1
|
Madapong A, Petro-Turnquist EM, Webby RJ, McCormick AA, Weaver EA. Immunity and Protective Efficacy of a Plant-Based Tobacco Mosaic Virus-like Nanoparticle Vaccine against Influenza a Virus in Mice. Vaccines (Basel) 2024; 12:1100. [PMID: 39460267 PMCID: PMC11510914 DOI: 10.3390/vaccines12101100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The rapid production of influenza vaccines is crucial to meet increasing pandemic response demands. Here, we developed plant-made vaccines comprising centralized consensus influenza hemagglutinin (HA-con) proteins (H1 and H3 subtypes) conjugated to a modified plant virus, tobacco mosaic virus (TMV) nanoparticle (TMV-HA-con). METHODS We compared immune responses and protective efficacy against historical H1 or H3 influenza A virus infections among TMV-HA-con, HA-con protein combined with AddaVax™ adjuvant, and whole-inactivated virus vaccine (Fluzone®). RESULTS Immunogenicity studies demonstrated robust IgG, IgM, and IgA responses in the TMV-HA-con and HA-con protein vaccinated groups, with relatively low induction of interferon (IFN)-γ+ T-cell responses across all vaccinated groups. The TMV-HA-con and HA-con protein groups displayed partial protection (100% and 80% survival) with minimal weight loss following challenge with two H1N1 strains. The HA-con protein group exhibited 80% and 100% survival against two H3 strains, whereas the TMV-HA-con groups showed reduced protection (20% survival). The Fluzone® group conferred 20-100% survival against two H1N1 strains and one H3N1 strain, but did not protect against H3N2 infection. CONCLUSIONS Our findings indicate that TMV-HA and HA-con protein vaccines with adjuvant induce protective immune responses against influenza A virus infections. Furthermore, our results underscore the potential of plant-based production using TMV-like nanoparticles for developing influenza A virus candidate vaccines.
Collapse
Affiliation(s)
- Adthakorn Madapong
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (A.M.); (E.M.P.-T.)
| | - Erika M. Petro-Turnquist
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (A.M.); (E.M.P.-T.)
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| | | | | | - Eric A. Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68503, USA; (A.M.); (E.M.P.-T.)
- School of Biological Sciences, College of Arts and Sciences, University of Nebraska-Lincoln, Lincoln, NE 68503, USA
| |
Collapse
|
2
|
Avižinienė A, Dalgėdienė I, Armalytė J, Petraitytė-Burneikienė R. Immunogenicity of novel vB_EcoS_NBD2 bacteriophage-originated nanotubes as a carrier for peptide-based vaccines. Virus Res 2024; 345:199370. [PMID: 38614253 PMCID: PMC11059446 DOI: 10.1016/j.virusres.2024.199370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Non-infectious virus-like nanoparticles mimic native virus structures and can be modified by inserting foreign protein fragments, making them immunogenic tools for antigen presentation. This study investigated, for the first time, the immunogenicity of long and flexible polytubes formed by yeast-expressed tail tube protein gp39 of bacteriophage vB_EcoS_NBD2 and evaluated their ability to elicit an immune response against the inserted protein fragments. Protein gp39-based polytubes induced humoral immune response in mice, even without the use of adjuvant. Bioinformatics analysis guided the selection of protein fragments from Acinetobacter baumannii for insertion into the C-terminus of gp39. Chimeric polytubes, displaying 28-amino acid long OmpA protein fragment, induced IgG response against OmpA protein fragment in immunized mice. These polytubes demonstrated their effectiveness both as antigen carrier and an adjuvant, when the OmpA fragments were either displayed on chimeric polytubes or used alongside with the unmodified polytubes. Our findings expand the potential applications of long and flexible polytubes, contributing to the development of novel antigen carriers with improved immunogenicity and antigen presentation capabilities.
Collapse
Affiliation(s)
- Aliona Avižinienė
- Department of Eukaryote Gene Engineering, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, Lithuania.
| | - Indrė Dalgėdienė
- Department of Immunology, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, Lithuania
| | - Julija Armalytė
- Institute of Biosciences, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, Lithuania
| | - Rasa Petraitytė-Burneikienė
- Department of Eukaryote Gene Engineering, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, Lithuania
| |
Collapse
|
3
|
Kumar R, Bera BC, Anand T, Pavulraj S, Kurian Mathew M, Gupta RP, Tripathi BN, Virmani N. Evaluation of immunogenicity and protective efficacy of bacteriophage conjugated haemagglutinin based subunit vaccine against equine influenza virus in a murine model. Vet Res Commun 2024; 48:1707-1726. [PMID: 38528300 DOI: 10.1007/s11259-024-10356-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/13/2024] [Indexed: 03/27/2024]
Abstract
Equine influenza (EI) is a highly contagious acute respiratory disease of equines caused by the H3N8 subtype of Influenza A virus i.e. equine influenza virus (EIV). Vaccination is an important and effective tool for the control of EI in equines. Most of the commercial influenza vaccines are produced in embryonated hen's eggs which has several inherent disadvantages. Hence, subunit vaccine based on recombinant haemagglutinin (HA) antigen, being the most important envelope glycoprotein has been extensively exploited for generating protective immune responses, against influenza A and B viruses. We hypothesized that novel vaccine formulation using baculovirus expressed recombinant HA1 (rHA1) protein coupled with bacteriophage will generate strong protective immune response against EIV. In the present study, the recombinant HA1 protein was produced in insect cells using recombinant baculovirus having cloned HA gene of EIV (Florida clade 2 sublineage) and the purified rHA1 was chemically coupled with bacteriophage using a crosslinker to produce rHA1-phage vaccine candidate. The protective efficacy of vaccine preparations of rHA1-phage conjugate and only rHA1 proteins were evaluated in mouse model through assessing serology, cytokine profiling, clinical signs, gross and histopathological changes, immunohistochemistry, and virus quantification. Immunization of vaccine preparations have stimulated moderate antibody response (ELISA titres-5760 ± 640 and 11,520 ± 1280 for rHA1 and rHA1-phage, respectively at 42 dpi) and elicited strong interferon (IFN)-γ expression levels after three immunizations of vaccine candidates. The immunized BALB/c mice were protected against challenge with wild EIV and resulted in reduced clinical signs and body weight loss, reduced pathological changes, decreased EIV antigen distribution, and restricted EIV replication in lungs and nasopharynx. In conclusion, the immune responses with moderate antibody titer and significantly higher cytokine responses generated by the rHA1-phage vaccine preparation without any adjuvant could be a novel vaccine candidate for quick vaccine preparation through further trials of vaccine in the natural host.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Veterinary Public Health and Epidemiology, LUVAS, Hisar, Haryana, 125004, India
| | | | - Taruna Anand
- ICAR- National Research Centre on Equines, Hisar, Haryana, 125 001, India
| | - Selvaraj Pavulraj
- Louisiana State University School of Veterinary Medicine, Skip Bertman Dr, Baton Rouge, LA, 70803, USA
| | - Manu Kurian Mathew
- ICAR- National Research Centre on Equines, Hisar, Haryana, 125 001, India
| | - R P Gupta
- Department of Veterinary Pathology, LUVAS, Hisar, Haryana, 125004, India
| | | | - Nitin Virmani
- ICAR- National Research Centre on Equines, Hisar, Haryana, 125 001, India.
| |
Collapse
|
4
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
5
|
Soo Hoo WI, Higa K, McCormick AA. Vaccination against Epstein-Barr Latent Membrane Protein 1 Protects against an Epstein-Barr Virus-Associated B Cell Model of Lymphoma. BIOLOGY 2023; 12:983. [PMID: 37508413 PMCID: PMC10376452 DOI: 10.3390/biology12070983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
In this study, we demonstrate that expression of viral latent membrane protein 1 (LMP1) in a mouse B cell line renders the animals responsive to protection from a 38C13-LMP1 tumor challenge with a novel vaccine. The Epstein-Barr virus (EBV) preferentially infects circulating B lymphocytes, has oncogenic potential, and is associated with a wide variety of B cell lymphomas. EBV is ectotrophic to human cells, and currently there are no B cell animal models of EBV-associated lymphoma that can be used to investigate vaccine immunotherapy. Since most EBV-infected human tumor cells express latent membrane protein 1 (LMP1) on their surface, this viral antigen was tested as a potential target for an anticancer vaccine in a mouse model. Here, we describe a new mouse model of LMP1-expressing B cell lymphoma produced with plasmid transduction of 38C13 into mouse B cells. The expression of LMP-1 was confirmed with a western blot analysis and immunocytochemistry. We then designed a novel LMP1 vaccine, by fusing viral antigen LMP1 surface loop epitopes to the surface of a viral antigen carrier, the Tobacco Mosaic virus (TMV). Vaccinated mice produced high titer antibodies against the TMV-LMP1 vaccine; however, cellular responses were at the baseline, as measured with IFNγ ELISpot. Despite this, the vaccine showed significant protection from a 38C13-LMP1 tumor challenge. To provide additional immune targets, we compared TMV-LMP1 peptide immunization with DNA immunization with the full-length LMP1 gene. Anti-LMP1 antibodies were significantly higher in TMV-LMP1-vaccinated mice compared to the DNA-immunized mice, but, as predicted, DNA-vaccinated mice had improved cellular responses using IFNγ ELISpot. Surprisingly, the TMV-LMP1 vaccine provided protection from a 38C13-LMP1 tumor challenge, while the DNA vaccine did not. Thus, we demonstrated that LMP1 expression in a mouse B cell line is responsive to antibody immunotherapy that may be applied to EBV-associated disease.
Collapse
Affiliation(s)
- Wesley I Soo Hoo
- College of Pharmacy, Touro University California, 1310 Club Drive, Mare Island, Vallejo, CA 94592, USA
| | - Kaylie Higa
- College of Pharmacy, Touro University California, 1310 Club Drive, Mare Island, Vallejo, CA 94592, USA
| | - Alison A McCormick
- College of Pharmacy, Touro University California, 1310 Club Drive, Mare Island, Vallejo, CA 94592, USA
| |
Collapse
|
6
|
Raj S, Vishwakarma P, Saxena S, Kumar V, Khatri R, Kumar A, Singh M, Mishra S, Asthana S, Ahmed S, Samal S. Intradermal Immunization of Soluble Influenza HA Derived from a Lethal Virus Induces High Magnitude and Breadth of Antibody Responses and Provides Complete Protection In Vivo. Vaccines (Basel) 2023; 11:780. [PMID: 37112692 PMCID: PMC10141624 DOI: 10.3390/vaccines11040780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023] Open
Abstract
Immunogens mimicking the native-like structure of surface-exposed viral antigens are considered promising vaccine candidates. Influenza viruses are important zoonotic respiratory viruses with high pandemic potential. Recombinant soluble hemagglutinin (HA) glycoprotein-based protein subunit vaccines against Influenza have been shown to induce protective efficacy when administered intramuscularly. Here, we have expressed a recombinant soluble trimeric HA protein in Expi 293F cells and purified the protein derived from the Inf A/Guangdong-Maonan/ SWL1536/2019 virus which was found to be highly virulent in the mouse. The trimeric HA protein was found to be in the oligomeric state, highly stable, and the efficacy study in the BALB/c mouse challenge model through intradermal immunization with the prime-boost regimen conferred complete protection against a high lethal dose of homologous and mouse-adapted InfA/PR8 virus challenge. Furthermore, the immunogen induced high hemagglutinin inhibition (HI) titers and showed cross-protection against other Inf A and Inf B subtypes. The results are promising and warrant trimeric HA as a suitable vaccine candidate.
Collapse
Affiliation(s)
- Sneha Raj
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Preeti Vishwakarma
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shikha Saxena
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Varun Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Ritika Khatri
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Amit Kumar
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Mrityunjay Singh
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Surbhi Mishra
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shailendra Asthana
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Shubbir Ahmed
- Centralized Core Research Facility (CCRF), All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sweety Samal
- Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| |
Collapse
|
7
|
Luo J, Zhang Z, Zhao S, Gao R. A Comparison of Etiology, Pathogenesis, Vaccinal and Antiviral Drug Development between Influenza and COVID-19. Int J Mol Sci 2023; 24:ijms24076369. [PMID: 37047339 PMCID: PMC10094131 DOI: 10.3390/ijms24076369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Influenza virus and coronavirus, two kinds of pathogens that exist widely in nature, are common emerging pathogens that cause respiratory tract infections in humans. In December 2019, a novel coronavirus SARS-CoV-2 emerged, causing a severe respiratory infection named COVID-19 in humans, and raising a global pandemic which has persisted in the world for almost three years. Influenza virus, a seasonally circulating respiratory pathogen, has caused four global pandemics in humans since 1918 by the emergence of novel variants. Studies have shown that there are certain similarities in transmission mode and pathogenesis between influenza and COVID-19, and vaccination and antiviral drugs are considered to have positive roles as well as several limitations in the prevention and control of both diseases. Comparative understandings would be helpful to the prevention and control of these diseases. Here, we review the study progress in the etiology, pathogenesis, vaccine and antiviral drug development for the two diseases.
Collapse
|
8
|
Langowski MD, Khan FA, Savransky S, Brown DR, Balasubramaniyam A, Harrison WB, Zou X, Beck Z, Matyas GR, Regules JA, Miller R, Soisson LA, Batchelor AH, Dutta S. Restricted valency (NPNA) n repeats and junctional epitope-based circumsporozoite protein vaccines against Plasmodium falciparum. NPJ Vaccines 2022; 7:13. [PMID: 35087099 PMCID: PMC8795123 DOI: 10.1038/s41541-022-00430-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 12/14/2021] [Indexed: 12/02/2022] Open
Abstract
The Circumsporozoite Protein (CSP) of Plasmodium falciparum contains an N-terminal region, a conserved Region I (RI), a junctional region, 25-42 copies of major (NPNA) and minor repeats followed by a C-terminal domain. The recently approved malaria vaccine, RTS,S/AS01 contains NPNAx19 and the C-terminal region of CSP. The efficacy of RTS,S against natural infection is low and short-lived, and mapping epitopes of inhibitory monoclonal antibodies may allow for rational improvement of CSP vaccines. Tobacco Mosaic Virus (TMV) was used here to display the junctional epitope (mAb CIS43), Region I (mAb 5D5), NPNAx5, and NPNAx20 epitope of CSP (mAbs 317 and 580). Protection studies in mice revealed that Region I did not elicit protective antibodies, and polyclonal antibodies against the junctional epitope showed equivalent protection to NPNAx5. Combining the junctional and NPNAx5 epitopes reduced immunogenicity and efficacy, and increasing the repeat valency to NPNAx20 did not improve upon NPNAx5. TMV was confirmed as a versatile vaccine platform for displaying small epitopes defined by neutralizing mAbs. We show that polyclonal antibodies against engineered VLPs can recapitulate the binding specificity of the mAbs and immune-focusing by reducing the structural complexity of an epitope may be superior to immune-broadening as a vaccine design approach. Most importantly the junctional and restricted valency NPNA epitopes can be the basis for developing highly effective second-generation malaria vaccine candidates.
Collapse
Affiliation(s)
- Mark D Langowski
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Farhat A Khan
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sofya Savransky
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Dallas R Brown
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Arasu Balasubramaniyam
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - William B Harrison
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Xiaoyan Zou
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Zoltan Beck
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Pfizer, 401N Middletown Rd, Pearl River, NY, 10965, USA
| | - Gary R Matyas
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jason A Regules
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Robin Miller
- United States Agency for International Development, Washington, DC, USA
| | | | - Adrian H Batchelor
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sheetij Dutta
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
9
|
Oluwagbemi OO, Oladipo EK, Dairo EO, Ayeni AE, Irewolede BA, Jimah EM, Oyewole MP, Olawale BM, Adegoke HM, Ogunleye AJ. Computational construction of a glycoprotein multi-epitope subunit vaccine candidate for old and new South-African SARS-CoV-2 virus strains. INFORMATICS IN MEDICINE UNLOCKED 2022; 28:100845. [PMID: 35071728 PMCID: PMC8760845 DOI: 10.1016/j.imu.2022.100845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 12/19/2022] Open
Abstract
The discovery of a new SARS-CoV-2 virus strain in South Africa presents a major public health threat, therefore contributing to increased infections and transmission rates during the second wave of the global pandemic. This study lays the groundwork for the development of a novel subunit vaccine candidate from the circulating strains of South African SARS-CoV-2 and provides an understanding of the molecular epidemiological trend of the circulating strains. A total of 475 whole-genome nucleotide sequences from South Africa submitted between December 1, 2020 and February 15, 2021 available at the GISAID database were retrieved based on its size, coverage level and hosts. To obtain the distribution of the clades and lineages of South African SARS-CoV-2 circulating strains, the metadata of the sequence retrieved were subjected to an epidemiological analysis. There was a prediction of the cytotoxic T lymphocytes (CTL), Helper T cells (HTL) and B-cell epitopes. Furthermore, there was allergenicity, antigenicity and toxicity predictions on the epitopes. The analysis of the physicochemical properties of the vaccine construct was performed; the secondary structure, tertiary structure and B-cell 3D conformational structure of the vaccine construct were predicted. Also, molecular binding simulations and dynamics simulations were adopted in the prediction of the vaccine construct's stability and binding affinity with TLRs. Result obtained from the metadata analysis indicated lineage B.1.351 to be in higher circulation among various circulating strains of SARS-CoV-2 in South Africa and GH has the highest number of circulating clades. The construct of the novel vaccine was antigenic, non-allergenic and non-toxic. The Instability index (II) score and aliphatic index were estimated as 41.74 and 78.72 respectively. The computed half-life in mammalian reticulocytes was 4.4 h in vitro, for yeast and in E. coli was >20 h and >10 h in vivo respectively. The grand average of hydropathicity (GRAVY) score is estimated to be -0.129, signifying the hydrophilic nature of the protein. The molecular docking indicates that the vaccine construct has a high binding affinity towards the TLRs with TLR 3 having the highest binding energy (-1203.2 kcal/mol) and TLR 9 with the lowest (-1559.5 kcal/mol). These results show that the vaccine construct is promising and should be evaluated using animal model.
Collapse
Affiliation(s)
- Olugbenga Oluseun Oluwagbemi
- Department of Computer Science and Information Technology, Sol Plaatje University, 8301, Kimberley, South Africa
- Department of Mathematical Sciences, Stellenbosch University, 7602, Matieland, South Africa
- National Institute of Theoretical and Computational Sciences (NiTheCS), South Africa
| | - Elijah Kolawole Oladipo
- Department of Microbiology, Laboratory of Molecular Biology, Immunology and Bioinformatics, Adeleke University, Ede, Osun State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Emmanuel Oluwatobi Dairo
- Department of Virology, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Ayodele Eugene Ayeni
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
- Department of Medical Microbiology and Parasitology, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo State, Nigeria
| | | | - Esther Moradeyo Jimah
- Department of Medical Microbiology and Parasitology, University of Ilorin, Kwara State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Moyosoluwa Precious Oyewole
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Boluwatife Mary Olawale
- Reproduction and Bioinformatics Unit, Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Genomics Unit, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | | | - Adewale Joseph Ogunleye
- Moscow Institute of Physics and Technology, 141701, Dolgoprudny, Moscow Oblast, Russian Federation
| |
Collapse
|
10
|
Development of a SARS-CoV-2 Vaccine Candidate Using Plant-Based Manufacturing and a Tobacco Mosaic Virus-like Nano-Particle. Vaccines (Basel) 2021; 9:vaccines9111347. [PMID: 34835278 PMCID: PMC8619098 DOI: 10.3390/vaccines9111347] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/19/2022] Open
Abstract
Stable, effective, easy-to-manufacture vaccines are critical to stopping the COVID-19 pandemic resulting from the coronavirus SARS-CoV-2. We constructed a vaccine candidate CoV-RBD121-NP, which is comprised of the SARS-CoV-2 receptor-binding domain (RBD) of the spike glycoprotein (S) fused to a human IgG1 Fc domain (CoV-RBD121) and conjugated to a modified tobacco mosaic virus (TMV) nanoparticle. In vitro, CoV-RBD121 bound to the host virus receptor ACE2 and to the monoclonal antibody CR3022, a neutralizing antibody that blocks S binding to ACE2. The CoV-RBD121-NP vaccine candidate retained key SARS-CoV-2 spike protein epitopes, had consistent manufacturing release properties of safety, identity, and strength, and displayed stable potency when stored for 12 months at 2–8 °C or 22–28 °C. Immunogenicity studies revealed strong antibody responses in C57BL/6 mice with non-adjuvanted or adjuvanted (7909 CpG) formulations. The non-adjuvanted vaccine induced a balanced Th1/Th2 response and antibodies that recognized both the S1 domain and full S protein from SARS2-CoV-2, whereas the adjuvanted vaccine induced a Th1-biased response. Both adjuvanted and non-adjuvanted vaccines induced virus neutralizing titers as measured by three different assays. Collectively, these data showed the production of a stable candidate vaccine for COVID-19 through the association of the SARS-CoV-2 RBD with the TMV-like nanoparticle.
Collapse
|
11
|
Producing Vaccines against Enveloped Viruses in Plants: Making the Impossible, Difficult. Vaccines (Basel) 2021; 9:vaccines9070780. [PMID: 34358196 PMCID: PMC8310165 DOI: 10.3390/vaccines9070780] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
The past 30 years have seen the growth of plant molecular farming as an approach to the production of recombinant proteins for pharmaceutical and biotechnological uses. Much of this effort has focused on producing vaccine candidates against viral diseases, including those caused by enveloped viruses. These represent a particular challenge given the difficulties associated with expressing and purifying membrane-bound proteins and achieving correct assembly. Despite this, there have been notable successes both from a biochemical and a clinical perspective, with a number of clinical trials showing great promise. This review will explore the history and current status of plant-produced vaccine candidates against enveloped viruses to date, with a particular focus on virus-like particles (VLPs), which mimic authentic virus structures but do not contain infectious genetic material.
Collapse
|
12
|
Venkataraman S, Hefferon K, Makhzoum A, Abouhaidar M. Combating Human Viral Diseases: Will Plant-Based Vaccines Be the Answer? Vaccines (Basel) 2021; 9:vaccines9070761. [PMID: 34358177 PMCID: PMC8310141 DOI: 10.3390/vaccines9070761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 12/28/2022] Open
Abstract
Molecular pharming or the technology of application of plants and plant cell culture to manufacture high-value recombinant proteins has progressed a long way over the last three decades. Whether generated in transgenic plants by stable expression or in plant virus-based transient expression systems, biopharmaceuticals have been produced to combat several human viral diseases that have impacted the world in pandemic proportions. Plants have been variously employed in expressing a host of viral antigens as well as monoclonal antibodies. Many of these biopharmaceuticals have shown great promise in animal models and several of them have performed successfully in clinical trials. The current review elaborates the strategies and successes achieved in generating plant-derived vaccines to target several virus-induced health concerns including highly communicable infectious viral diseases. Importantly, plant-made biopharmaceuticals against hepatitis B virus (HBV), hepatitis C virus (HCV), the cancer-causing virus human papillomavirus (HPV), human immunodeficiency virus (HIV), influenza virus, zika virus, and the emerging respiratory virus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have been discussed. The use of plant virus-derived nanoparticles (VNPs) and virus-like particles (VLPs) in generating plant-based vaccines are extensively addressed. The review closes with a critical look at the caveats of plant-based molecular pharming and future prospects towards further advancements in this technology. The use of biopharmed viral vaccines in human medicine and as part of emergency response vaccines and therapeutics in humans looks promising for the near future.
Collapse
Affiliation(s)
- Srividhya Venkataraman
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
- Correspondence:
| | - Kathleen Hefferon
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| | - Abdullah Makhzoum
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Palapye, Botswana;
| | - Mounir Abouhaidar
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| |
Collapse
|
13
|
Butkovich N, Li E, Ramirez A, Burkhardt AM, Wang SW. Advancements in protein nanoparticle vaccine platforms to combat infectious disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1681. [PMID: 33164326 PMCID: PMC8052270 DOI: 10.1002/wnan.1681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
Infectious diseases are a major threat to global human health, yet prophylactic treatment options can be limited, as safe and efficacious vaccines exist only for a fraction of all diseases. Notably, devastating diseases such as acquired immunodeficiency syndrome (AIDS) and coronavirus disease of 2019 (COVID-19) currently do not have vaccine therapies. Conventional vaccine platforms, such as live attenuated vaccines and whole inactivated vaccines, can be difficult to manufacture, may cause severe side effects, and can potentially induce severe infection. Subunit vaccines carry far fewer safety concerns due to their inability to cause vaccine-based infections. The applicability of protein nanoparticles (NPs) as vaccine scaffolds is promising to prevent infectious diseases, and they have been explored for a number of viral, bacterial, fungal, and parasitic diseases. Many types of protein NPs exist, including self-assembling NPs, bacteriophage-derived NPs, plant virus-derived NPs, and human virus-based vectors, and these particular categories will be covered in this review. These vaccines can elicit strong humoral and cellular immune responses against specific pathogens, as well as provide protection against infection in a number of animal models. Furthermore, published clinical trials demonstrate the promise of applying these NP vaccine platforms, which include bacteriophage-derived NPs, in addition to multiple viral vectors that are currently used in the clinic. The continued investigations of protein NP vaccine platforms are critical to generate safer alternatives to current vaccines, advance vaccines for diseases that currently lack effective prophylactic therapies, and prepare for the rapid development of new vaccines against emerging infectious diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Nina Butkovich
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Enya Li
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Aaron Ramirez
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Amanda M. Burkhardt
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089 USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697 USA
| |
Collapse
|
14
|
D'Arco C, McCormick AA, Arnaboldi PM. Single-dose intranasal subunit vaccine rapidly clears secondary sepsis in a high-dose pneumonic plague infection. Vaccine 2021; 39:1435-1444. [PMID: 33531196 DOI: 10.1016/j.vaccine.2021.01.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023]
Abstract
Yersinia pestis, the causative agent of plague, has killed millions throughout human history. Though public health initiatives have reduced the number of plague cases, it remains endemic in many areas of the world. It also remains a significant threat for use as a biological weapon. Naturally occurring multi-drug antibiotic resistance has been observed in Y. pestis, and resistant strains have been engineered for use as a biological weapon. Vaccines represent our best means of protection against the threat of antibiotic resistant plague. We have developed a vaccine consisting of two Y. pestis virulence factors, LcrV (V) and F1, conjugated to Tobacco Mosaic Virus (TMV), a safe, non-replicating plant virus that can be administered mucosally, providing complete protection against pneumonic plague, the deadliest form of the disease and the one most likely to be seen in a biological attack. A single intranasal (i.n.) dose of TMV-F1 + TMV-V (TMV-F1/V) protected 88% of mice against lethal challenge with 100 LD50 of Y. pestis CO92pgm-, while immunization with rF1 + rV without TMV was not protective. Serum and tissues were collected at various timepoints after challenge to assess bacterial clearance, histopathology, cytokine production, and antibody production. Overall, TMV-F1/V immunized mice showed a significant reduction in histopathology, bacterial burden, and inflammatory cytokine production following challenge compared to rF1 + rV vaccinated and unvaccinated mice. Pneumonic challenge resulted in systemic dissemination of the bacteria in all groups, but only TMV-F1/V immunized mice rapidly cleared bacteria from the spleen and liver. There was a direct correlation between pre-challenge serum F1 titers and recovery in all immunized mice, strongly suggesting a role for antibody in the neutralization and/or opsonization of Y. pestis in this model. Mucosal administration of a single dose of a Y. pestis TMV-based subunit vaccine, without any additional adjuvant, can effectively protect mice from lethal infection.
Collapse
Affiliation(s)
- Christina D'Arco
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, United States
| | - Alison A McCormick
- Department of Biology and Pharmaceutical Sciences, College of Pharmacy, Touro University California, Vallejo, CA 94592, United States
| | - Paul M Arnaboldi
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, United States; Biopeptides, Corp., East Setauket, NY 11733, United States.
| |
Collapse
|
15
|
Evtushenko EA, Ryabchevskaya EM, Nikitin NA, Atabekov JG, Karpova OV. Plant virus particles with various shapes as potential adjuvants. Sci Rep 2020; 10:10365. [PMID: 32587281 PMCID: PMC7316779 DOI: 10.1038/s41598-020-67023-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 06/01/2020] [Indexed: 01/15/2023] Open
Abstract
Plant viruses are biologically safe for mammals and can be successfully used as a carrier/platform to present foreign epitopes in the course of creating novel putative vaccines. However, there is mounting evidence that plant viruses, their virus-like and structurally modified particles may also have an immunopotentiating effect on antigens not bound with their surface covalently. Here, we present data on the adjuvant properties of plant viruses with various shapes (Tobacco mosaic virus, TMV; Potato virus X, PVX; Cauliflower mosaic virus, CaMV; Bean mild mosaic virus, BMMV) and structurally modified TMV spherical particles (SPs). We have analysed the effectiveness of immune response to individual model antigens (ovalbumin, OVA/hen egg lysozyme, HEL) and to OVA/HEL in compositions with plant viruses/SPs, and have shown that CaMV, TMV and SPs can effectively induce total IgG titers to model antigen. Some intriguing data were obtained when analysing the immune response to the plant viruses/SPs themselves. Strong immunity was induced to CaMV, BMMV and PVX, whereas TMV and SPs stimulated considerably lower self-IgG titers. Our results provide new insights into the immunopotentiating properties of plant viruses and can be useful in devising adjuvants based on plant viruses.
Collapse
Affiliation(s)
- Ekaterina A Evtushenko
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119234, Russian Federation.
| | - Ekaterina M Ryabchevskaya
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119234, Russian Federation
| | - Nikolai A Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119234, Russian Federation
| | - Joseph G Atabekov
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119234, Russian Federation
| | - Olga V Karpova
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory, Moscow, 119234, Russian Federation
| |
Collapse
|
16
|
Kumar S, Sunagar R, Gosselin EJ. Preclinical Efficacy of a Trivalent Human FcγRI-Targeted Adjuvant-Free Subunit Mucosal Vaccine against Pulmonary Pneumococcal Infection. Vaccines (Basel) 2020; 8:vaccines8020193. [PMID: 32340134 PMCID: PMC7349865 DOI: 10.3390/vaccines8020193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/03/2020] [Accepted: 04/17/2020] [Indexed: 12/25/2022] Open
Abstract
Lack of safe and effective mucosal adjuvants has severely hampered the development of mucosal subunit vaccines. In this regard, we have previously shown that immunogenicity of vaccine antigens can be improved by targeting the antigens to the antigen-presenting cells. Specifically, groups of mice immunized intranasally with a fusion protein (Bivalent-FP) containing a fragment of pneumococcal-surface-protein-A (PspA) as antigen and a single-chain bivalent antibody raised against the anti-human Fc-gamma-receptor-I (hFcγRI) elicited protective immunity to pulmonary Streptococcus pneumoniae infection. In order to further enhance the immunogenicity, an additional hFcγRI-binding moiety of the single chain antibody was incorporated. The modified vaccine (Trivalent-FP) induced significantly improved protection against lethal pulmonary S. pneumoniae challenge compared to Bivalent-FP. In addition, the modified vaccine exhibited over 85% protection with only two immunizations. Trivalent-FP also induced S. pneumoniae-specific systemic and mucosal antibodies. Moreover, Trivalent-FP also induced IL-17- and IL-22-producing CD4+ T cells. Furthermore, it was found that the hFcγRI facilitated uptake and presentation of Trivalent-FP. In addition, Trivalent-FP also induced IL-1α, MIP-1α, and TNF-α; modulated recruitment of dendritic cells and macrophages; and induced CD80/86 and MHC-II expression on antigen presenting cells.
Collapse
Affiliation(s)
- Sudeep Kumar
- Department of Immunology and Microbial Diseases, Albany Medical College, Albany, NY 12208, USA;
| | - Raju Sunagar
- Ella Foundation, Genome Valley, Hyderabad 500078, India;
| | - Edmund J. Gosselin
- Department of Immunology and Microbial Diseases, Albany Medical College, Albany, NY 12208, USA;
- Correspondence:
| |
Collapse
|
17
|
Sharma J, Shepardson K, Johns LL, Wellham J, Avera J, Schwarz B, Rynda-Apple A, Douglas T. A Self-Adjuvanted, Modular, Antigenic VLP for Rapid Response to Influenza Virus Variability. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18211-18224. [PMID: 32233444 DOI: 10.1021/acsami.9b21776] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The continuous evolution of influenza A virus (IAV) requires the influenza vaccine formulations to be updated annually to provide adequate protection. Recombinant protein-based vaccines provide safer, faster, and a more scalable alternative to the conventional embryonated egg approach for developing vaccines. However, these vaccines are typically poorer in immunogenicity than the vaccines containing inactivated or attenuated influenza viruses and require administration of a large antigen dosage together with potent adjuvants. The presentation of protein antigens on the surface of virus-like particles (VLP) provides an attractive strategy to rapidly induce stronger antigen-specific immune responses. Here we have examined the immunogenic potential and protective efficacy of P22 VLPs conjugated with multiple copies of the globular head domain of the hemagglutinin (HA) protein from the PR8 strain of IAV in a murine model of influenza pathogenesis. Using a covalent attachment strategy (SpyTag/SpyCatcher), we conjugated the HA globular head, which was recombinantly expressed in a genetically modified E. coli strain and found to refold as a monomer, to preassembled P22 VLPs. Immunization of mice with this P22-HAhead conjugate provided full protection from morbidity and mortality following infection with a homologous IAV strain. Moreover, the P22-HAhead conjugate also elicited an accelerated and enhanced HA head specific IgG response, which was significantly higher than the soluble HA head, or the admixture of P22 and HA head without the need for adjuvants. Thus, our results show that the HA head can be easily prepared by in vitro refolding in a modified E. coli strain, maintaining its intact structure and enabling the induction of a strong immune response when conjugated to P22 VLPs, even when presented as a monomer. These results also demonstrate that the P22 VLPs can be rapidly modified in a modular fashion, resulting in an effective vaccine construct that can generate protective immunity without the need for additional adjuvants.
Collapse
Affiliation(s)
- Jhanvi Sharma
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Kelly Shepardson
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Laura L Johns
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Julia Wellham
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - John Avera
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
- Matrivax Research and Development Corporation, Boston, Massachusetts 02118, United Sates
| | - Benjamin Schwarz
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
- Immunity to Pulmonary Pathogens section, Laboratory of Bacteriology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana 59840, United States
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Trevor Douglas
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
18
|
Eapen P, Cates J, Mundell R, Palmer KE, Fuqua JL. In Preparation for Outdoor Pharming: Griffithsin Can Be Expressed in Nicotiana excelsiana and Retains Activity After Storage as Silage. Front Bioeng Biotechnol 2020; 8:199. [PMID: 32258012 PMCID: PMC7093594 DOI: 10.3389/fbioe.2020.00199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/28/2020] [Indexed: 11/13/2022] Open
Abstract
Griffithsin is an algae-derived lectin with strong anti-viral activity against HIV and a positive safety profile. Multiple clinical studies are investigating griffithsin's utility as topical HIV microbicide. HIV microbicides are an extremely cost-sensitive market and plant-based griffithsin protein expression has the potential to meet those demands. The griffithsin product used in the clinic has been expressed and purified in N. benthamiana, using a TMV-based viral vector system, Geneware®. Outdoor pharming of biopharmaceuticals would further alleviate startup costs for biotechnology firms and may allow broader product accessibility. Therefore, this study assessed expression in a hybrid tobacco line, N. excelsiana, that is susceptible to TMV-based viral vectors and can be grown outdoors. In addition to using this hybrid line we expand on methods for in planta storage of griffithsin in leafy plants by ensiling kilogram quantities of griffithsin. The ensiling process allows year-round biomanufacturing, minimal environmental-controlled storage, and reduces the industry need for multiple growth areas to maintain multi-product manufacturing of plant-based pharmaceuticals. This study shows that griffithsin can be expressed in N. excelsiana and is stable, recoverable, and active from ensiled tissue. These studies can pave the way for future plant-based pharmaceuticals to be expressed and stored in this manner.
Collapse
Affiliation(s)
- Paul Eapen
- University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States
| | - Jennifer Cates
- University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States
| | - Rich Mundell
- University of Kentucky College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, United States
| | - Kenneth E Palmer
- University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States.,Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States
| | - Joshua L Fuqua
- University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States.,Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, University of Louisville, Louisville, KY, United States
| |
Collapse
|
19
|
Gasanova TV, Koroleva AA, Skurat EV, Ivanov PA. Complexes Formed via Bioconjugation of Genetically Modified TMV Particles with Conserved Influenza Antigen: Synthesis and Characterization. BIOCHEMISTRY. BIOKHIMIIA 2020; 85:224-233. [PMID: 32093598 DOI: 10.1134/s0006297920020091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/08/2019] [Accepted: 11/16/2019] [Indexed: 06/10/2023]
Abstract
Recently we obtained complexes between genetically modified Tobacco Mosaic Virus (TMV) particles and proteins carrying conserved influenza antigen such as M2e epitope. Viral vector TMV-N-lys based on TMV-U1 genome was constructed by insertion of chemically active lysine into the exposed N-terminal part of the coat protein. Nicotiana benthamiana plants were agroinjected and TMV-N-lys virions were purified from non-inoculated leaves. Preparation was analyzed by SDS-PAGE/Coomassie staining; main protein with electrophoretic mobility of 21 kDa was detected. Electron microscopy confirmed the stability of modified particles. Chemical conjugation of TMV-N-lys virions and target influenza antigen M2e expressed in E. coli was performed using 5 mM 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide and 1 mM N-hydroxysuccinimide. The efficiency of chemical conjugation was confirmed by Western blotting. For additional characterization we used conventional electron microscopy. The diameter of the complexes did not differ significantly from the initial TMV-N-lys virions, but complexes formed highly organized and extensive network with dense "grains" on the surface. Dynamic light scattering demonstrated that the single peaks, reflecting the complexes TMV-N-lys/DHFR-M2e were significantly shifted relative to the control TMV-N-lys virions. The indirect enzyme-linked immunosorbent assay with TMV- and DHFR-M2e-specific antibodies showed that the complexes retain stability during overnight adsorption. Thus, the results allow using these complexes for immunization of animals with the subsequent preparation of a candidate universal vaccine against the influenza virus.
Collapse
Affiliation(s)
- T V Gasanova
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia.
| | - A A Koroleva
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - E V Skurat
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| | - P A Ivanov
- Lomonosov Moscow State University, Faculty of Biology, Moscow, 119991, Russia
| |
Collapse
|
20
|
Venkataraman S, Reddy VS, Khurana SMP. Biomedical Applications of Viral Nanoparticles in Vaccine Therapy. Nanobiomedicine (Rij) 2020. [DOI: 10.1007/978-981-32-9898-9_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
21
|
Tsekoa TL, Singh AA, Buthelezi SG. Molecular farming for therapies and vaccines in Africa. Curr Opin Biotechnol 2019; 61:89-95. [PMID: 31786432 DOI: 10.1016/j.copbio.2019.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
Local manufacturing of protein-based vaccines and therapies in Africa is limited and contributes to a trade deficit, security of supply concerns and poor access to biopharmaceuticals by the poor. Plant molecular farming is a potential technology solution that has received growing adoption by African scientists attracted by the potential for the competitive cost of goods, safety and efficacy. Plant-made pharmaceutical technologies for veterinary and human vaccination and treatment of non-communicable and infectious diseases are available at different stages of development in Africa. There is also growth in the translation of these technologies to commercial operations. Africa is poised to benefit from the real-world impact of molecular farming in the next few years.
Collapse
Affiliation(s)
- Tsepo L Tsekoa
- NextGen Health and Future Production: Chemistry Clusters, Council for Scientific and Industrial Research, Pretoria, South Africa.
| | - Advaita Acarya Singh
- NextGen Health and Future Production: Chemistry Clusters, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Sindisiwe G Buthelezi
- NextGen Health and Future Production: Chemistry Clusters, Council for Scientific and Industrial Research, Pretoria, South Africa
| |
Collapse
|
22
|
Gebauer M, Hürlimann HC, Behrens M, Wolff T, Behrens SE. Subunit vaccines based on recombinant yeast protect against influenza A virus in a one-shot vaccination scheme. Vaccine 2019; 37:5578-5587. [PMID: 31399274 DOI: 10.1016/j.vaccine.2019.07.094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/22/2019] [Accepted: 07/27/2019] [Indexed: 01/03/2023]
Abstract
Here we report on new subunit vaccines based on recombinant yeast of the type Kluyveromyces lactis (K. lactis), which protect mice from a lethal influenza A virus infection. Applying a genetic system that enables the rapid generation of transgenic yeast, we have developed K. lactis strains that express the influenza A virus hemagglutinin, HA, either individually or in combination with the viral M1 matrix protein. Subcutaneous application of the inactivated, but otherwise non-processed yeast material shows a complete protection of BALB/c mice in prime/boost and even one-shot/single dose vaccination schemes against a subsequent, lethal challenge with the cognate influenza virus. The yeast vaccines induce titers of neutralizing antibodies that are readily comparable to those induced by an inactivated virus vaccine. These data suggest that HA and M1 are produced with a high antigenicity in the yeast cells. Based on these findings, multivalent, DIVA-capable, yeast-based subunit vaccines may be developed as promising alternatives to conventional virus-based anti-flu vaccines for veterinary applications.
Collapse
Affiliation(s)
- Mandy Gebauer
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Hans C Hürlimann
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biology, Weinbergweg 10, 06120 Halle (Saale), Germany
| | - Martina Behrens
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Thorsten Wolff
- Robert Koch Institute, Unit 17 "Influenza and Other Respiratory Viruses", Seestr. 10, 13353 Berlin, Germany
| | - Sven-Erik Behrens
- Martin Luther University Halle-Wittenberg, Faculty of Life Sciences (NFI), Institute of Biochemistry and Biotechnology, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany.
| |
Collapse
|
23
|
Aksular M, Calvo-Pinilla E, Marín-López A, Ortego J, Chambers AC, King LA, Castillo-Olivares J. A single dose of African horse sickness virus (AHSV) VP2 based vaccines provides complete clinical protection in a mouse model. Vaccine 2018; 36:7003-7010. [PMID: 30309744 PMCID: PMC6219453 DOI: 10.1016/j.vaccine.2018.09.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/06/2018] [Accepted: 09/27/2018] [Indexed: 11/24/2022]
Abstract
Baculovirus-expressed AHS-VP2 and MVA-VP2 vaccines were evaluated in mice. Clinical protection was complete in mice receiving one or two doses of MVA-VP2. Clinical protection complete after two doses of baculovirus-expressed VP2. Significant reduction of viraemia in all vaccinated groups. Significant levels of immunity were achieved with one dose of either vaccine.
African horse sickness is a severe, often fatal, arboviral disease of equids. The control of African horse sickness virus (AHSV) in endemic countries is based currently on the use of live attenuated vaccines despite some biosafety concerns derived from its biological properties. Thus, experimental vaccination platforms have been developed over the years in order to avoid the biosafety concerns associated with the use of attenuated vaccines. Various studies showed that baculovirus-expressed AHSV-VP2 or modified Vaccinia Ankara virus expressing AHSV-VP2 (MVA-VP2) induced virus neutralising antibodies and protective immunity in small animals and horses. AHSV is an antigenically diverse pathogen and immunity against AHS is serotype-specific. Therefore, AHS vaccines for use in endemic countries need to induce an immune response capable of protecting against all existing serotypes. For this reason, current live attenuated vaccines are administered as polyvalent preparations comprising combinations of AHSV attenuated strains of different serotypes. Previous studies have shown that it is possible to induce cross-reactive virus neutralising antibodies against different serotypes of AHSV by using polyvalent vaccines comprising combinations of either different serotype-specific VP2 proteins, or MVA-VP2 viruses. However, these strategies could be difficult to implement if induction of protective immunity is highly dependent on using a two-dose vaccination regime for each serotype the vaccine intends to protect against. In our study, we have tested the protective capacity of MVA-VP2 and baculovirus-expressed VP2 vaccines when a single dose was used. Groups of interferon alpha receptor knock-out mice were inoculated with either MVA-VP2 or baculovirus-expressed VP2 vaccines using one dose or the standard two-dose vaccination regime. After vaccination, all four vaccinated groups were challenged with AHSV and clinical responses, lethality and viraemia compared between the groups. Our results show that complete clinical protection was achieved after a single vaccination with either MVA-VP2 or baculovirus sub-unit VP2 vaccines.
Collapse
Affiliation(s)
- Mine Aksular
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; Oxford Expression Technologies Ltd, Gipsy Lane, Oxford OX3 0BP, UK; The Pirbright Institute, Ash Road, Woking, Surrey GU24 0NB, UK
| | | | | | | | - Adam C Chambers
- Oxford Expression Technologies Ltd, Gipsy Lane, Oxford OX3 0BP, UK
| | - Linda A King
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| | - Javier Castillo-Olivares
- Department of Biological & Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK; The Pirbright Institute, Ash Road, Woking, Surrey GU24 0NB, UK; Department of Veterinary Medicine, University of Cambridge, Madingley Road, CB3 0ES Cambridge, UK.
| |
Collapse
|
24
|
Mansour AA, Banik S, Suresh RV, Kaur H, Malik M, McCormick AA, Bakshi CS. An Improved Tobacco Mosaic Virus (TMV)-Conjugated Multiantigen Subunit Vaccine Against Respiratory Tularemia. Front Microbiol 2018; 9:1195. [PMID: 29922267 PMCID: PMC5996085 DOI: 10.3389/fmicb.2018.01195] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 05/16/2018] [Indexed: 12/22/2022] Open
Abstract
Francisella tularensis, the causative agent of the fatal human disease known as tularemia is classified as a Category A Select Agent by the Centers for Disease Control. No licensed vaccine is currently available for prevention of tularemia in the United States. Previously, we published that a tri-antigen tobacco mosaic virus (TMV) vaccine confers 50% protection in immunized mice against respiratory tularemia caused by F. tularensis. In this study, we refined the TMV-vaccine formulation to improve the level of protection in immunized C57BL/6 mice against respiratory tularemia. We developed a tetra-antigen vaccine by conjugating OmpA, DnaK, Tul4, and SucB proteins of Francisella to TMV. CpG was also included in the vaccine formulation as an adjuvant. Primary intranasal (i.n.) immunization followed by two booster immunizations with the tetra-antigen TMV vaccine protected 100% mice against i.n. 10LD100 challenges dose of F. tularensis live vaccine strain (LVS). Mice receiving three immunization doses of tetra-antigen TMV vaccine showed only transient body weight loss, cleared the infection rapidly, and showed minimal histopathological lesions in lungs, liver, and spleen following a lethal respiratory challenge with F. tularensis LVS. Mice immunized with the tetra-antigen TMV vaccine also induced strong ex vivo recall responses and were protected against a lethal challenge as late as 163 days post-primary immunization. Three immunization with the tetra-antigen TMV vaccine also induced a stronger humoral immune response predominated by IgG1, IgG2b, and IgG2c antibodies than mice receiving only a single or two immunizations. Remarkably, a single dose protected 40% of mice, while two doses protected 80% of mice from lethal pathogen challenge. Immunization of Interferon-gamma (IFN-γ)-deficient mice with the tetra-antigen TMV vaccine demonstrated an absolute requirement of IFN-γ for the generation of protective immune response against a lethal respiratory challenge with F. tularensis LVS. Collectively, this study further demonstrates the feasibility of TMV as an efficient platform for the delivery of multiple F. tularensis antigens and that tetra-antigen TMV vaccine formulation provides complete protection, and induces long-lasting protective and memory immune responses against respiratory tularemia caused by F. tularensis LVS.
Collapse
Affiliation(s)
- Ahd A Mansour
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Sukalyani Banik
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Ragavan V Suresh
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Hardeep Kaur
- College of Pharmacy, Touro University California, Vallejo, CA, United States
| | - Meenakshi Malik
- Department of Basic and Clinical Sciences, School of Arts and Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Alison A McCormick
- College of Pharmacy, Touro University California, Vallejo, CA, United States
| | - Chandra S Bakshi
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
25
|
McCormick AA, Shakeel A, Yi C, Kaur H, Mansour AM, Bakshi CS. Intranasal administration of a two-dose adjuvanted multi-antigen TMV-subunit conjugate vaccine fully protects mice against Francisella tularensis LVS challenge. PLoS One 2018; 13:e0194614. [PMID: 29684046 PMCID: PMC5912714 DOI: 10.1371/journal.pone.0194614] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/06/2018] [Indexed: 12/03/2022] Open
Abstract
Tularemia is a fatal human disease caused by Francisella tularensis, a Gram-negative encapsulated coccobacillus bacterium. Due to its low infectious dose, ease of aerosolized transmission, and lethal effects, the CDC lists F. tularensis as a Category A pathogen, the highest level for a potential biothreat agent. Previous vaccine studies have been conducted with live attenuated, inactivated, and subunit vaccines, which have achieved partial or full protection from F. tularensis live vaccine strain (LVS) challenge, but no vaccine has been approved for human use. We demonstrate the improved efficacy of a multi-antigen subunit vaccine by using Tobacco Mosaic virus (TMV) as an antigen carrier for the F. tularensis SchuS4 proteins DnaK, OmpA, SucB and Tul4 (DOST). The magnitude and quality of immune responses were compared after mice were immunized by subcutaneous or intranasal routes of administration with a TMV-DOST mixture, with or without four different adjuvants. Immune responses varied in magnitude and isotype profile, by antigen, by route of administration, and by protection in an F. tularensis LVS challenge model of disease. Interestingly, our analysis demonstrates an overwhelming IgG2 response to SucB after intranasal dosing, as well as a robust cellular response, which may account for the improved two-dose survival imparted by the tetravalent vaccine, compared to a previous study that tested efficacy of TMV-DOT. Our study provides evidence that potent humoral, cellular and mucosal immunity can be achieved by optimal antigen combination, delivery, adjuvant and appropriate route of administration, to improve vaccine potency and provide protection from pathogen challenge.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/analysis
- Antibodies, Bacterial/metabolism
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/metabolism
- Bacterial Vaccines/immunology
- Disease Models, Animal
- Female
- Francisella tularensis/immunology
- Immunity, Cellular
- Immunoglobulin G/analysis
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Immunoglobulin Isotypes/immunology
- Immunoglobulin Isotypes/metabolism
- Mice
- Mice, Inbred C57BL
- Survival Rate
- Tobacco Mosaic Virus/genetics
- Tobacco Mosaic Virus/metabolism
- Tularemia/immunology
- Tularemia/microbiology
- Tularemia/prevention & control
- Vaccines, Conjugate/immunology
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
| | - Aisha Shakeel
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Chris Yi
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Hardeep Kaur
- Touro University California, College of Pharmacy, Vallejo, CA
| | - Ahd M. Mansour
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
26
|
Lebel MÈ, Langlois MP, Daudelin JF, Tarrab E, Savard P, Leclerc D, Lamarre A. Complement Component 3 Regulates IFN-α Production by Plasmacytoid Dendritic Cells following TLR7 Activation by a Plant Virus-like Nanoparticle. THE JOURNAL OF IMMUNOLOGY 2016; 198:292-299. [PMID: 27864474 DOI: 10.4049/jimmunol.1601271] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/31/2016] [Indexed: 12/14/2022]
Abstract
The increasing use of plant viruses for the development of new vaccines and immunotherapy approaches poses questions regarding the mechanism by which the mammalian immune system recognizes these viruses. For example, although natural Abs (NA) and complement are key components of the innate immune system involved in the opsonization, phagocytosis, and destruction of microorganisms infecting mammals, their implication in plant virus recognition and immunogenicity is not well defined. In this study, we address the involvement of NA and the complement system in the activation of innate immunity through engagement of TLR7 with papaya mosaic virus (PapMV)-like nanoparticles. We demonstrate that NA, although binding to PapMV, are not involved in its recognition by the immune system. On the other hand, C3 strongly binds to PapMV nanoparticles and its depletion significantly reduces PapMV's interaction with immune cells. Unexpectedly, however, we observed increased immune cell activation following administration of PapMV to complement-depleted mice. TLR7 activation by PapMV in the absence of C3 induced higher IFN-α production, resulting in superior immune cell activation and increased immunotherapeutic properties. In conclusion, in this study we established the involvement of the complement system in the recognition and the phagocytosis of PapMV nanoparticles and identified an unsuspected role for C3 in regulating the production of IFN-α following TLR7 activation.
Collapse
Affiliation(s)
- Marie-Ève Lebel
- Immunovirology Laboratory, Institut national de la recherche scientifique (INRS), INRS-Institut Armand-Frappier, Laval, Quebec H7V 1B7, Canada
| | - Marie-Pierre Langlois
- Immunovirology Laboratory, Institut national de la recherche scientifique (INRS), INRS-Institut Armand-Frappier, Laval, Quebec H7V 1B7, Canada
| | - Jean-François Daudelin
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H1T 2M4, Canada
| | - Esther Tarrab
- Immunovirology Laboratory, Institut national de la recherche scientifique (INRS), INRS-Institut Armand-Frappier, Laval, Quebec H7V 1B7, Canada
| | - Pierre Savard
- Department of Molecular Biology, Medical Biology and Pathology, Laval University, Quebec City, Quebec G1V 4G2, Canada; and
| | - Denis Leclerc
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Centre, Laval University, Quebec City, Quebec G1V 4G2, Canada
| | - Alain Lamarre
- Immunovirology Laboratory, Institut national de la recherche scientifique (INRS), INRS-Institut Armand-Frappier, Laval, Quebec H7V 1B7, Canada;
| |
Collapse
|
27
|
Intranasal delivery of a protein subunit vaccine using a Tobacco Mosaic Virus platform protects against pneumonic plague. Vaccine 2016; 34:5768-5776. [PMID: 27745954 DOI: 10.1016/j.vaccine.2016.09.063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 11/22/2022]
Abstract
Yersinia pestis, one of history's deadliest pathogens, has killed millions over the course of human history. It has attributes that make it an ideal choice to produce mass casualties and is a prime candidate for use as a biological weapon. When aerosolized, Y. pestis causes pneumonic plague, a pneumonia that is 100% lethal if not promptly treated with effective antibiotics. Currently, there is no FDA approved plague vaccine. The current lead vaccine candidate, a parenterally administered protein subunit vaccine comprised of the Y. pestis virulence factors, F1 and LcrV, demonstrated variable levels of protection in primate pneumonic plague models. As the most likely mode of exposure in biological attack with Y. pestis is by aerosol, this raises a question of whether this parenteral vaccine will adequately protect humans against pneumonic plague. In the present study we evaluated two distinct mucosal delivery platforms for the intranasal (IN) administration of LcrV and F1 vaccine proteins, a live bacterial vector, Lactobacillus plantarum, and a Tobacco Mosaic Virus (TMV) based delivery platform. IN administration of L. plantarum expressing LcrV, or TMV-conjugated to LcrV and F1 (TMV-LcrV+TMV-F1) resulted in the similar induction of high titers of IgG antibodies and evidence of proinflammatory cytokine secretion. However, only the TMV-conjugate delivery platform protected against subsequent lethal challenge with Y. pestis. TMV-LcrV+TMV-F1 co-vaccinated mice had no discernable morbidity and no mortality, while mice vaccinated with L. plantarum expressing LcrV or rLcrV+rF1 without TMV succumbed to infection or were only partially protected. Thus, TMV is a suitable mucosal delivery platform for an F1-LcrV subunit vaccine that induces complete protection against pneumonic infection with a lethal dose of Y. pestis in mice.
Collapse
|
28
|
Gasanova TV, Petukhova NV, Ivanov PA. Chimeric particles of tobacco mosaic virus as a platform for the development of next-generation nanovaccines. ACTA ACUST UNITED AC 2016. [DOI: 10.1134/s1995078016020051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Luckanagul JA, Metavarayuth K, Feng S, Maneesaay P, Clark AY, Yang X, García AJ, Wang Q. Tobacco Mosaic Virus Functionalized Alginate Hydrogel Scaffolds for Bone Regeneration in Rats with Cranial Defect. ACS Biomater Sci Eng 2016; 2:606-615. [DOI: 10.1021/acsbiomaterials.5b00561] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jittima Amie Luckanagul
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
- Department
of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Wangmai, Pathumwan, Bangkok, Thailand 10330
| | - Kamolrat Metavarayuth
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Sheng Feng
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| | - Phudit Maneesaay
- Department
of Pathology, Faculty of Veterinary Medicine, Kasetsart University, 50 Ngamwongwan Road, Lat Yao, Chatuchak, Bangkok, Thailand 10903
| | - Amy Y. Clark
- Woodruff
School of Mechanical Engineering and Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Xiaoming Yang
- Medical
Chronobiology Laboratory and Center for Colon Cancer Research, WJB Dorn VA Medical Center, 6439 Garners Ferry Road, Columbia, South Carolina 29209, United States
| | - Andrés J. García
- Woodruff
School of Mechanical Engineering and Petit Institute for Bioengineering
and Bioscience, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Qian Wang
- Department
of Chemistry and Biochemistry, University of South Carolina, 631
Sumter Street, Columbia, South Carolina 29208, United States
| |
Collapse
|
30
|
Plant Viruses as Nanoparticle-Based Vaccines and Adjuvants. Vaccines (Basel) 2015; 3:620-37. [PMID: 26350598 PMCID: PMC4586470 DOI: 10.3390/vaccines3030620] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022] Open
Abstract
Vaccines are considered one of the greatest medical achievements in the battle against infectious diseases. However, the intractability of various diseases such as hepatitis C, HIV/AIDS, malaria, tuberculosis, and cancer poses persistent hurdles given that traditional vaccine-development methods have proven to be ineffective; as such, these challenges have driven the emergence of novel vaccine design approaches. In this regard, much effort has been put into the development of new safe adjuvants and vaccine platforms. Of particular interest, the utilization of plant virus-like nanoparticles and recombinant plant viruses has gained increasing significance as an effective tool in the development of novel vaccines against infectious diseases and cancer. The present review summarizes recent advances in the use of plant viruses as nanoparticle-based vaccines and adjuvants and their mechanism of action. Harnessing plant-virus immunogenic properties will enable the design of novel, safe, and efficacious prophylactic and therapeutic vaccines against disease.
Collapse
|
31
|
Recombinant H7 hemagglutinin forms subviral particles that protect mice and ferrets from challenge with H7N9 influenza virus. Vaccine 2015. [PMID: 26207590 DOI: 10.1016/j.vaccine.2015.07.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel avian-origin influenza A H7N9 virus emerged in China in 2013 and continues to cause sporadic human infections with mortality rates approaching 35%. Currently there are no approved human vaccines for H7N9 virus. Recombinant approaches including hemagglutinin (HA) and virus-like particles (VLPs) have resulted in experimental vaccines with advantageous safety and manufacturing characteristics. While high immunogenicity of VLP vaccines has been attributed to the native conformation of HA arranged in the regular repeated patterns within virus-like structures, there is limited data regarding molecular organization of HA within recombinant HA vaccine preparations. In this study, the full-length recombinant H7 protein (rH7) of A/Anhui/1/2013 (H7N9) virus was expressed in Sf9 cells. We showed that purified full-length rH7 retained functional ability to agglutinate red blood cells and formed oligomeric pleomorphic subviral particles (SVPs) of ∼20nm in diameter composed of approximately 10 HA0 molecules. No significant quantities of free monomeric HA0 were observed in rH7 preparation by size exclusion chromatography. Immunogenicity and protective efficacy of rH7 SVPs was confirmed in the mouse and ferret challenge models suggesting that SVPs can be used for vaccination against H7N9 virus.
Collapse
|
32
|
Development of a Multivalent Subunit Vaccine against Tularemia Using Tobacco Mosaic Virus (TMV) Based Delivery System. PLoS One 2015; 10:e0130858. [PMID: 26098553 PMCID: PMC4476615 DOI: 10.1371/journal.pone.0130858] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/26/2015] [Indexed: 11/21/2022] Open
Abstract
Francisella tularensis is a facultative intracellular pathogen, and is the causative agent of a fatal human disease known as tularemia. F. tularensis is classified as a Category A Biothreat agent by the CDC based on its use in bioweapon programs by several countries in the past and its potential to be used as an agent of bioterrorism. No licensed vaccine is currently available for prevention of tularemia. In this study, we used a novel approach for development of a multivalent subunit vaccine against tularemia by using an efficient tobacco mosaic virus (TMV) based delivery platform. The multivalent subunit vaccine was formulated to contain a combination of F. tularensis protective antigens: OmpA-like protein (OmpA), chaperone protein DnaK and lipoprotein Tul4 from the highly virulent F. tularensis SchuS4 strain. Two different vaccine formulations and immunization schedules were used. The immunized mice were challenged with lethal (10xLD100) doses of F. tularensis LVS on day 28 of the primary immunization and observed daily for morbidity and mortality. Results from this study demonstrate that TMV can be used as a carrier for effective delivery of multiple F. tularensis antigens. TMV-conjugate vaccine formulations are safe and multiple doses can be administered without causing any adverse reactions in immunized mice. Immunization with TMV-conjugated F. tularensis proteins induced a strong humoral immune response and protected mice against respiratory challenges with very high doses of F. tularensis LVS. This study provides a proof-of-concept that TMV can serve as a suitable platform for simultaneous delivery of multiple protective antigens of F. tularensis. Refinement of vaccine formulations coupled with TMV-targeting strategies developed in this study will provide a platform for development of an effective tularemia subunit vaccine as well as a vaccination approach that may broadly be applicable to many other bacterial pathogens.
Collapse
|
33
|
Zhou Y, Maharaj PD, Mallajosyula JK, McCormick AA, Kearney CM. In planta production of flock house virus transencapsidated RNA and its potential use as a vaccine. Mol Biotechnol 2015; 57:325-36. [PMID: 25432792 DOI: 10.1007/s12033-014-9826-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have developed a transencapsidated vaccine delivery system based on the insect virus, Flock House virus (FHV). FHV is attractive due to its small genome size, simple organization, and nonpathogenic characteristics. With the insertion of a Tobacco mosaic virus (TMV) origin of assembly (Oa), the independently replicating FHV RNA1 can be transencapsidated by TMV coat protein. In this study, we demonstrated that the Oa-adapted FHV RNA1 transencapsidation process can take place in planta, by using a bipartite plant expression vector system, where TMV coat protein is expressed by another plant virus vector, Foxtail mosaic virus (FoMV). Dual infection in the same cell by both FHV and FoMV was observed. Though an apparent classical coat protein-mediated resistance repressed FHV expression, this was overcome by delaying inoculation of the TMV coat protein vector by 3 days after FHV vector inoculation. Expression of the transgene marker in animals by these in vivo-generated transencapsidated nanoparticles was confirmed by mouse vaccination, which also showed an improved vaccine response compared to similar in vitro-produced vaccines.
Collapse
Affiliation(s)
- Yiyang Zhou
- Biomedical Studies Program, Baylor University, Waco, TX, USA,
| | | | | | | | | |
Collapse
|
34
|
Zhang N, Zheng BJ, Lu L, Zhou Y, Jiang S, Du L. Advancements in the development of subunit influenza vaccines. Microbes Infect 2014; 17:123-34. [PMID: 25529753 DOI: 10.1016/j.micinf.2014.12.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/07/2014] [Accepted: 12/08/2014] [Indexed: 12/19/2022]
Abstract
The ongoing threat of influenza epidemics and pandemics has emphasized the importance of developing safe and effective vaccines against infections from divergent influenza viruses. In this review, we first introduce the structure and life cycle of influenza A viruses, describing major influenza A virus-caused pandemics. We then compare different types of influenza vaccines and discuss current advancements in the development of subunit influenza vaccines, particularly those based on nucleoprotein (NP), extracellular domain of matrix protein 2 (M2e) and hemagglutinin (HA) proteins. We also illustrate potential strategies for improving the efficacy of subunit influenza vaccines.
Collapse
Affiliation(s)
- Naru Zhang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Bo-Jian Zheng
- Department of Microbiology, University of Hong Kong, Pokfulam, Hong Kong
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA; Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology, Fudan University, Shanghai, China.
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.
| |
Collapse
|
35
|
Abstract
Plant-made or "biofarmed" viral vaccines are some of the earliest products of the technology of plant molecular farming, and remain some of the brightest prospects for the success of this field. Proofs of principle and of efficacy exist for many candidate viral veterinary vaccines; the use of plant-made viral antigens and of monoclonal antibodies for therapy of animal and even human viral disease is also well established. This review explores some of the more prominent recent advances in the biofarming of viral vaccines and therapies, including the recent use of ZMapp for Ebolavirus infection, and explores some possible future applications of the technology.
Collapse
Affiliation(s)
- Edward P Rybicki
- Biopharming Research Unit, Department of Molecular & Cell Biology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Private Bag X3, Rondebosch, 7701, Cape Town, South Africa.
| |
Collapse
|
36
|
Nanoparticle encapsidation of Flock house virus by auto assembly of Tobacco mosaic virus coat protein. Int J Mol Sci 2014; 15:18540-56. [PMID: 25318056 PMCID: PMC4227231 DOI: 10.3390/ijms151018540] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 09/09/2014] [Accepted: 09/29/2014] [Indexed: 01/01/2023] Open
Abstract
Tobacco Mosaic virus (TMV) coat protein is well known for its ability to self-assemble into supramolecular nanoparticles, either as protein discs or as rods originating from the ~300 bp genomic RNA origin-of-assembly (OA). We have utilized TMV self-assembly characteristics to create a novel Flock House virus (FHV) RNA nanoparticle. FHV encodes a viral polymerase supporting autonomous replication of the FHV genome, which makes it an attractive candidate for viral transgene expression studies and targeted RNA delivery into host cells. However, FHV viral genome size is strictly limited by native FHV capsid. To determine if this packaging restriction could be eliminated, FHV was adapted to express enhanced green fluorescent protein (GFP), to allow for monitoring of functional FHV RNA activity. Then TMV OA was introduced in six 3' insertion sites, with only site one supporting functional FHV GFP expression. To create nanoparticles, FHV GFP-OA modified genomic RNA was mixed in vitro with TMV coat protein and monitored for encapsidation by agarose electrophoresis and electron microscopy. The production of TMV-like rod shaped nanoparticles indicated that modified FHV RNA can be encapsidated by purified TMV coat protein by self-assembly. This is the first demonstration of replication-independent packaging of the FHV genome by protein self-assembly.
Collapse
|
37
|
Kemnade JO, Seethammagari M, Collinson-Pautz M, Kaur H, Spencer DM, McCormick AA. Tobacco mosaic virus efficiently targets DC uptake, activation and antigen-specific T cell responses in vivo. Vaccine 2014; 32:4228-33. [PMID: 24923637 DOI: 10.1016/j.vaccine.2014.04.051] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/07/2014] [Accepted: 04/17/2014] [Indexed: 11/28/2022]
Abstract
Over the past 20 years, dendritic cells (DCs) have been utilized to activate immune responses capable of eliminating cancer cells. Currently, ex vivo DC priming has been the mainstay of DC cancer immunotherapies. However, cell-based treatment modalities are inherently flawed due to a lack of standardization, specialized facilities and personnel, and cost. Therefore, direct modes of DC manipulation, circumventing the need for ex vivo culture, must be investigated. To facilitate the development of next-generation, in vivo targeted DC vaccines, we characterized the DC interaction and activation potential of the Tobacco Mosaic virus (TMV), a plant virus that enjoys a relative ease of production and the ability to deliver protein payloads via surface conjugation. In this study we show that TMV is readily taken up by mouse bone marrow-derived DCs, in vitro. Footpad injection of fluorophore-labeled TMV reveals preferential uptake by draining lymph node resident DCs in vivo. Uptake leads to activation, as measured by the upregulation of key DC surface markers. When peptide antigen-conjugated TMV is injected into the footpad of mice, DC-mediated uptake and activation leads to robust antigen-specific CD8(+) T cell responses, as measured by antigen-specific tetramer analysis. Remarkably, TMV priming induced a greater magnitude T cell response than Adenovirus (Ad) priming. Finally, TMV is capable of boosting either Ad-induced or TMV-induced antigen-specific T cell responses, demonstrating that TMV, uniquely, does not induce neutralizing self-immunity. Overall, this study elucidates the in vivo DC delivery and activation properties of TMV and indicates its potential as a vaccine vector in stand alone or prime-boost strategies.
Collapse
Affiliation(s)
| | | | | | - Hardeep Kaur
- Touro University California, Vallejo, CA, United States.
| | | | | |
Collapse
|