1
|
Rader NA, Lee KS, Loes AN, Miller-Stump OA, Cooper M, Wong TY, Boehm DT, Barbier M, Bevere JR, Heath Damron F. Influenza virus strains expressing SARS-CoV-2 receptor binding domain protein confer immunity in K18-hACE2 mice. Vaccine X 2024; 20:100543. [PMID: 39221180 PMCID: PMC11364132 DOI: 10.1016/j.jvacx.2024.100543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease (COVID-19), rapidly spread across the globe in 2019. With the emergence of the Omicron variant, COVID-19 shifted into an endemic phase. Given the anticipated rise in cases during the fall and winter seasons, the strategy of implementing seasonal booster vaccines for COVID-19 is becoming increasingly valuable to protect public health. This practice already exists for seasonal influenza vaccines to combat annual influenza seasons. Our goal was to investigate an easily modifiable vaccine platform for seasonal use against SARS-CoV-2. In this study, we evaluated the genetically modified influenza virus ΔNA(RBD) as an intranasal vaccine candidate for COVID-19. This modified virus was engineered to replace the coding sequence for the neuraminidase (NA) protein with a membrane-anchored form of the receptor binding domain (RBD) protein of SARS-CoV-2. We designed experiments to assess the protection of ΔNA(RBD) in K18-hACE2 mice using lethal (Delta) and non-lethal (Omicron) challenge models. Controls of COVID-19 mRNA vaccine and our lab's previously described intranasal virus like particle vaccine were used as comparisons. Immunization with ΔNA(RBD) expressing ancestral RBD elicited high anti-RBD IgG levels in the serum of mice, high anti-RBD IgA in lung tissue, and improved survival after Delta variant challenge. Modifying ΔNA(RBD) to express Omicron variant RBD shifted variant-specific antibody responses and limited viral burden in the lungs of mice after Omicron variant challenge. Overall, this data suggests that ΔNA(RBD) could be an effective intranasal vaccine platform that generates mucosal and systemic immunity towards SARS-CoV-2.
Collapse
Affiliation(s)
- Nathaniel A. Rader
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Katherine S. Lee
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - 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
| | - Olivia A. Miller-Stump
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Melissa Cooper
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Ting Y. Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Dylan T. Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Justin R. Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, USA
- Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, USA
| |
Collapse
|
2
|
Simpson J, Kasson PM. Structural prediction of chimeric immunogens to elicit targeted antibodies against betacoronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.31.526494. [PMID: 36778336 PMCID: PMC9915606 DOI: 10.1101/2023.01.31.526494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Betacoronaviruses pose an ongoing pandemic threat. Antigenic evolution of the SARS-CoV-2 virus has shown that much of the spontaneous antibody response is narrowly focused rather than broadly neutralizing against even SARS-CoV-2 variants, let alone future threats. One way to overcome this is by focusing the antibody response against better-conserved regions of the viral spike protein. Here, we present a design approach to predict stable chimeras between SARS-CoV-2 and other coronaviruses, creating synthetic spike proteins that display a desired conserved region and vary other regions. We leverage AlphaFold to predict chimeric structures and create a new metric for scoring chimera stability based on AlphaFold outputs. We evaluated 114 candidate spike chimeras using this approach. Top chimeras were further evaluated using molecular dynamics simulation as an intermediate validation technique, showing good stability compared to low-scoring controls. Experimental testing of five predicted-stable and two predicted-unstable chimeras confirmed 5/7 predictions, with one intermediate result. This demonstrates the feasibility of the underlying approach, which can be used to design custom immunogens to focus the immune response against a desired viral glycoprotein epitope.
Collapse
Affiliation(s)
- Jamel Simpson
- Program in Biophysics and Department of Biomedical Engineering, Box 800886, Charlottesville VA 22908
| | - Peter M. Kasson
- Program in Biophysics and Department of Biomedical Engineering, Box 800886, Charlottesville VA 22908
- Departments of Chemistry & Biochemistry and Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
- Department of Cell and Molecular Biology, Uppsala University, Box 256, Uppsala, Sweden
| |
Collapse
|
3
|
Wang Y, Wei X, Liu Y, Li S, Pan W, Dai J, Yang Z. Towards broad-spectrum protection: the development and challenges of combined respiratory virus vaccines. Front Cell Infect Microbiol 2024; 14:1412478. [PMID: 38903942 PMCID: PMC11188343 DOI: 10.3389/fcimb.2024.1412478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
In the post-COVID-19 era, the co-circulation of respiratory viruses, including influenza, SARS-CoV-2, and respiratory syncytial virus (RSV), continues to have significant health impacts and presents ongoing public health challenges. Vaccination remains the most effective measure for preventing viral infections. To address the concurrent circulation of these respiratory viruses, extensive efforts have been dedicated to the development of combined vaccines. These vaccines utilize a range of platforms, including mRNA-based vaccines, viral vector vaccines, and subunit vaccines, providing opportunities in addressing multiple pathogens at once. This review delves into the major advancements in the field of combined vaccine research, underscoring the strategic use of various platforms to tackle the simultaneous circulation of respiratory viruses effectively.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xiaotong Wei
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yang Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Customs Technology Center, Guangzhou, China
| | - Shengfeng Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiqi Pan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Respiratory Disease AI Laboratory on Epidemic and Medical Big Data Instrument Applications, Faculty of Innovation Engineering, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jun Dai
- Guangzhou National Laboratory, Guangzhou, China
- Guangzhou Customs Technology Center, Guangzhou, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
- Respiratory Disease AI Laboratory on Epidemic and Medical Big Data Instrument Applications, Faculty of Innovation Engineering, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
4
|
Moser MJ, Hill-Batorski L, Bowen RA, Matejka SM, Marshall D, Kawaoka Y, Neumann G, Bilsel P. Intranasal Single-Replication Influenza Vector Induces Cross-Reactive Serum and Mucosal Antibodies against SARS-CoV-2 Variants. Vaccines (Basel) 2023; 11:1063. [PMID: 37376452 DOI: 10.3390/vaccines11061063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Current SARS-CoV-2 vaccines provide protection for COVID-19-associated hospitalization and death, but remain inefficient at inhibiting initial infection and transmission. Despite updated booster formulations, breakthrough infections and reinfections from emerging SARS-CoV-2 variants are common. Intranasal vaccination to elicit mucosal immunity at the site of infection can improve the performance of respiratory virus vaccines. We developed SARS-CoV-2 M2SR, a dual SARS-CoV-2 and influenza vaccine candidate, employing our live intranasal M2-deficient single replication (M2SR) influenza vector expressing the receptor binding domain (RBD) of the SARS-CoV-2 Spike protein of the prototype strain, first reported in January 2020. The intranasal vaccination of mice with this dual vaccine elicits both high serum IgG and mucosal IgA titers to RBD. Sera from inoculated mice show that vaccinated mice develop neutralizing SARS-CoV-2 antibody titers against the prototype and Delta virus strains, which are considered to be sufficient to protect against viral infection. Moreover, SARS-CoV-2 M2SR elicited cross-reactive serum and mucosal antibodies to the Omicron BA.4/BA.5 variant. The SARS-CoV-2 M2SR vaccine also maintained strong immune responses to influenza A with high titers of anti H3 serum IgG and hemagglutination inhibition (HAI) antibody titers corresponding to those seen from the control M2SR vector alone. With a proven safety record and robust immunological profile in humans that includes mucosal immunity, the M2SR influenza viral vector expressing key SARS-CoV-2 antigens could provide more efficient protection against influenza and SARS-CoV-2 variants.
Collapse
Affiliation(s)
| | | | - Richard A Bowen
- Department of Biomedical Sciences, Colorado State University, 1601 Campus Delivery, Fort Collins, CO 80523, USA
| | | | | | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin, 2015 Linden Dr., Madison, WI 53706, USA
| | - Gabriele Neumann
- Department of Pathobiological Sciences, University of Wisconsin, 2015 Linden Dr., Madison, WI 53706, USA
| | - Pamuk Bilsel
- FluGen, Inc., 597 Science Drive, Madison, WI 53711, USA
| |
Collapse
|
5
|
Boosting BCG with recombinant influenza A virus tuberculosis vaccines increases pulmonary T cell responses but not protection against Mycobacterium tuberculosis infection. PLoS One 2021; 16:e0259829. [PMID: 34793507 PMCID: PMC8601556 DOI: 10.1371/journal.pone.0259829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 10/27/2021] [Indexed: 11/19/2022] Open
Abstract
The current Mycobacterium bovis BCG vaccine provides inconsistent protection against pulmonary infection with Mycobacterium tuberculosis. Immunity induced by subcutaneous immunization with BCG wanes and does not promote early recruitment of T cell to the lungs after M. tuberculosis infection. Delivery of Tuberculosis (TB) vaccines to the lungs may increase and prolong immunity at the primary site of M. tuberculosis infection. Pulmonary immunization with recombinant influenza A viruses (rIAVs) expressing an immune-dominant M. tuberculosis CD4+ T cell epitope (PR8-p25 and X31-p25) stimulates protective immunity against lung TB infection. Here, we investigated the potential use of rIAVs to improve the efficacy of BCG using simultaneous immunization (SIM) and prime-boost strategies. SIM with parenteral BCG and intranasal PR8-p25 resulted in equivalent protection to BCG alone against early, acute and chronic M. tuberculosis infection. Boosting BCG with rIAVs increased the frequency of IFN-γ-secreting specific T cells (p<0.001) and polyfunctional CD4+ T cells (p<0.05) in the lungs compared to the BCG alone, however, this did not result in a significant increase in protection against M. tuberculosis compared to BCG alone. Therefore, sequential pulmonary immunization with these rIAVs after BCG increased M. tuberculosis-specific memory T cell responses in the lung, but not protection against M. tuberculosis infection.
Collapse
|
6
|
Koonpaew S, Kaewborisuth C, Srisutthisamphan K, Wanitchang A, Thaweerattanasinp T, Saenboonrueng J, Poonsuk S, Jengarn J, Viriyakitkosol R, Kramyu J, Jongkaewwattana A. A Single-Cycle Influenza A Virus-Based SARS-CoV-2 Vaccine Elicits Potent Immune Responses in a Mouse Model. Vaccines (Basel) 2021; 9:vaccines9080850. [PMID: 34451975 PMCID: PMC8402467 DOI: 10.3390/vaccines9080850] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/24/2021] [Accepted: 07/30/2021] [Indexed: 01/14/2023] Open
Abstract
The use of virus-vectored platforms has increasingly gained attention in vaccine development as a means for delivering antigenic genes of interest into target hosts. Here, we describe a single-cycle influenza virus-based SARS-CoV-2 vaccine designated as scPR8-RBD-M2. The vaccine utilizes the chimeric gene encoding 2A peptide-based bicistronic protein cassette of the SARS-CoV-2 receptor-binding domain (RBD) and influenza matrix 2 (M2) protein. The C-terminus of the RBD was designed to link with the cytoplasmic domain of the influenza virus hemagglutinin (HA) to anchor the RBD on the surface of producing cells and virus envelope. The chimeric RBD-M2 gene was incorporated in place of the HA open-reading frame (ORF) between the 3′ and 5′ UTR of HA gene for the virus rescue in MDCK cells stably expressing HA. The virus was also constructed with the disrupted M2 ORF in segment seven to ensure that M2 from the RBD-M2 was utilized. The chimeric gene was intact and strongly expressed in infected cells upon several passages, suggesting that the antigen was stably maintained in the vaccine candidate. Mice inoculated with scPR8-RBD-M2 via two alternative prime-boost regimens (intranasal-intranasal or intranasal-intramuscular routes) elicited robust mucosal and systemic humoral immune responses and cell-mediated immunity. Notably, we demonstrated that immunized mouse sera exhibited neutralizing activity against pseudotyped viruses bearing SARS-CoV-2 spikes from various variants, albeit with varying potency. Our study warrants further development of a replication-deficient influenza virus as a promising SARS-CoV-2 vaccine candidate.
Collapse
Affiliation(s)
- Surapong Koonpaew
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Kanjana Srisutthisamphan
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Asawin Wanitchang
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Theeradej Thaweerattanasinp
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Janya Saenboonrueng
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Sukontip Poonsuk
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Juggragarn Jengarn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | | | - Jarin Kramyu
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani 12120, Thailand; (S.K.); (C.K.); (K.S.); (A.W.); (T.T.); (J.S.); (S.P.); (J.J.); (J.K.)
- Correspondence:
| |
Collapse
|
7
|
Haque S, Swami P, Khan A. S. Typhi derived vaccines and a proposal for outer membrane vesicles (OMVs) as potential vaccine for typhoid fever. Microb Pathog 2021; 158:105082. [PMID: 34265371 DOI: 10.1016/j.micpath.2021.105082] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022]
Abstract
Typhoid fever is a serious systemic infection caused by Salmonella Typhi (S. Typhi), spread by the feco-oral route and closely associated with poor food hygiene and inadequate sanitation. Nearly 93% of S. Typhi strains have acquired antibiotic resistance against most antibiotics. Vaccination is the only promising way to prevent typhoid fever. This review covers the nature and composition of S. Typhi, pathogenecity and mode of infection, epidemiology, and nature of drug resistance. Several components (Vi-polysaccharides, O-antigens, flagellar antigens, full length OMPs, and short peptides from OMPs) of S. Typhi have been utilized for vaccine design for protection against typhoid fever. Vaccine delivery systems also contribute to efficacy of the vaccines. In this study, we propose to develop S. Typhi derived OMVs as vaccine for protection against typhoid fevers.
Collapse
Affiliation(s)
- Shabirul Haque
- Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Pooja Swami
- Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| | - Azhar Khan
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal, Pradesh, India.
| |
Collapse
|
8
|
Mucosal Priming with a Recombinant Influenza A Virus-Vectored Vaccine Elicits T-Cell and Antibody Responses to HIV-1 in Mice. J Virol 2021; 95:JVI.00059-21. [PMID: 33789991 DOI: 10.1128/jvi.00059-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Recombinant influenza A viral (IAV) vectors are potential to stimulate systemic and mucosal immunity, but the packaging capacity is limited and only one or a few epitopes can be carried. Here, we report the generation of a replication-competent IAV vector that carries a full-length HIV-1 p24 gene linked to the 5'-terminal coding region of the neuraminidase segment via a protease cleavage sequence (IAV-p24). IAV-p24 was successfully rescued and stably propagated, and P24 protein was efficiently expressed in infected mammalian cells. In BALB/c mice, IAV-p24 showed attenuated pathogenicity compared to that of the parental A/PR/8/34 (H1N1) virus. An intranasal inoculation with IAV-p24 elicited moderate HIV-specific cell-mediated immune (CMI) responses in the airway and vaginal tracts and in the spleen, and an intranasal boost with a replication-incompetent adenovirus type 2 vector expressing the HIV-1 gag gene (Ad2-gag) greatly improved these responses. Importantly, compared to an Ad2-gag prime plus IAV-p24 boost regimen, the IAV-p24 prime plus Ad2-gag boost regimen had a greater efficacy in eliciting HIV-specific CMI responses. P24-specific CD8+ T cells and antibodies were robustly provoked both systemically and in mucosal sites and showed long-term durability, revealing that IAV-p24 may be used as a mucosa-targeted priming vaccine. Our results illustrate that IAV-p24 is able to prime systemic and mucosal immunity against HIV-1 and warrants further evaluation in nonhuman primates.IMPORTANCE An effective HIV-1 vaccine remains elusive despite nearly 40 years of research. CD8+ T cells and protective antibodies may both be desirable for preventing HIV-1 infection in susceptible mucosal sites. Recombinant influenza A virus (IAV) vector has the potential to stimulate these immune responses, but the packaging capacity is extremely limited. Here, we describe a replication-competent IAV vector expressing the HIV-1 p24 gene (IAV-p24). Unlike most other IAV vectors that carried one or several antigenic epitopes, IAV-p24 stably expressed the full-length P24 protein, which contains multiple epitopes and is highly conserved among all known HIV-1 sequences. Compared to the parental A/PR/8/34 (H1N1) virus, IAV-p24 showed an attenuated pathogenicity in BALB/c mice. When combined with an adenovirus vector expressing the HIV-1 gag gene, IAV-p24 was able to prime P24-specific systemic and mucosal immune responses. IAV-p24 as an alternative priming vaccine against HIV-1 warrants further evaluation in nonhuman primates.
Collapse
|
9
|
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: 19] [Impact Index Per Article: 4.8] [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
|
10
|
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: 1.0] [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
|
11
|
Gerlach T, Elbahesh H, Saletti G, Rimmelzwaan GF. Recombinant influenza A viruses as vaccine vectors. Expert Rev Vaccines 2019; 18:379-392. [PMID: 30777467 DOI: 10.1080/14760584.2019.1582338] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Various viruses, including poxviruses, adenoviruses and vesicular stomatitis virus, have been considered as vaccine vectors for the delivery of antigens of interest in the development of vaccines against newly emerging pathogens. AREAS COVERED Here, we review results that have been obtained with influenza A viruses (IAV) as vaccine vectors. With the advent of reverse genetics technology, IAV-based recombinant vaccine candidates have been constructed that induce protective immunity to a variety of different pathogens of interest, including West Nile virus, Plasmodium falciparum and respiratory syncytial virus. The various cloning strategies to produce effective and attenuated, safe to use IAV-based viral vectors are discussed. EXPERT COMMENTARY It was concluded that IAV-based vector system has several advantages and holds promise for further development.
Collapse
Affiliation(s)
- Thomas Gerlach
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Husni Elbahesh
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Giulietta Saletti
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| | - Guus F Rimmelzwaan
- a Research Center for Emerging Infections and Zoonoses (RIZ) , University of Veterinary Medicine Hannover (TiHo) , Hannover , Germany
| |
Collapse
|
12
|
Muflihah H, Flórido M, Lin L, Xia Y, Triccas J, Stambas J, Britton W. Sequential pulmonary immunization with heterologous recombinant influenza A virus tuberculosis vaccines protects against murine M. tuberculosis infection. Vaccine 2018; 36:2462-2470. [DOI: 10.1016/j.vaccine.2018.03.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/22/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
|
13
|
Engineering Recombinant Reoviruses To Display gp41 Membrane-Proximal External-Region Epitopes from HIV-1. mSphere 2016; 1:mSphere00086-16. [PMID: 27303748 PMCID: PMC4888892 DOI: 10.1128/msphere.00086-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/25/2016] [Indexed: 12/30/2022] Open
Abstract
Vaccines to protect against HIV-1, the causative agent of AIDS, are not approved for use. Antibodies that neutralize genetically diverse strains of HIV-1 bind to discrete regions of the envelope glycoproteins, including the gp41 MPER. We engineered recombinant reoviruses that displayed MPER epitopes in attachment protein σ1 (REO-MPER vectors). The REO-MPER vectors replicated with wild-type efficiency, were genetically stable, and retained native antigenicity. However, we did not detect HIV-1-specific immune responses following inoculation of the REO-MPER vectors into small animals. This work provides proof of principle for engineering reovirus to express antigenic epitopes and illustrates the difficulty in eliciting MPER-specific immune responses. The gp41 membrane-proximal external region (MPER) is a target for broadly neutralizing antibody responses against human immunodeficiency virus type 1 (HIV-1). However, replication-defective virus vaccines currently under evaluation in clinical trials do not efficiently elicit MPER-specific antibodies. Structural modeling suggests that the MPER forms an α-helical coiled coil that is required for function and immunogenicity. To maintain the native MPER conformation, we used reverse genetics to engineer replication-competent reovirus vectors that displayed MPER sequences in the α-helical coiled-coil tail domain of viral attachment protein σ1. Sequences in reovirus strain type 1 Lang (T1L) σ1 were exchanged with sequences encoding HIV-1 strain Ba-L MPER epitope 2F5 or the entire MPER. Individual 2F5 or MPER substitutions were introduced at virion-proximal or virion-distal sites in the σ1 tail. Recombinant reoviruses containing heterologous HIV-1 sequences were viable and produced progeny yields comparable to those with wild-type virus. HIV-1 sequences were retained following 10 serial passages in cell culture, indicating that the substitutions were genetically stable. Recombinant viruses engineered to display the 2F5 epitope or full-length MPER in σ1 were recognized by purified 2F5 antibody. Inoculation of mice with 2F5-containing vectors or rabbits with 2F5- or MPER-containing vectors elicited anti-reovirus antibodies, but HIV-1-specific antibodies were not detected. Together, these findings indicate that heterologous sequences that form α-helices can functionally replace native sequences in the α-helical tail domain of reovirus attachment protein σ1. However, although these vectors retain native antigenicity, they were not immunogenic, illustrating the difficulty of experimentally inducing immune responses to this essential region of HIV-1. IMPORTANCE Vaccines to protect against HIV-1, the causative agent of AIDS, are not approved for use. Antibodies that neutralize genetically diverse strains of HIV-1 bind to discrete regions of the envelope glycoproteins, including the gp41 MPER. We engineered recombinant reoviruses that displayed MPER epitopes in attachment protein σ1 (REO-MPER vectors). The REO-MPER vectors replicated with wild-type efficiency, were genetically stable, and retained native antigenicity. However, we did not detect HIV-1-specific immune responses following inoculation of the REO-MPER vectors into small animals. This work provides proof of principle for engineering reovirus to express antigenic epitopes and illustrates the difficulty in eliciting MPER-specific immune responses.
Collapse
|
14
|
Tan HX, Gilbertson BP, Jegaskanda S, Alcantara S, Amarasena T, Stambas J, McAuley JL, Kent SJ, De Rose R. Recombinant influenza virus expressing HIV-1 p24 capsid protein induces mucosal HIV-specific CD8 T-cell responses. Vaccine 2016; 34:1172-9. [PMID: 26826545 DOI: 10.1016/j.vaccine.2016.01.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/24/2015] [Accepted: 01/17/2016] [Indexed: 10/22/2022]
Abstract
Influenza viruses are promising mucosal vaccine vectors for HIV but their use has been limited by difficulties in engineering the expression of large amounts of foreign protein. We developed recombinant influenza viruses incorporating the HIV-1 p24 gag capsid into the NS-segment of PR8 (H1N1) and X31 (H3N2) influenza viruses with the use of multiple 2A ribosomal skip sequences. Despite the insertion of a sizable HIV-1 gene into the influenza genome, recombinant viruses were readily rescued to high titers. Intracellular expression of p24 capsid was confirmed by in vitro infection assays. The recombinant influenza viruses were subsequently tested as mucosal vaccines in BALB/c mice. Recombinant viruses were attenuated and safe in immunized mice. Systemic and mucosal HIV-specific CD8 T-cell responses were elicited in mice that were immunized via intranasal route with a prime-boost regimen. Isolated HIV-specific CD8 T-cells displayed polyfunctional cytokine and degranulation profiles. Mice boosted via intravaginal route induced recall responses from the distal lung mucosa and developed heightened HIV-specific CD8 T-cell responses in the vaginal mucosa. These findings demonstrate the potential utility of recombinant influenza viruses as vaccines for mucosal immunity against HIV-1 infection.
Collapse
Affiliation(s)
- Hyon-Xhi Tan
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| | - Brad P Gilbertson
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| | - Sinthujan Jegaskanda
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia; Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, MD 20892, United States
| | - Sheilajen Alcantara
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| | - Thakshila Amarasena
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| | - John Stambas
- School of Medicine, Deakin University, Geelong, Victoria, Australia; CSIRO Animal Health Laboratories, Geelong, Victoria, Australia
| | - Julie L McAuley
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia; Melbourne Sexual Health Centre, Alfred Hospital, Monash University Central Clinical School, Victoria, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Melbourne, Victoria, Australia.
| | - Robert De Rose
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3010, Australia
| |
Collapse
|
15
|
Banerjee S, Shi H, Habte HH, Qin Y, Cho MW. Modulating immunogenic properties of HIV-1 gp41 membrane-proximal external region by destabilizing six-helix bundle structure. Virology 2016; 490:17-26. [PMID: 26803471 DOI: 10.1016/j.virol.2016.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/05/2016] [Accepted: 01/09/2016] [Indexed: 01/31/2023]
Abstract
The C-terminal alpha-helix of gp41 membrane-proximal external region (MPER; (671)NWFDITNWLWYIK(683)) encompassing 4E10/10E8 epitopes is an attractive target for HIV-1 vaccine development. We previously reported that gp41-HR1-54Q, a trimeric protein comprised of the MPER in the context of a stable six-helix bundle (6HB), induced strong immune responses against the helix, but antibodies were directed primarily against the non-neutralizing face of the helix. To better target 4E10/10E8 epitopes, we generated four putative fusion intermediates by introducing double point mutations or deletions in the heptad repeat region 1 (HR1) that destabilize 6HB in varying degrees. One variant, HR1-∆10-54K, elicited antibodies in rabbits that targeted W672, I675 and L679, which are critical for 4E10/10E8 recognition. Overall, the results demonstrated that altering structural parameters of 6HB can influence immunogenic properties of the MPER and antibody targeting. Further exploration of this strategy could allow development of immunogens that could lead to induction of 4E10/10E8-like antibodies.
Collapse
Affiliation(s)
- Saikat Banerjee
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Heliang Shi
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Habtom H Habte
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Yali Qin
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States
| | - Michael W Cho
- Department of Biomedical Sciences, College of Veterinary Medicine; and Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, Ames, IA 50011, United States.
| |
Collapse
|
16
|
Habte HH, Banerjee S, Shi H, Qin Y, Cho MW. Immunogenic properties of a trimeric gp41-based immunogen containing an exposed membrane-proximal external region. Virology 2015; 486:187-97. [PMID: 26454663 DOI: 10.1016/j.virol.2015.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/09/2015] [Accepted: 09/22/2015] [Indexed: 01/15/2023]
Abstract
The membrane-proximal external region (MPER) of HIV-1 gp41 is an attractive target for vaccine development. Thus, better understanding of its immunogenic properties in various structural contexts is important. We previously described the crystal structure of a trimeric protein complex named gp41-HR1-54Q, which consists of the heptad repeat regions 1 and 2 and the MPER. The protein was efficiently recognized by broadly neutralizing antibodies. Here, we describe its immunogenic properties in rabbits. The protein was highly immunogenic, especially the C-terminal end of the MPER containing 4E10 and 10E8 epitopes ((671)NWFDITNWLWYIK(683)). Although antibodies exhibited strong competition activity against 4E10 and 10E8, neutralizing activity was not detected. Detailed mapping analyses indicated that amino acid residues critical for recognition resided on faces of the alpha helix that are either opposite of or perpendicular to the epitopes recognized by 4E10 and 10E8. These results provide critical information for designing the next generation of MPER-based immunogens.
Collapse
Affiliation(s)
- Habtom H Habte
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Saikat Banerjee
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Heliang Shi
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Yali Qin
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA
| | - Michael W Cho
- College of Veterinary Medicine, Department of Biomedical Sciences, Center for Advanced Host Defenses, Immunobiotics and Translational Comparative Medicine, Iowa State University, 1600 S 16th Street, Ames, IA 50011-1250, USA.
| |
Collapse
|
17
|
Zang Y, Du D, Li N, Su W, Liu X, Zhang Y, Nie J, Wang Y, Kong W, Jiang C. Eliciting neutralizing antibodies against the membrane proximal external region of HIV-1 Env by chimeric live attenuated influenza A virus vaccines. Vaccine 2015; 33:3859-64. [PMID: 26126669 DOI: 10.1016/j.vaccine.2015.06.072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/14/2015] [Accepted: 06/17/2015] [Indexed: 10/23/2022]
|
18
|
Abstract
The advent of reverse genetic approaches to manipulate the genomes of both positive (+) and negative (-) sense RNA viruses allowed researchers to harness these genomes for basic research. Manipulation of positive sense RNA virus genomes occurred first largely because infectious RNA could be transcribed directly from cDNA versions of the RNA genomes. Manipulation of negative strand RNA virus genomes rapidly followed as more sophisticated approaches to provide RNA-dependent RNA polymerase complexes coupled with negative-strand RNA templates were developed. These advances have driven an explosion of RNA virus vaccine vector development. That is, development of approaches to exploit the basic replication and expression strategies of RNA viruses to produce vaccine antigens that have been engineered into their genomes. This study has led to significant preclinical testing of many RNA virus vectors against a wide range of pathogens as well as cancer targets. Multiple RNA virus vectors have advanced through preclinical testing to human clinical evaluation. This review will focus on RNA virus vectors designed to express heterologous genes that are packaged into viral particles and have progressed to clinical testing.
Collapse
Affiliation(s)
- Mark A Mogler
- Harrisvaccines, Inc., 1102 Southern Hills Drive, Suite 101, Ames, IA 50010, USA
| | | |
Collapse
|
19
|
Lai RPJ, Hock M, Radzimanowski J, Tonks P, Hulsik DL, Effantin G, Seilly DJ, Dreja H, Kliche A, Wagner R, Barnett SW, Tumba N, Morris L, LaBranche CC, Montefiori DC, Seaman MS, Heeney JL, Weissenhorn W. A fusion intermediate gp41 immunogen elicits neutralizing antibodies to HIV-1. J Biol Chem 2014; 289:29912-26. [PMID: 25160627 PMCID: PMC4208001 DOI: 10.1074/jbc.m114.569566] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 08/01/2014] [Indexed: 11/06/2022] Open
Abstract
The membrane-proximal external region (MPER) of the human immunodeficiency virus, type 1 (HIV-1) envelope glycoprotein subunit gp41 is targeted by potent broadly neutralizing antibodies 2F5, 4E10, and 10E8. These antibodies recognize linear epitopes and have been suggested to target the fusion intermediate conformation of gp41 that bridges viral and cellular membranes. Anti-MPER antibodies exert different degrees of membrane interaction, which is considered to be the limiting factor for the generation of such antibodies by immunization. Here we characterize a fusion intermediate conformation of gp41 (gp41(int)-Cys) and show that it folds into an elongated ∼ 12-nm-long extended structure based on small angle x-ray scattering data. Gp41(int)-Cys was covalently linked to liposomes via its C-terminal cysteine and used as immunogen. The gp41(int)-Cys proteoliposomes were administered alone or in prime-boost regimen with trimeric envelope gp140(CA018) in guinea pigs and elicited high anti-gp41 IgG titers. The sera interacted with a peptide spanning the MPER region, demonstrated competition with broadly neutralizing antibodies 2F5 and 4E10, and exerted modest lipid binding, indicating the presence of MPER-specific antibodies. Although the neutralization potency generated solely by gp140(CA018) was higher than that induced by gp41(int)-Cys, the majority of animals immunized with gp41(int)-Cys proteoliposomes induced modest breadth and potency in neutralizing tier 1 pseudoviruses and replication-competent simian/human immunodeficiency viruses in the TZM-bl assay as well as responses against tier 2 HIV-1 in the A3R5 neutralization assay. Our data thus demonstrate that liposomal gp41 MPER formulation can induce neutralization activity, and the strategy serves to improve breadth and potency of such antibodies by improved vaccination protocols.
Collapse
Affiliation(s)
- Rachel P J Lai
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Miriam Hock
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Jens Radzimanowski
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Paul Tonks
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - David Lutje Hulsik
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - Gregory Effantin
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France
| | - David J Seilly
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Hanna Dreja
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Alexander Kliche
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Susan W Barnett
- Novartis Vaccines and Diagnostics Inc., Cambridge, Massachusetts 02139
| | - Nancy Tumba
- National Institute for Communicable Diseases, Centre for HIV and Sexually Transmitted Infections, 1 Modderfontein Road, Sandringham 2131, South Africa
| | - Lynn Morris
- National Institute for Communicable Diseases, Centre for HIV and Sexually Transmitted Infections, 1 Modderfontein Road, Sandringham 2131, South Africa
| | - Celia C LaBranche
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, and
| | - David C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, and
| | - Michael S Seaman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02115
| | - Jonathan L Heeney
- From the Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom,
| | - Winfried Weissenhorn
- Université Grenoble Alpes, Unit of Virus Host Cell Interactions (UVHCI), F-38000 Grenoble, France, CNRS, UVHCI, F-38000 Grenoble, France,
| |
Collapse
|
20
|
Ondondo BO. The influence of delivery vectors on HIV vaccine efficacy. Front Microbiol 2014; 5:439. [PMID: 25202303 PMCID: PMC4141443 DOI: 10.3389/fmicb.2014.00439] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/03/2014] [Indexed: 12/31/2022] Open
Abstract
Development of an effective HIV/AIDS vaccine remains a big challenge, largely due to the enormous HIV diversity which propels immune escape. Thus novel vaccine strategies are targeting multiple variants of conserved antibody and T cell epitopic regions which would incur a huge fitness cost to the virus in the event of mutational escape. Besides immunogen design, the delivery modality is critical for vaccine potency and efficacy, and should be carefully selected in order to not only maximize transgene expression, but to also enhance the immuno-stimulatory potential to activate innate and adaptive immune systems. To date, five HIV vaccine candidates have been evaluated for efficacy and protection from acquisition was only achieved in a small proportion of vaccinees in the RV144 study which used a canarypox vector for delivery. Conversely, in the STEP study (HVTN 502) where human adenovirus serotype 5 (Ad5) was used, strong immune responses were induced but vaccination was more associated with increased risk of HIV acquisition than protection in vaccinees with pre-existing Ad5 immunity. The possibility that pre-existing immunity to a highly promising delivery vector may alter the natural course of HIV to increase acquisition risk is quite worrisome and a huge setback for HIV vaccine development. Thus, HIV vaccine development efforts are now geared toward delivery platforms which attain superior immunogenicity while concurrently limiting potential catastrophic effects likely to arise from pre-existing immunity or vector-related immuno-modulation. However, it still remains unclear whether it is poor immunogenicity of HIV antigens or substandard immunological potency of the safer delivery vectors that has limited the success of HIV vaccines. This article discusses some of the promising delivery vectors to be harnessed for improved HIV vaccine efficacy.
Collapse
Affiliation(s)
- Beatrice O Ondondo
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford Oxford, UK
| |
Collapse
|
21
|
Chanzu N, Ondondo B. Induction of Potent and Long-Lived Antibody and Cellular Immune Responses in the Genitorectal Mucosa Could be the Critical Determinant of HIV Vaccine Efficacy. Front Immunol 2014; 5:202. [PMID: 24847327 PMCID: PMC4021115 DOI: 10.3389/fimmu.2014.00202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/23/2014] [Indexed: 01/28/2023] Open
Abstract
The field of HIV prevention has indeed progressed in leaps and bounds, but with major limitations of the current prevention and treatment options, the world remains desperate for an HIV vaccine. Sadly, this continues to be elusive, because more than 30 years since its discovery there is no licensed HIV vaccine. Research aiming to define immunological biomarkers to accurately predict vaccine efficacy have focused mainly on systemic immune responses, and as such, studies defining correlates of protection in the genitorectal mucosa, the primary target site for HIV entry and seeding are sparse. Clearly, difficulties in sampling and analysis of mucosal specimens, as well as their limited size have been a major deterrent in characterizing the type (mucosal antibodies, cytokines, chemokines, or CTL), threshold (magnitude, depth, and breadth) and viral inhibitory capacity of HIV-1-specific immune responses in the genitorectal mucosa, where they are needed to immediately block HIV acquisition and arrest subsequent virus dissemination. Nevertheless, a few studies document the existence of HIV-specific immune responses in the genitorectal mucosa of HIV-infected aviremic and viremic controllers, as well as in highly exposed persistently seronegative (HEPS) individuals with natural resistance to HIV-1. Some of these responses strongly correlate with protection from HIV acquisition and/or disease progression, thus providing significant clues of the ideal components of an efficacious HIV vaccine. In this study, we provide an overview of the key features of protective immune responses found in HEPS, elite and viremic controllers, and discuss how these can be achieved through mucosal immunization. Inevitably, HIV vaccine development research will have to consider strategies that elicit potent antibody and cellular immune responses within the genitorectal mucosa or induction of systemic immune cells with an inherent potential to home and persist at mucosal sites of HIV entry.
Collapse
Affiliation(s)
- Nadia Chanzu
- Institute of Tropical and Infectious Diseases, College of Health Sciences, University of Nairobi , Nairobi , Kenya
| | - Beatrice Ondondo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford , Oxford , UK
| |
Collapse
|
22
|
Abstract
Reverse genetics systems allow artificial generation of non-segmented and segmented negative-sense RNA viruses, like influenza viruses, entirely from cloned cDNA. Since the introduction of reverse genetics systems over a decade ago, the ability to generate ‘designer’ influenza viruses in the laboratory has advanced both basic and applied research, providing a powerful tool to investigate and characterise host–pathogen interactions and advance the development of novel therapeutic strategies. The list of applications for reverse genetics has expanded vastly in recent years. In this review, we discuss the development and implications of this technique, including the recent controversy surrounding the generation of a transmissible H5N1 influenza virus. We will focus on research involving the identification of viral protein function, development of live-attenuated influenza virus vaccines, host–pathogen interactions, immunity and the generation of recombinant influenza virus vaccine vectors for the prevention and treatment of infectious diseases and cancer.
Collapse
|
23
|
Abstract
The influenza virus is a respiratory pathogen with a negative-sense, segmented RNA genome. Construction of recombinant influenza viruses in the laboratory was reported starting in the 1980s. Within a short period of time, pioneer researchers had devised methods that made it possible to construct influenza viral vectors from cDNA plasmid systems. Herein, we discuss the evolution of influenza virus reverse genetics, from helper virus-dependent systems, to helper virus-independent 17-plasmid systems, and all the way to 3- and 1- plasmid systems. Successes in the modification of different gene segments for various applications, including vaccine and gene therapies are highlighted.
Collapse
Affiliation(s)
- Junwei Li
- Center of Excellence for Infectious Diseases, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | | | | |
Collapse
|
24
|
Ye L, Wen Z, Dong K, Wang X, Bu Z, Zhang H, Compans RW, Yang C. Induction of HIV neutralizing antibodies against the MPER of the HIV envelope protein by HA/gp41 chimeric protein-based DNA and VLP vaccines. PLoS One 2011; 6:e14813. [PMID: 21625584 PMCID: PMC3098228 DOI: 10.1371/journal.pone.0014813] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 04/12/2011] [Indexed: 02/06/2023] Open
Abstract
Several conserved neutralizing epitopes have been identified in the HIV Env protein and among these, the MPER of gp41 has received great attention and is widely recognized as a promising target. However, little success has been achieved in eliciting MPER-specific HIV neutralizing antibodies by a number of different vaccine strategies. We investigated the ability of HA/gp41 chimeric protein-based vaccines, which were designed to enhance the exposure of the MPER in its native conformation, to induce MPER-specific HIV neutralizing antibodies. In characterization of the HA/gp41 chimeric protein, we found that by mutating an unpaired Cys residue (Cys-14) in its HA1 subunit to a Ser residue, the modified chimeric protein HA-C14S/gp41 showed increased reactivity to a conformation-sensitive monoclonal antibody against HA and formed more stable trimers in VLPs. On the other hand, HA-C14S/gp41 and HA/gp41 chimeric proteins expressed on the cell surfaces exhibited similar reactivity to monoclonal antibodies 2F5 and 4E10. Immunization of guinea pigs using the HA-C14S/gp41 DNA or VLP vaccines induced antibodies against the HIV gp41 as well as to a peptide corresponding to a segment of MPER at higher levels than immunization by standard HIV VLPs. Further, sera from vaccinated guinea pigs were found to exhibit HIV neutralizing activities. Moreover, sera from guinea pigs vaccinated by HA-C14S/gp41 DNA and VLP vaccines but not the standard HIV VLPs, were found to neutralize HIV pseudovirions containing a SIV-4E10 chimeric Env protein. The virus neutralization could be blocked by a MPER-specific peptide, thus demonstrating induction of MPER-specific HIV neutralizing antibodies by this novel vaccine strategy. These results show that induction of MPER-specific HIV neutralizing antibodies can be achieved through a rationally designed vaccine strategy.
Collapse
Affiliation(s)
- Ling Ye
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (LY); (CY)
| | - Zhiyuan Wen
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- Agriculture Ministry Key Laboratory of Veterinary Public Health, Harbin Veterinary Research Institute, Harbin, People's Republic of China
| | - Ke Dong
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- Central Laboratory, Tangdu Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xi Wang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- Central Laboratory, Tangdu Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Zhigao Bu
- Agriculture Ministry Key Laboratory of Veterinary Public Health, Harbin Veterinary Research Institute, Harbin, People's Republic of China
| | - Huizhong Zhang
- Central Laboratory, Tangdu Hospital, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Richard W. Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Chinglai Yang
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
- * E-mail: (LY); (CY)
| |
Collapse
|
25
|
Krenn BM, Egorov A, Romanovskaya-Romanko E, Wolschek M, Nakowitsch S, Ruthsatz T, Kiefmann B, Morokutti A, Humer J, Geiler J, Cinatl J, Michaelis M, Wressnigg N, Sturlan S, Ferko B, Batishchev OV, Indenbom AV, Zhu R, Kastner M, Hinterdorfer P, Kiselev O, Muster T, Romanova J. Single HA2 mutation increases the infectivity and immunogenicity of a live attenuated H5N1 intranasal influenza vaccine candidate lacking NS1. PLoS One 2011; 6:e18577. [PMID: 21490925 PMCID: PMC3072404 DOI: 10.1371/journal.pone.0018577] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 03/06/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND H5N1 influenza vaccines, including live intranasal, appear to be relatively less immunogenic compared to seasonal analogs. The main influenza virus surface glycoprotein hemagglutinin (HA) of highly pathogenic avian influenza viruses (HPAIV) was shown to be more susceptible to acidic pH treatment than that of human or low pathogenic avian influenza viruses. The acidification machinery of the human nasal passageway in response to different irritation factors starts to release protons acidifying the mucosal surface (down to pH of 5.2). We hypothesized that the sensitivity of H5 HA to the acidic environment might be the reason for the low infectivity and immunogenicity of intranasal H5N1 vaccines for mammals. METHODOLOGY/PRINCIPAL FINDINGS We demonstrate that original human influenza viruses infect primary human nasal epithelial cells at acidic pH (down to 5.4), whereas H5N1 HPAIVs lose infectivity at pH ≤ 5.6. The HA of A/Vietnam/1203/04 was modified by introducing the single substitution HA2 58K→I, decreasing the pH of the HA conformational change. The H5N1 reassortants containing the indicated mutation displayed an increased resistance to acidic pH and high temperature treatment compared to those lacking modification. The mutation ensured a higher viral uptake as shown by immunohistochemistry in the respiratory tract of mice and 25 times lower mouse infectious dose₅₀. Moreover, the reassortants keeping 58K→I mutation designed as a live attenuated vaccine candidate lacking an NS1 gene induced superior systemic and local antibody response after the intranasal immunization of mice. CONCLUSION/SIGNIFICANCE Our finding suggests that an efficient intranasal vaccination with a live attenuated H5N1 virus may require a certain level of pH and temperature stability of HA in order to achieve an optimal virus uptake by the nasal epithelial cells and induce a sufficient immune response. The pH of the activation of the H5 HA protein may play a substantial role in the infectivity of HPAIVs for mammals.
Collapse
Affiliation(s)
| | - Andrej Egorov
- Avir Green Hills Biotechnology AG, Vienna, Austria
- * E-mail:
| | | | | | | | | | | | | | | | - Janina Geiler
- Institute for Medical Virology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Jindrich Cinatl
- Institute for Medical Virology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Martin Michaelis
- Institute for Medical Virology, Johann Wolfgang Goethe University, Frankfurt, Germany
| | | | | | - Boris Ferko
- Avir Green Hills Biotechnology AG, Vienna, Austria
| | - Oleg V. Batishchev
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Andrey V. Indenbom
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry of Russian Academy of Sciences (RAS), Moscow, Russia
| | - Rong Zhu
- Christian Doppler Laboratory of Nanoscopic Methods in Biophysics, Institute for Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Markus Kastner
- Christian Doppler Laboratory of Nanoscopic Methods in Biophysics, Institute for Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Peter Hinterdorfer
- Christian Doppler Laboratory of Nanoscopic Methods in Biophysics, Institute for Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Oleg Kiselev
- Influenza Research Institute, Russian Academy of Medical Sciences, St. Petersburg, Russia
| | | | | |
Collapse
|
26
|
Generation of replication-competent recombinant influenza A viruses carrying a reporter gene harbored in the neuraminidase segment. J Virol 2010; 84:12075-81. [PMID: 20826692 DOI: 10.1128/jvi.00046-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Replication-competent influenza viruses carrying reporter genes are of great use for basic research, screening of antiviral drugs, and neutralizing of antibodies. In this study, two recombinant influenza A viruses with a neuraminidase (NA) segment harboring enhanced green fluorescent protein (EGFP) in the background of A/PR/8/34 (PR8) were generated. The viral RNA (vRNA)-specific packaging signals for NA were largely retained for efficient packaging. An "autocleave" 2A peptide sequence, which was inserted at the N terminus or the COOH terminus of NA to link with EGFP, enabled NA and EGFP to be expressed monocistronically. Further analysis demonstrated that both viruses, named rPR8-EGFP+NA and rPR8-NA+EGPF, although with some characteristic differences in growth and EGFP expression, could replicate in noncomplementary cells and propagate to large quantities while maintaining genome stability after multiple passages in embryonated eggs. These replication-competent influenza viruses carrying reporter genes are a great addition to the tool set for developing antiviral therapeutics and vaccines and for in vivo studies of viral dissemination and pathogenicity.
Collapse
|
27
|
Machado AV, Caetano BC, Barbosa RP, Salgado APC, Rabelo RH, Garcia CC, Bruna-Romero O, Escriou N, Gazzinelli RT. Prime and boost immunization with influenza and adenovirus encoding the Toxoplasma gondii surface antigen 2 (SAG2) induces strong protective immunity. Vaccine 2010; 28:3247-56. [PMID: 20189485 DOI: 10.1016/j.vaccine.2010.02.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 01/21/2010] [Accepted: 02/03/2010] [Indexed: 11/25/2022]
Abstract
In this work, we explored an original vaccination protocol using recombinant influenza and adenovirus. We constructed recombinant influenza viruses harboring dicistronic NA segments containing the surface antigen 2 (SAG2) from Toxoplasma gondii under control of the duplicated 3' promoter. Recombinant influenza viruses were able to drive the expression of the foreign SAG2 sequence in cell culture and to replicate efficiently both in cell culture and in lungs of infected mice. In addition, mice primed with recombinant influenza virus and boosted with a recombinant adenovirus encoding SAG2 elicited both humoral and cellular immune responses specific for SAG2. Moreover, when immunized animals were challenged with the cystogenic P-Br strain of T. gondii, they displayed up to 85% of reduction in parasite burden. These results demonstrate the potential use of recombinant influenza vectors harboring the dicistronic segments in the development of vaccines against infectious diseases.
Collapse
Affiliation(s)
- Alexandre V Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-910, MG, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wu R, Guan Y, Yang Z, Chen J, Wang H, Chen Q, Sui Z, Fang F, Chen Z. A live bivalent influenza vaccine based on a H9N2 virus strain. Vaccine 2009; 28:673-80. [PMID: 19892041 DOI: 10.1016/j.vaccine.2009.10.102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2008] [Revised: 10/11/2009] [Accepted: 10/19/2009] [Indexed: 10/20/2022]
Abstract
The purpose of this study was to construct an H9N2 virus-based bivalent live vaccine expressing the protective antigen of a different subtype of influenza virus. Reverse genetics was used to generate an influenza virus containing nine gene segments derived from the A/Chicken/Jiangsu/11/2002 (H9N2) strain, including independent M1 and M2 matrix gene segments. A recombinant virus expressing the H1N1 HA1 hemagglutinin protein was produced on this framework by substituting the extracellular domain of the H9N2 M2 gene with the H1N1 HA1 fragment from A/PR/8/34 (PR8, H1N1). The resulting hybrid virus H9N2-PR8/HA1 was genetically stable and of low pathogenicity. Intra-nasal immunization of BALB/c mice with H9N2-PR8/HA1 virus induced both anti-H9N2 virus and anti-PR8 HA antibodies and conferred protection to mice against lethal challenge (40x LD(50)) with either H1N1 or H9N2 viruses. This study provides a new influenza H9N2 virus model for the expression and/or delivery of foreign antigens.
Collapse
Affiliation(s)
- Rui Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Characterization of recombinant influenza A virus as a vector for HIV-1 p17Gag. Vaccine 2009; 27:5735-9. [PMID: 19647812 DOI: 10.1016/j.vaccine.2009.07.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/10/2009] [Accepted: 07/14/2009] [Indexed: 11/21/2022]
Abstract
We have generated a recombinant influenza A virus with the HIV-1 p17(Gag) (rFlu-p17) gene inserted into the influenza virus neuraminidase (NA) gene. Expression of HIV-1 p17 protein was detected by conventional Western blot analysis and also by liquid chromatography/tandem mass spectrometry (LC-MS/MS) analysis of rFlu-p17 infected cells. The latter method does not depend on protein-specific antibody preparations and proved to be a powerful tool to detect proteins of interest. Next, antigen presentation of p17 expressed after infection of antigen-presenting cells was determined. Cloned p17-specific CD8+ T-cells were co-cultured with rFlu-p17 infected B-cells and produced IFN-gamma upon stimulation. Furthermore, we showed that immunization with rFlu-p17 elicited a humoral immune response in mice. This study shows that replication-deficient rFlu-p17 is antigenic in vitro and immunogenic in vivo and warrants further development as a candidate vaccine vector.
Collapse
|
30
|
Abstract
The antiquated system used to manufacture the currently licensed inactivated influenza virus vaccines would not be adequate during an influenza virus pandemic. There is currently a search for vaccines that can be developed faster and provide superior, long-lasting immunity to influenza virus as well as other highly pathogenic viruses and bacteria. Recombinant vectors provide a safe and effective method to elicit a strong immune response to a foreign protein or epitope. This review explores the advantages and limitations of several different vectors that are currently being tested, and highlights some of the newer viruses being used as recombinant vectors.
Collapse
|
31
|
Kopecky-Bromberg SA, Fraser KA, Pica N, Carnero E, Moran TM, Franck RW, Tsuji M, Palese P. Alpha-C-galactosylceramide as an adjuvant for a live attenuated influenza virus vaccine. Vaccine 2009; 27:3766-74. [PMID: 19464560 DOI: 10.1016/j.vaccine.2009.03.090] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Revised: 03/10/2009] [Accepted: 03/26/2009] [Indexed: 10/20/2022]
Abstract
There is a substantial need to develop better influenza virus vaccines that can protect populations that are not adequately protected by the currently licensed vaccines. While live attenuated influenza virus vaccines induce superior immune responses compared to inactivated vaccines, the manufacturing process of both types of influenza virus vaccines is time consuming and may not be adequate during a pandemic. Adjuvants would be particularly useful if they could enhance the immune response to live attenuated influenza virus vaccines so that the amount of vaccine needed for a protective dose could be reduced. The glycolipid, alpha-galactosylceramide (alpha-GalCer), has recently been shown to have adjuvant activity for both inactivated and replicating recombinant vaccines. The goal of these experiments was to determine whether a derivative of alpha-GalCer, alpha-C-galactosylceramide (alpha-C-GalCer) can enhance the immune response elicited by a live attenuated influenza virus vaccine containing an NS1 protein truncation and reduce the amount of vaccine required to provide protection after challenge. Our results indicated that the adjuvant reduced both morbidity and mortality in BALB/c mice after challenge with wild type influenza virus. The adjuvant also increased the amount of influenza virus specific total IgG, IgG1, and IgG2a antibodies as well as IFN-gamma secreting CD8(+) T cells. By using knockout mice that are not able to generate NKT cells, we were able to demonstrate that the mechanism of adjuvant activity is dependent on NKT cells. Thus, our data indicate that stimulators of NKT cells represent a new avenue of adjuvants to pursue for live attenuated virus vaccines.
Collapse
Affiliation(s)
- Sarah A Kopecky-Bromberg
- Department of Microbiology, Mount Sinai School of Medicine, New York, NY 10029-6574, United States
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Zolla-Pazner S, Cohen SS, Krachmarov C, Wang S, Pinter A, Lu S. Focusing the immune response on the V3 loop, a neutralizing epitope of the HIV-1 gp120 envelope. Virology 2007; 372:233-46. [PMID: 18061228 DOI: 10.1016/j.virol.2007.09.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 06/25/2007] [Accepted: 09/20/2007] [Indexed: 11/28/2022]
Abstract
Rabbits were immunized with a novel regimen designed to focus the immune response on a single neutralizing epitope of HIV-1 gp120 and thereby preferentially induce neutralizing antibodies (Abs). Animals were primed with gp120 DNA from a clade A Env bearing the GPGR V3 motif and/or a clade C Env bearing the GPGQ V3 motif, and boosted with one or more fusion proteins containing V3 sequences from clades A, B and/or C. Immune sera neutralized three of four Tier 1 primary isolates, including strains heterologous to the immunizing strains, and potent cross-clade-neutralizing activity was demonstrated against V3 chimeric pseudoviruses carrying in a Tier 1 Env, the consensus V3 sequences from clades A1, AG, B, AE, or F. The broadest and most potent neutralizing responses were elicited with the clade C gp120 DNA and a combination of V3-fusion proteins from clades A, B and C. Neutralizing activity was primarily due to V3-specific Abs. The results demonstrate that the immune response can be focused on a neutralizing epitope and show that the anti-V3 Abs induced recognize a diverse set of V3 loops.
Collapse
|
33
|
Abstract
Newly optimized reverse genetics techniques have allowed influenza researchers to generate recombinant influenza viruses expressing mutant viral proteins, as well as foreign proteins. Approaches include the insertion of noninfluenza epitopes and polypeptides into viral glycoproteins, foreign open reading frames as additional segments, and the fusion of independent proteins into viral genes encoding glycoproteins or the nonstructural protein 1. These genetically engineered viruses have been demonstrated to be good viral vectors for mounting B- and T-cell responses and are attractive candidates for vaccine development. As the molecular biology of influenza viral infection is more fully understood, influenza vectors can be concurrently manipulated to produce designed chimeric viruses, unveiling the possibility of a prosperous future with cheap, effective and safe vaccines against different human diseases.
Collapse
Affiliation(s)
- Luis Martínez-Sobrido
- Mount Sinai School of Medicine, Department of Microbiology, Emerging Pathogens Institute, 1 Gustave L Levy Place, Box #1124, NY 10029, USA
| | - Adolfo García-Sastre
- Mount Sinai School of Medicine, Department of Microbiology, Emerging Pathogens Institute & Department of Medicine, Division of Infectious Diseases, 1 Gustave L Levy Place, Box #1124, NY 10029, USA
| |
Collapse
|
34
|
Garulli B, Meola M, Stillitano MG, Kawaoka Y, Castrucci MR. Efficient vagina-to-lower respiratory tract immune trafficking in a murine model of influenza A virus infection. Virology 2007; 361:274-82. [PMID: 17222437 DOI: 10.1016/j.virol.2006.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 08/23/2006] [Accepted: 12/01/2006] [Indexed: 11/30/2022]
Abstract
Effective vaccination strategies for infectious diseases take into account the induction, long-term maintenance and recall of memory T-cell populations. To understand the immunological cross-talk within the mucosal compartments, we compared intranasal to vaginal immunization and demonstrated that vaginal infection of BALB/c mice with influenza A virus provides protective mucosal immunity against both homosubtypic and heterosubtypic virus challenge in the respiratory tract. We found that, prior to the viral challenge, in vaginally primed mice, antigen-specific CD8+ T cells were not detected in the lung airways and levels of serum antibodies were lower than those observed in intranasally immunized mice. However, following pulmonary challenge, NP147-specific CD8+ T cells were recruited and amplified in vaginally primed mice to the same extent as those in intranasally primed mice. Thus, the long-term memory immune response elicited by vaginal immunization with influenza virus is efficiently recalled and offers reasonable protection against infection in the respiratory tract.
Collapse
Affiliation(s)
- Bruno Garulli
- Department of Infectious, Parasitic and Immune-mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | |
Collapse
|
35
|
Renegar KB, Menge A, Mestecky J. Influenza Virus Infection of the Murine Uterus: A New Model for Antiviral Immunity in the Female Reproductive Tract. Viral Immunol 2006; 19:613-22. [PMID: 17201656 DOI: 10.1089/vim.2006.19.613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Secretory IgA (S-IgA) mediates local immunity to influenza virus in the murine upper respiratory tract and may play an important role in local immunity to various microorganisms in the female reproductive tract as well. Although the presence of IgA in cervicovaginal or uterine secretions has been correlated with immunity to a number of pathogens, there has been no direct demonstration of the mediation of uterine antiviral immunity by S-IgA. Influenza virus, although not a normal pathogen of the reproductive tract, was used to develop a model for the investigation of mucosal immunity in the uterus. PR8 (H1N1) influenza virus injected into the ovarian bursa of BALB/c mice grew well, with peak titers between days 3 and 5. Intravenous injection of polymeric IgA anti-influenza virus monoclonal antibody before or 30 min after viral challenge protected mice against viral infection. We believe this work to be the first direct demonstration of S-IgA-mediated antiviral uterine immunity. It provides a model for further investigation of immunity in the female reproductive tract.
Collapse
Affiliation(s)
- Kathryn B Renegar
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | |
Collapse
|
36
|
Ye L, Sun Y, Lin J, Bu Z, Wu Q, Jiang S, Steinhauer DA, Compans RW, Yang C. Antigenic properties of a transport-competent influenza HA/HIV Env chimeric protein. Virology 2006; 352:74-85. [PMID: 16725170 DOI: 10.1016/j.virol.2006.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Revised: 02/15/2006] [Accepted: 04/12/2006] [Indexed: 10/24/2022]
Abstract
The transmembrane subunit (gp41) of the HIV Env glycoprotein contains conserved neutralizing epitopes which are not well-exposed in wild-type HIV Env proteins. To enhance the exposure of these epitopes, a chimeric protein, HA/gp41, in which the gp41 of HIV-1 89.6 envelope protein was fused to the C-terminus of the HA1 subunit of the influenza HA protein, was constructed. Characterization of protein expression showed that the HA/gp41 chimeric proteins were expressed on cell surfaces and formed trimeric oligomers, as found in the HIV Env as well as influenza HA proteins. In addition, the HA/gp41 chimeric protein expressed on the cell surface can also be cleaved into 2 subunits by trypsin treatment, similar to the influenza HA. Moreover, the HA/gp41 chimeric protein was found to maintain a pre-fusion conformation. Interestingly, the HA/gp41 chimeric proteins on cell surfaces exhibited increased reactivity to monoclonal antibodies against the HIV Env gp41 subunit compared with the HIV-1 envelope protein, including the two broadly neutralizing monoclonal antibodies 2F5 and 4E10. Immunization of mice with a DNA vaccine expressing the HA/gp41 chimeric protein induced antibodies against the HIV gp41 protein and these antibodies exhibit neutralizing activity against infection by an HIV SF162 pseudovirus. These results demonstrate that the construction of such chimeric proteins can provide enhanced exposure of conserved epitopes in the HIV Env gp41 and may represent a novel vaccine design strategy for inducing broadly neutralizing antibodies against HIV.
Collapse
Affiliation(s)
- Ling Ye
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Room 3086 Rollins Research Center, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
HIV poses a serious health threat in the world. Mucosal transmission of HIV through the genitourinary tract may be the most important route of transmission. Intranasal immunisations induce vaginal and systemic immune responses. Various protein-, DNA- and RNA-based immunopotentiating adjuvants/delivery systems and live bacterial and viral vectors are available for intranasal immunisations, and these systems may differ in their ability to induce a specific type of immune response (e.g., a cytotoxic T cell versus an antibody response). As the protection against HIV may require both cytotoxic T cell and antibodies, a combination of adjuvants/delivery systems for combinations of mucosal and parenteral immunisations may be required in order to develop a protective anti-HIV vaccine.
Collapse
Affiliation(s)
- Michael Vajdy
- Chiron Vaccines, 4560 Horton Street, Emeryville, CA 94608, USA.
| | | |
Collapse
|
38
|
Vieira Machado A, Naffakh N, Gerbaud S, van der Werf S, Escriou N. Recombinant influenza A viruses harboring optimized dicistronic NA segment with an extended native 5' terminal sequence: induction of heterospecific B and T cell responses in mice. Virology 2005; 345:73-87. [PMID: 16271378 DOI: 10.1016/j.virol.2005.09.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 06/23/2005] [Accepted: 09/23/2005] [Indexed: 11/27/2022]
Abstract
We generated novel recombinant influenza A viruses (vNA38) harboring dicistronic NA segments with an extended native 5' terminal sequence of 70 nucleotides comprised of the last 42 nucleotides of the NA ORF and the 5' noncoding region (5' NCR). vNA38 viruses replicated stably and more efficiently than vNA35 viruses with a dicistronic NA segment comprised of the native 5' NCR only, that we described previously (Vieira Machado, A., Naffakh, N., van der Werf, S., Escriou, N., 2003. Expression of a foreign gene by stable recombinant influenza viruses harboring a dicistronic genomic segment with an internal promoter. Virology 313, 235-249). In addition, vNA38 viruses drove the expression of higher levels of encoded heterologous proteins than corresponding vNA35 viruses, both in cell culture and in the pulmonary tissue of infected mice. These data demonstrate that a sequence overlapping 5' coding and noncoding regions of the NA segment determines efficient replication and/or propagation of the vRNA. Intranasal immunization of mice with live vNA38 viruses induced B and T cell responses specific for the heterologous protein expressed, establishing the usefulness of such recombinant influenza viruses with a dicistronic segment for the development of live bivalent vaccines.
Collapse
Affiliation(s)
- Alexandre Vieira Machado
- Unité de Génétique Moléculaire des Virus Respiratoires, URA 1966 CNRS, Institut Pasteur, 25 rue du Dr. Roux, 75724 Paris Cedex 15, France.
| | | | | | | | | |
Collapse
|
39
|
Li ZN, Mueller SN, Ye L, Bu Z, Yang C, Ahmed R, Steinhauer DA. Chimeric influenza virus hemagglutinin proteins containing large domains of the Bacillus anthracis protective antigen: protein characterization, incorporation into infectious influenza viruses, and antigenicity. J Virol 2005; 79:10003-12. [PMID: 16014960 PMCID: PMC1181555 DOI: 10.1128/jvi.79.15.10003-10012.2005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Large polypeptides of the Bacillus anthracis protective antigen (PA) were inserted into an influenza A virus hemagglutinin glycoprotein (HA), and the chimeric proteins were functionally characterized and incorporated into infectious influenza viruses. PA domain 1', the region responsible for binding to the other toxin components, the lethal factor and edema factor, and domain 4, the receptor binding domain (RBD), were inserted at the C-terminal flank of the HA signal peptide and incorporated into the HA1 subunit of HA. The chimeric proteins, designated as LEF/HA (90 amino acid insertion) and RBD/HA (140 amino acid insertion), were initially analyzed following expression using recombinant vaccinia viruses. Both chimeric proteins were shown to display functional phenotypes similar to that of the wild-type HA. They transport to the cell surface, can be cleaved into the HA1 and HA2 subunits by trypsin to activate membrane fusion potential, are able to undergo the low-pH-induced conformational changes required for fusion, and are capable of inducing the fusion process. We were also able to generate recombinant influenza viruses containing the chimeric RBD/HA and LEF/HA genes, and the inserted PA domains were maintained in the HA gene segments following several passages in MDCK cells or embryonated chicken eggs. Furthermore, DNA immunization of mice with plasmids that express the chimeric RBD/HA and LEF/HA proteins, and the recombinant viruses containing them, induced antibody responses against both the HA and PA components of the protein. These approaches may provide useful tools for vaccines against anthrax and other diseases.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/blood
- Antibodies, Viral/blood
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacillus anthracis/genetics
- Bacillus anthracis/immunology
- Bacterial Toxins/chemistry
- Bacterial Toxins/genetics
- Bacterial Toxins/immunology
- Chick Embryo
- Female
- Genetic Vectors
- Hemagglutinin Glycoproteins, Influenza Virus/chemistry
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Influenza A virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Mice
- Mice, Inbred C57BL
- Models, Molecular
- Plasmids
- Recombination, Genetic
- Vaccination
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccinia virus
Collapse
Affiliation(s)
- Zhu-Nan Li
- Department of Microbiology and Immunology, Emory University School of Medicine, Rollins Research Center, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Binley JM, Wrin T, Korber B, Zwick MB, Wang M, Chappey C, Stiegler G, Kunert R, Zolla-Pazner S, Katinger H, Petropoulos CJ, Burton DR. Comprehensive cross-clade neutralization analysis of a panel of anti-human immunodeficiency virus type 1 monoclonal antibodies. J Virol 2004; 78:13232-52. [PMID: 15542675 PMCID: PMC524984 DOI: 10.1128/jvi.78.23.13232-13252.2004] [Citation(s) in RCA: 585] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2004] [Accepted: 07/09/2004] [Indexed: 12/20/2022] Open
Abstract
Broadly neutralizing monoclonal antibodies (MAbs) are potentially important tools in human immunodeficiency virus type 1 (HIV-1) vaccine design. A few rare MAbs have been intensively studied, but we still have a limited appreciation of their neutralization breadth. Using a pseudovirus assay, we evaluated MAbs from clade B-infected donors and a clade B HIV(+) plasma against 93 viruses from diverse backgrounds. Anti-gp120 MAbs exhibited greater activity against clade B than non-B viruses, whereas anti-gp41 MAbs exhibited broad interclade activity. Unexpectedly, MAb 4E10 (directed against the C terminus of the gp41 ectodomain) neutralized all 90 viruses with moderate potency. MAb 2F5 (directed against an epitope adjacent to that of 4E10) neutralized 67% of isolates, but none from clade C. Anti-gp120 MAb b12 (directed against an epitope overlapping the CD4 binding site) neutralized 50% of viruses, including some from almost every clade. 2G12 (directed against a high-mannose epitope on gp120) neutralized 41% of the viruses, but none from clades C or E. MAbs to the gp120 V3 loop, including 447-52D, neutralized a subset of clade B viruses (up to 45%) but infrequently neutralized other clades (=7%). MAbs b6 (directed against the CD4 binding site) and X5 (directed against a CD4-induced epitope of gp120) neutralized only sensitive primary clade B viruses. The HIV(+) plasma neutralized 70% of the viruses, including some from all major clades. Further analysis revealed five neutralizing immunotypes that were somewhat associated with clades. As well as the significance for vaccine design, our data have implications for passive-immunization studies in countries where clade C viruses are common, given that only MAbs b12 and 4E10 were effective against viruses from this clade.
Collapse
Affiliation(s)
- James M Binley
- IMM2, Department of Immunology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nakaya Y, Nakaya T, Park MS, Cros J, Imanishi J, Palese P, García-Sastre A. Induction of cellular immune responses to simian immunodeficiency virus gag by two recombinant negative-strand RNA virus vectors. J Virol 2004; 78:9366-75. [PMID: 15308731 PMCID: PMC506935 DOI: 10.1128/jvi.78.17.9366-9375.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A recombinant Newcastle disease virus (rNDV) expressing simian immunodeficiency virus (SIV) Gag protein (rNDV/SIVgag) was generated. The rNDV/SIVgag virus induced Gag-specific cellular immune responses in mice, leading to a specific anti-Gag antiviral immunity. This was evidenced by the inhibition of growth of recombinant vaccinia virus expressing an identical Gag antigen (rVac/SIVgag) but not of wild-type vaccinia virus in rNDV/SIVgag-immunized mice. Among intravenous, intraperitoneal, or intranasal immunization routes, intranasal administration induced the strongest protective response against challenge with rVac/SIVgag. We further demonstrated that these immune responses were greatly enhanced after booster immunization with recombinant influenza viruses expressing immunogenic portions of SIV Gag. The magnitude of the protective immune response correlated with the levels of cellular immune responses to Gag, which were still evident 9 weeks after immunization. These results suggest that rNDV and influenza virus vectors are suitable candidate vaccines against AIDS as well as against other infectious diseases.
Collapse
Affiliation(s)
- Yurie Nakaya
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The BM2 protein of influenza B virus functions as an ion channel, which is suggested to be important for virus uncoating in endosomes of virus-infected cells. Because direct support for this function is lacking, whether BM2 plays an essential role in the viral life cycle remains unknown. We therefore attempted to generate BM2 knockout viruses by reverse genetics. Mutant viruses possessing M segments with the mutated initiation codon of BM2 protein at the stop-start pentanucleotide were viable and still expressed BM2. The introduction of multiple stop codons and a one-nucleotide deletion downstream of the stop-start pentanucleotide, in addition to disablement of the BM2 initiation codon, failed to generate viable mutant viruses, but the mutant M segments still expressed proteins that reacted with the BM2 peptide antiserum. To completely abolish BM2 expression, we generated a mutant M gene whose BM2 open reading frame was deleted. Although this mutant was not able to replicate in normal MDCK cells, it did replicate in a cell line that we established which constitutively expresses BM2. Furthermore, a virus possessing the mutant M gene lacking the BM2 open reading frame and a mutant NA gene containing the BM2 open reading frame instead of the NA open reading frame underwent multiple cycles of replication in MDCK cells, with exogenous sialidase used to supplement the deleted viral sialidase activity. These findings demonstrate that the BM2 protein is essential for influenza B virus replication.
Collapse
Affiliation(s)
- Masato Hatta
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | |
Collapse
|
43
|
Schlehuber LD, Rose JK. Prediction and identification of a permissive epitope insertion site in the vesicular stomatitis virus glycoprotein. J Virol 2004; 78:5079-87. [PMID: 15113889 PMCID: PMC400361 DOI: 10.1128/jvi.78.10.5079-5087.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2003] [Accepted: 12/29/2003] [Indexed: 11/20/2022] Open
Abstract
We developed a rational approach to identify a site in the vesicular stomatitis virus (VSV) glycoprotein (G) that is exposed on the protein surface and tolerant of foreign epitope insertion. The foreign epitope inserted was the six-amino-acid sequence ELDKWA, a sequence in a neutralizing epitope from human immunodeficiency virus type 1. This sequence was inserted into six sites within the VSV G protein (Indiana serotype). Four sites were selected based on hydrophilicity and high sequence variability identified by sequence comparison with other vesiculovirus G proteins. The site showing the highest variability was fully tolerant of the foreign peptide insertion. G protein containing the insertion at this site folded correctly, was transported normally to the cell surface, had normal membrane fusion activity, and could reconstitute fully infectious VSV. The virus was neutralized by the human 2F5 monoclonal antibody that binds the ELDKWA epitope. Additional studies showed that this site in G protein tolerated insertion of at least 16 amino acids while retaining full infectivity. The three other insertions in somewhat less variable sequences interfered with VSV G folding and transport to the cell surface. Two additional insertions were made in a conserved sequence adjacent to a glycosylation site and near the transmembrane domain. The former blocked G-protein transport, while the latter allowed transport to the cell surface but blocked membrane fusion activity of G protein. Identification of an insertion-tolerant site in VSV G could be important in future vaccine and targeting studies, and the general principle might also be useful in other systems.
Collapse
Affiliation(s)
- Lisa D Schlehuber
- Section of Microbial Pathogenesis, Yale University School of Medicine, 310 Cedar Street, New Haven, CT 06510, USA
| | | |
Collapse
|
44
|
Shinya K, Fujii Y, Ito H, Ito T, Kawaoka Y. Characterization of a neuraminidase-deficient influenza a virus as a potential gene delivery vector and a live vaccine. J Virol 2004; 78:3083-8. [PMID: 14990727 PMCID: PMC353727 DOI: 10.1128/jvi.78.6.3083-3088.2004] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We recently identified a packaging signal in the neuraminidase (NA) viral RNA (vRNA) segment of an influenza A virus, allowing us to produce a mutant virus [GFP(NA)-Flu] that lacks most of the NA open reading frame but contains instead the gene encoding green fluorescent protein (GFP). To exploit the expanding knowledge of vRNA packaging signals to establish influenza virus vectors for the expression of foreign genes, we studied the replicative properties of this virus in cell culture and mice. Compared to wild-type virus, GFP(NA)-Flu was highly attenuated in normal cultured cells but was able to grow to a titer of >10(6) PFU/ml in a mutant cell line expressing reduced levels of sialic acid on the cell surface. GFP expression from this virus was stable even after five passages in the latter cells. In intranasally infected mice, GFP was detected in the epithelial cells of nasal mucosa, bronchioles, and alveoli for up to 4 days postinfection. We attribute the attenuated growth of GFP(NA)-Flu to virion aggregation at the surface of bronchiolar epithelia. In studies to test the potential of this mutant as a live attenuated influenza vaccine, all mice vaccinated with >/==" BORDER="0">10(5) PFU of GFP(NA)-Flu survived when challenged with lethal doses of the parent virus. These results suggest that influenza virus could be a useful vector for expressing foreign genes and that a sialidase-deficient virus may offer an alternative to the live influenza vaccines recently approved for human use.
Collapse
Affiliation(s)
- Kyoko Shinya
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, USA
| | | | | | | | | |
Collapse
|
45
|
Garulli B, Kawaoka Y, Castrucci MR. Mucosal and systemic immune responses to a human immunodeficiency virus type 1 epitope induced upon vaginal infection with a recombinant influenza A virus. J Virol 2004; 78:1020-5. [PMID: 14694134 PMCID: PMC368805 DOI: 10.1128/jvi.78.2.1020-1025.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The humoral and cellular immune responses in the genital mucosa likely play an important role in the prevention of sexually transmitted infections, including infection with human immunodeficiency virus type 1 (HIV-1). Here we show that vaginal infection of progesterone-treated BALB/c mice with a recombinant influenza virus bearing the immunodominant P18IIIB cytotoxic T-lymphocyte (CTL) epitope of the gp160 envelope protein from an HIV-1 IIIB isolate (P18IIIB; RIQRGPGRAFVTIGK) can induce a specific immune response in regional mucosal lymph nodes, as well as in a systemic site (the spleen). A single inoculation of mice with the recombinant influenza virus induced long-lasting (at least 5 months) antigen-specific CTL memory detectable as a rapid recall of effector CTLs upon vaginal infection with recombinant vaccinia virus expressing HIV-1 IIIB envelope gene products. Long-term antigen-specific CTL memory was also induced and maintained in distant mucosal tissues when mice were intranasally immunized with the recombinant influenza virus. These results indicate that mucosal immunization and, in particular, local vaginal immunization with recombinant influenza virus can provide strong, durable immune responses in the female genital tract of mice.
Collapse
Affiliation(s)
- Bruno Garulli
- Laboratory of Virology, Istituto Superiore di Sanità, 00161 Rome, Italy
| | | | | |
Collapse
|
46
|
Abstract
This article gives an overview about the development of an HIV-1 vaccine. Tremendous numbers of papers have been published on this topic during the last 10 years, and this article can only touch on the different directions taken toward the development of an HIV-1 vaccine, and not give a complete overview of the entire field.
Collapse
Affiliation(s)
- James P McGettigan
- Department of Biochemistry and Molecular Pharmacology, Dorrance H. Hamilton Laboratories, Center for Human Virology, Philadelphia, PA 19107-6799, USA
| | | | | |
Collapse
|
47
|
Fujii Y, Goto H, Watanabe T, Yoshida T, Kawaoka Y. Selective incorporation of influenza virus RNA segments into virions. Proc Natl Acad Sci U S A 2003; 100:2002-7. [PMID: 12574509 PMCID: PMC149948 DOI: 10.1073/pnas.0437772100] [Citation(s) in RCA: 241] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The genome of influenza A virus is comprised of eight viral RNA (vRNA) segments. Although the products of all eight vRNA segments must be present for viral replication, little is known about the mechanism(s) responsible for incorporation of these segments into virions. Two models have been proposed for the generation of infectious virions containing eight vRNA segments. The random-incorporation model assumes a common structural feature in all the vRNAs, enabling any combination of vRNAs to be incorporated randomly into virions. The selective-incorporation model predicts the presence of specific structures in each vRNA segment, leading to the incorporation of a set of eight vRNA segments into virions. Here we demonstrate that eight different vRNA segments must be present for efficient virion formation and that sequences within the coding region of (and thus unique to) the neuraminidase vRNA possess a signal that drives incorporation of this segment into virions. These findings indicate a unique contribution from individual vRNA segments and thus suggest a selective (rather than random) mechanism of vRNA recruitment into virions. The neuraminidase vRNA incorporation signal and others yet to be identified should provide attractive targets for the attenuation of influenza viruses in vaccine production and the design of new antiviral drugs.
Collapse
Affiliation(s)
- Yutaka Fujii
- Department of Virology, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | | | | | | | | |
Collapse
|
48
|
Neumann G, Whitt MA, Kawaoka Y. A decade after the generation of a negative-sense RNA virus from cloned cDNA - what have we learned? J Gen Virol 2002; 83:2635-2662. [PMID: 12388800 DOI: 10.1099/0022-1317-83-11-2635] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Since the first generation of a negative-sense RNA virus entirely from cloned cDNA in 1994, similar reverse genetics systems have been established for members of most genera of the Rhabdo- and Paramyxoviridae families, as well as for Ebola virus (Filoviridae). The generation of segmented negative-sense RNA viruses was technically more challenging and has lagged behind the recovery of nonsegmented viruses, primarily because of the difficulty of providing more than one genomic RNA segment. A member of the Bunyaviridae family (whose genome is composed of three RNA segments) was first generated from cloned cDNA in 1996, followed in 1999 by the production of influenza virus, which contains eight RNA segments. Thus, reverse genetics, or the de novo synthesis of negative-sense RNA viruses from cloned cDNA, has become a reliable laboratory method that can be used to study this large group of medically and economically important viruses. It provides a powerful tool for dissecting the virus life cycle, virus assembly, the role of viral proteins in pathogenicity and the interplay of viral proteins with components of the host cell immune response. Finally, reverse genetics has opened the way to develop live attenuated virus vaccines and vaccine vectors.
Collapse
Affiliation(s)
- Gabriele Neumann
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, 2015 Linden Drive West, Madison, WI 53706, USA1
| | - Michael A Whitt
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN, USA2
| | - Yoshihiro Kawaoka
- CREST, Japan Science and Technology Corporation, Japan4
- Institute of Medical Science, University of Tokyo, Tokyo, Japan3
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, 2015 Linden Drive West, Madison, WI 53706, USA1
| |
Collapse
|
49
|
Decroix N, Quan CP, Pamonsinlapatham P, Bouvet JP. Mucosal immunity induced by intramuscular administration of free peptides in-line with PADRE: IgA antibodies to the ELDKWA epitope of HIV gp41. Scand J Immunol 2002; 56:59-65. [PMID: 12100472 DOI: 10.1046/j.1365-3083.2002.01113.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To improve the mucosal antibody response against a short amino acid (aa) sequence (ELDKWA) of HIV gp41, we have investigated a construction including this peptide in-line with the Pan DR epitope (PADRE). ELDKWA is a conserved peptide playing a key role in the pathogenicity of HIV transmission. PADRE is a non-natural peptide with multipotential immunogenic properties. The results show striking differences between mucosal and systemic immune systems, with a preferential response of the mucosal organs. In contrast with most mucosal immunizations, the intracellular response persists for over two months after the last injection. This strongly suggests that further investigations of conserved key epitopes from various pathogens may lead to safe and chemically defined mucosal vaccines with synthetic peptides. These candidate vaccines with free peptides may be suitable for mass campaigns even in developing countries.
Collapse
Affiliation(s)
- N Decroix
- Unité d'Immunopathologie humaine INSERM UR 430, Université Paris VI, France
| | | | | | | |
Collapse
|
50
|
Parks GD, Alexander-Miller MA. High avidity cytotoxic T lymphocytes to a foreign antigen are efficiently activated following immunization with a recombinant paramyxovirus, simian virus 5. J Gen Virol 2002; 83:1167-1172. [PMID: 11961272 DOI: 10.1099/0022-1317-83-5-1167] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Our previous work has shown that high avidity cytotoxic T lymphocytes (CTL) are optimal for virus clearance in vivo and thus it is necessary that an effective vaccine is capable of eliciting high avidity CTL. To determine if vaccination with the paramyxovirus simian virus 5 (SV5) elicits a high avidity response to a model foreign antigen, a recombinant virus was engineered to express chicken ovalbumin (rSV5-Ova). To compare the CTL response elicited with rSV5-Ova and a recombinant vaccinia virus expressing ovalbumin (rVV-Ova), mice were vaccinated intranasally with various doses of each vector and the Ova-specific CTL response was determined by ELISPOT analysis. Here, it has been shown that rSV5 can be equally as effective as rVV in eliciting antigen-specific CTL, in terms of both the total number of CTL and the number of high avidity cells. This has implications for both the design of vaccine vectors and the route utilized for vaccine administration for the elicitation of high avidity CTL responses. The advantages and future potential use of rSV5 vaccine vectors are discussed.
Collapse
Affiliation(s)
- Griffith D Parks
- Department of Microbiology, Wake Forest University School of Medicine, Room 5108 Gray Building, Medical Center Boulevard, Winston-Salem, NC 27157, USA1
| | - Martha A Alexander-Miller
- Department of Microbiology, Wake Forest University School of Medicine, Room 5108 Gray Building, Medical Center Boulevard, Winston-Salem, NC 27157, USA1
| |
Collapse
|