombinant influenza A virus nonstructural-protein-encoding vectors induce human immunodeficiency virus type 1 Nef-specific systemic and mucosal immune responses in mice. J Virol 2001; 75:8899-908. [PMID: 11533153 PMCID: PMC114458 DOI: 10.1128/jvi.75.19.8899-8908.2001] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have generated recombinant influenza A viruses belonging to the H1N1 and H3N2 virus subtypes containing an insertion of the 137 C-terminal amino acid residues of the human immunodeficiency virus type 1 (HIV-1) Nef protein into the influenza A virus nonstructural-protein (NS1) reading frame. These viral vectors were found to be genetically stable and capable of growing efficiently in embryonated chicken eggs and tissue culture cells but did not replicate in the murine respiratory tract. Despite the hyperattenuated phenotype of influenza/NS-Nef viruses, a Nef and influenza virus (nucleoprotein)-specific CD8(+)-T-cell response was detected in spleens and the lymph nodes draining the respiratory tract after a single intranasal immunization of mice. Compared to the primary response, a marked enhancement of the CD8(+)-T-cell response was detected in the systemic and mucosal compartments, including mouse urogenital tracts, if mice were primed with the H1N1 subtype vector and subsequently boosted with the H3N2 subtype vector. In addition, Nef-specific serum IgG was detected in mice which were immunized twice with the recombinant H1N1 and then boosted with the recombinant H3N2 subtype virus. These findings may contribute to the development of alternative immunization strategies utilizing hyperattenuated live recombinant influenza virus vectors to prevent or control infectious diseases, e.g., HIV-1 infection.
Collapse
Affiliation(s)
- B Ferko
- Institut für Angewandte Mikrobiologie, Universität für Bodenkultur, A-1190 Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
21
|
von Hoegen P. Synthetic biomimetic supra molecular Biovector (SMBV) particles for nasal vaccine delivery. Adv Drug Deliv Rev 2001; 51:113-25. [PMID: 11516783 DOI: 10.1016/s0169-409x(01)00175-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
For the optimal delivery of antigens to mucosal tissues, especially as nasal sprays, protein antigen alone is often not sufficient. A clear need for nasal delivery systems has therefore evolved, especially for Influenza A vaccines. Such technologies will be even more essential for new modern vaccines based on recombinant antigens. Here we describe synthetic biomimetic supra molecular Biovector (SMBV) which have proven in preclinical and clinical evaluation to be suitable candidates for the delivery of nasal vaccines. They also demonstrate the potential to work with multiple antigens and furthermore allow combination with adjuvants. These Biovectors can associate with internal or lipid layer membrane proteins and peptides due to their charged polysaccharide core. The mimicry with viruses is also provided through their size of 60-80 nm, which allows sterilization by filtration. This makes them an ideal tool for the development of modern nasal vaccines, as they have shown to be able to induce the desired types of humoral immunity (serosal and mucosal immunity, IgA and IgG antibodies) as well as cellular immunity (CD4 and CD8 responses).
Collapse
Affiliation(s)
- P von Hoegen
- Biovector Therapeutics S.A., Chemin du Chêne Vert, BP 169, 31676 Cedex, Labege, France.
| |
Collapse
|