101
|
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
|
102
|
Choi HJ, Song JM, Bondy BJ, Compans RW, Kang SM, Prausnitz MR. Effect of Osmotic Pressure on the Stability of Whole Inactivated Influenza Vaccine for Coating on Microneedles. PLoS One 2015; 10:e0134431. [PMID: 26230936 PMCID: PMC4521748 DOI: 10.1371/journal.pone.0134431] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 07/10/2015] [Indexed: 11/18/2022] Open
Abstract
Enveloped virus vaccines can be damaged by high osmotic strength solutions, such as those used to protect the vaccine antigen during drying, which contain high concentrations of sugars. We therefore studied shrinkage and activity loss of whole inactivated influenza virus in hyperosmotic solutions and used those findings to improve vaccine coating of microneedle patches for influenza vaccination. Using stopped-flow light scattering analysis, we found that the virus underwent an initial shrinkage on the order of 10% by volume within 5 s upon exposure to a hyperosmotic stress difference of 217 milliosmolarity. During this shrinkage, the virus envelope had very low osmotic water permeability (1 - 6×10-4 cm s-1) and high Arrhenius activation energy (Ea = 15.0 kcal mol-1), indicating that the water molecules diffused through the viral lipid membranes. After a quasi-stable state of approximately 20 s to 2 min, depending on the species and hypertonic osmotic strength difference of disaccharides, there was a second phase of viral shrinkage. At the highest osmotic strengths, this led to an undulating light scattering profile that appeared to be related to perturbation of the viral envelope resulting in loss of virus activity, as determined by in vitro hemagglutination measurements and in vivo immunogenicity studies in mice. Addition of carboxymethyl cellulose effectively prevented vaccine activity loss in vitro and in vivo, believed to be due to increasing the viscosity of concentrated sugar solution and thereby reducing osmotic stress during coating of microneedles. These results suggest that hyperosmotic solutions can cause biphasic shrinkage of whole inactivated influenza virus which can damage vaccine activity at high osmotic strength and that addition of a viscosity enhancer to the vaccine coating solution can prevent osmotically driven damage and thereby enable preparation of stable microneedle coating formulations for vaccination.
Collapse
Affiliation(s)
- Hyo-Jick Choi
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Jae-Min Song
- Department of Global Medical Science, Sungshin Women's University, Seoul, Korea
| | - Brian J. Bondy
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Richard W. Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Sang-Moo Kang
- Center for Inflammation, Immunity, & Infection and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
103
|
Incorporation of conserved nucleoprotein into influenza virus-like particles could provoke a broad protective immune response in BALB/c mice and chickens. Virus Res 2015; 195:35-42. [PMID: 25312452 DOI: 10.1016/j.virusres.2014.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/05/2014] [Accepted: 09/30/2014] [Indexed: 10/24/2022]
Abstract
We engineered influenza A/goose/GD/1996 (H5N1) (clade 0) virus-like particles (VLPs) by coinfecting Sf9 cells with triple/quadruple recombinant baculovirus that expressed hemagglutinin (HA), neuraminidase (NA), and matrix 1 (M1) with or without nucleoprotein (NP). VLP3 (HA, NA, and M1) and VLP4 (HA, NA, M1, and NP) vaccines (containing 1 μg HA) with oil emulsion were administered to mice and chickens by intramuscular injection, and the immune responses were analyzed. The VLP-vaccinated mice demonstrated high antigen specific antibody titers and effective cellular immune responses. The mice and chickens vaccinated with VLP4 demonstrated more robust humoral and cellular immune responses than those vaccinated with VLP3. The VLP4 vaccine afforded 100% protection against a heterologous lethal influenza virus challenge (clade 2.3.4) whereas the VLP3 vaccine conferred 50% protection in chickens. These results implied that the incorporation of conserved NP protein into the VLPs could elicit a broad protective immune response in BALB/c mice and chickens. To the best of our knowledge, this study is the first report describing the immunological profile of the NP-containing VLPs vaccines in mice and chicken models, and the results demonstrate that the non-infectious, genome less VLPs, particularly those containing NP, represent a promising strategy for the development of a safe and effective vaccine to control pandemic influenza.
Collapse
|
104
|
Lee YT, Kim KH, Hwang HS, Lee Y, Kwon YM, Ko EJ, Jung YJ, Lee YN, Kim MC, Kang SM. Innate and adaptive cellular phenotypes contributing to pulmonary disease in mice after respiratory syncytial virus immunization and infection. Virology 2015. [PMID: 26196232 DOI: 10.1016/j.virol.2015.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Respiratory syncytial virus (RSV) is the major leading cause of infantile viral bronchiolitis. However, cellular phenotypes contributing to the RSV protection and vaccine-enhanced disease remain largely unknown. Upon RSV challenge, we analyzed phenotypes and cellularity in the lung of mice that were naïve, immunized with formalin inactivated RSV (FI-RSV), or re-infected with RSV. In comparison with naïve and live RSV re-infected mice, the high levels of eosinophils, neutrophils, plasmacytoid and CD11b(+) dendritic cells, and IL-4(+) CD4(+) T cells were found to be contributing to pulmonary inflammation in FI-RSV immune mice despite lung viral clearance. Alveolar macrophages appeared to play differential roles in protection and inflammation upon RSV infection of different RSV immune mice. These results suggest that multiple innate and adaptive immune components differentially contribute to RSV disease and inflammation.
Collapse
Affiliation(s)
- Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Animal and Plant Quarantine Agency, 175 Anyangro, Anyangsi, Gyeonggido 430-757, Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
105
|
Influenza bivalent vaccine comprising recombinant H3 hemagglutinin (HA) and H1 HA containing replaced H3 hemagglutinin transmembrane domain exhibited improved heterosubtypic protection immunity in mice. Vaccine 2015; 33:4035-40. [DOI: 10.1016/j.vaccine.2015.05.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/30/2015] [Accepted: 05/19/2015] [Indexed: 02/02/2023]
|
106
|
Rynda-Apple A, Patterson DP, Douglas T. Virus-like particles as antigenic nanomaterials for inducing protective immune responses in the lung. Nanomedicine (Lond) 2015; 9:1857-68. [PMID: 25325241 DOI: 10.2217/nnm.14.107] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The lung is a major entry point for many of the most detrimental pathogens to human health. The onslaught of pathogens encountered by the lung is counteracted by protective immune responses that are generated locally, which can be stimulated through vaccine strategies to prevent pathogen infections. Here, we discuss the use of virus-like particles (VLPs), nonpathogen derivatives of viruses or protein cage structures, to construct new vaccines exploiting the lung as a site for immunostimulation. VLPs are unique in their ability to be engineered with near molecular level detail and knowledge of their composition and structure. A summary of research in developing VLP-based vaccines for the lung is presented that suggests promising results for future vaccine development.
Collapse
|
107
|
Microneedle patch delivery to the skin of virus-like particles containing heterologous M2e extracellular domains of influenza virus induces broad heterosubtypic cross-protection. J Control Release 2015; 210:208-16. [PMID: 26003039 DOI: 10.1016/j.jconrel.2015.05.278] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 01/24/2023]
Abstract
A broadly cross-protective influenza vaccine that can be administrated by a painless self-immunization method would be a value as a potential universal mass vaccination strategy. This study developed a minimally-invasive microneedle (MN) patch for skin vaccination with virus-like particles containing influenza virus heterologous M2 extracellular (M2e) domains (M2e5x VLPs) as a universal vaccine candidate without adjuvants. The stability of M2e5x VLP-coated microneedles was maintained for 8weeks at room temperature without losing M2e antigenicity and immunogenicity. MN skin immunization induced strong humoral and mucosal M2e antibody responses and conferred cross-protection against heterosubtypic H1N1, H3N2, and H5N1 influenza virus challenges. In addition, M2e5x VLP MN skin vaccination induced T-helper type 1 responses such as IgG2a isotype antibodies and IFN-γ producing cells at higher levels than those by conventional intramuscular injection. These potential immunological and logistic advantages for skin delivery of M2e5x VLP MN vaccines could offer a promising approach to develop an easy-to-administer universal influenza vaccine.
Collapse
|
108
|
Sokolova V, Westendorf AM, Buer J, Überla K, Epple M. The potential of nanoparticles for the immunization against viral infections. J Mater Chem B 2015; 3:4767-4779. [PMID: 32262665 DOI: 10.1039/c5tb00618j] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vaccination has a great impact on the prevention and control of infectious diseases. However, there are still many infectious diseases for which an effective vaccine is missing. Thirty years after the discovery of the AIDS-pathogen (human immunodeficiency virus, HIV) and intensive research, there is still no protective immunity against the HIV infection. Over the past decade, nanoparticulate systems such as virus-like particles, liposomes, polymers and inorganic nanoparticles have received attention as potential delivery vehicles which can be loaded or functionalized with active biomolecules (antigens and adjuvants). Here we compare the properties of different nanoparticulate systems and assess their potential for the development of new vaccines against a range of viral infections.
Collapse
Affiliation(s)
- Viktoriya Sokolova
- Inorganic Chemistry and Centre for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitaetsstr. 5-7, 45117 Essen, Germany.
| | | | | | | | | |
Collapse
|
109
|
Thompson CM, Petiot E, Mullick A, Aucoin MG, Henry O, Kamen AA. Critical assessment of influenza VLP production in Sf9 and HEK293 expression systems. BMC Biotechnol 2015; 15:31. [PMID: 25981500 PMCID: PMC4432999 DOI: 10.1186/s12896-015-0152-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/22/2015] [Indexed: 12/22/2022] Open
Abstract
Background Each year, influenza is responsible for hundreds of thousand cases of illness and deaths worldwide. Due to the virus’ fast mutation rate, the World Health Organization (WHO) is constantly on alert to rapidly respond to emerging pandemic strains. Although anti-viral therapies exist, the most proficient way to stop the spread of disease is through vaccination. The majority of influenza vaccines on the market are produced in embryonic hen’s eggs and are composed of purified viral antigens from inactivated whole virus. This manufacturing system, however, is limited in its production capacity. Cell culture produced vaccines have been proposed for their potential to overcome the problems associated with egg-based production. Virus-like particles (VLPs) of influenza virus are promising candidate vaccines under consideration by both academic and industry researchers. Methods In this study, VLPs were produced in HEK293 suspension cells using the Bacmam transduction system and Sf9 cells using the baculovirus infection system. The proposed systems were assessed for their ability to produce influenza VLPs composed of Hemagglutinin (HA), Neuraminidase (NA) and Matrix Protein (M1) and compared through the lens of bioprocessing by highlighting baseline production yields and bioactivity. VLPs from both systems were characterized using available influenza quantification techniques, such as single radial immunodiffusion assay (SRID), HA assay, western blot and negative staining transmission electron microscopy (NSTEM) to quantify total particles. Results For the HEK293 production system, VLPs were found to be associated with the cell pellet in addition to those released in the supernatant. Sf9 cells produced 35 times more VLPs than HEK293 cells. Sf9-VLPs had higher total HA activity and were generally more homogeneous in morphology and size. However, Sf9 VLP samples contained 20 times more baculovirus than VLPs, whereas 293 VLPs were produced along with vesicles. Conclusions This study highlights key production hurdles that must be overcome in both expression platforms, namely the presence of contaminants and the ensuing quantification challenges, and brings up the question of what truly constitutes an influenza VLP candidate vaccine. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0152-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine M Thompson
- National Research Council Canada, Human Health Therapeutics, Montréal, Canada. .,Ecole Polytechnique de Montréal, Montréal, Canada.
| | - Emma Petiot
- National Research Council Canada, Human Health Therapeutics, Montréal, Canada. .,Laboratoire Virologie et pathologies Humaine (VirPath), EA4610, Lyon, France.
| | - Alaka Mullick
- National Research Council Canada, Human Health Therapeutics, Montréal, Canada.
| | | | | | - Amine A Kamen
- National Research Council Canada, Human Health Therapeutics, Montréal, Canada. .,Department of Bioengineering, McGill University, 817 Sherbrooke St. W. Macdonald Engineering Building, Room 387, Montréal, Canada.
| |
Collapse
|
110
|
Talactac MR, Chowdhury MYE, Park ME, Weeratunga P, Kim TH, Cho WK, Kim CJ, Ma JY, Lee JS. Antiviral Effects of Novel Herbal Medicine KIOM-C, on Diverse Viruses. PLoS One 2015; 10:e0125357. [PMID: 25942440 PMCID: PMC4420246 DOI: 10.1371/journal.pone.0125357] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 03/11/2015] [Indexed: 01/28/2023] Open
Abstract
In order to identify new potential antiviral agents, recent studies have advocated thorough testing of herbal medicines or natural substances that are traditionally used to prevent viral infections. Antiviral activities and the mechanism of action of the total aqueous extract preparation of KIOM-C, a novel herbal medicine, against diverse types of viruses were investigated. In vitro antiviral activity against A/Puerto Rico/8/34 (H1N1) (PR8), vesicular stomatitis virus (VSV), and Newcastle disease virus (NDV) through the induction of type-I interferon related protein phosphorylation and up-regulation of pro-inflammatory cytokines in murine macrophage cells (RAW264.7) were determined. In vivo, KIOM-C-treated BALB/c mice showed higher survivability and lower lung viral titers when challenged with A/Aquatic bird/Korea/W81/2005 (H5N2), A/PR/8/34(H1N1), A/Aquatic bird/Korea/W44/2005(H7N3) or A/Chicken/Korea/116 /2004(H9N2) influenza subtypes in contrast with the non-treated group. The present study revealed that total aqueous extract preparation of KIOM-C stimulates an antiviral state in murine macrophage cells and in mice leading to inhibition of viral infection and protection against lethal challenges.
Collapse
Affiliation(s)
- Melbourne R. Talactac
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Cavite, Philippines
| | - Mohammed Y. E. Chowdhury
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Faculty of Veterinary Medicine, Chittagong Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Min-Eun Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Prasanna Weeratunga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Tae-Hwan Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Won-Kyung Cho
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon, and Republic of Korea
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon, and Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
111
|
Tsen SWD, Donthi N, La V, Hsieh WH, Li YD, Knoff J, Chen A, Wu TC, Hung CF, Achilefu S, Tsen KT. Chemical-free inactivated whole influenza virus vaccine prepared by ultrashort pulsed laser treatment. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:051008. [PMID: 25423046 PMCID: PMC4242973 DOI: 10.1117/1.jbo.20.5.051008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/24/2014] [Indexed: 05/22/2023]
Abstract
There is an urgent need for rapid methods to develop vaccines in response to emerging viral pathogens. Whole inactivated virus (WIV) vaccines represent an ideal strategy for this purpose; however, a universal method for producing safe and immunogenic inactivated vaccines is lacking. Conventional pathogen inactivation methods such as formalin, heat, ultraviolet light, and gamma rays cause structural alterations in vaccines that lead to reduced neutralizing antibody specificity, and in some cases, disastrous T helper type 2-mediated immune pathology. We have evaluated the potential of a visible ultrashort pulsed (USP) laser method to generate safe and immunogenic WIV vaccines without adjuvants. Specifically, we demonstrate that vaccination of mice with laser-inactivated H1N1 influenza virus at about a 10-fold lower dose than that required using conventional formalin-inactivated influenza vaccines results in protection against lethal H1N1 challenge in mice. The virus, inactivated by the USP laser irradiation, has been shown to retain its surface protein structure through hemagglutination assay. Unlike conventional inactivation methods, laser treatment did not generate carbonyl groups in protein, thereby reducing the risk of adverse vaccine-elicited T helper type 2 responses. Therefore, USP laser treatment is an attractive potential strategy to generate WIV vaccines with greater potency and safety than vaccines produced by current inactivation techniques.
Collapse
Affiliation(s)
- Shaw-Wei David Tsen
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri 63110, United States
| | - Nisha Donthi
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
| | - Victor La
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
| | - Wen-Han Hsieh
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
| | - Yen-Der Li
- National Taiwan University, College of Medicine, Taipei 10617, Taiwan
| | - Jayne Knoff
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
| | - Alexander Chen
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
| | - Tzyy-Choou Wu
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
- Johns Hopkins Medical Institutions, Department of Obstetrics and Gynecology, Baltimore, Maryland 21231, United States
- Johns Hopkins Medical Institutions, Department of Molecular Microbiology and Immunology, Baltimore, Maryland 21231, United States
- Johns Hopkins Medical Institutions, Department of Oncology, Baltimore, Maryland 21231, United States
| | - Chien-Fu Hung
- Johns Hopkins Medical Institutions, Department of Pathology, Baltimore, Maryland 21231, United States
- Johns Hopkins Medical Institutions, Department of Oncology, Baltimore, Maryland 21231, United States
- Address all correspondence to: Kong-Thon Tsen, E-mail: ; Chien-Fu Hung, E-mail:
| | - Samuel Achilefu
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biochemistry and Molecular Biophysics, St. Louis, Missouri 63110, United States
- Washington University School of Medicine, Department of Biomedical Engineering, St. Louis, Missouri 63110, United States
| | - Kong-Thon Tsen
- Arizona State University, Department of Physics and Center for Biophysics, Tempe, Arizona 85287, United States
- Address all correspondence to: Kong-Thon Tsen, E-mail: ; Chien-Fu Hung, E-mail:
| |
Collapse
|
112
|
Cao L, Mao F, Pang Z, Yi Y, Qiu F, Tian R, Meng Q, Jia Z, Bi S. Protective effect of enterovirus‑71 (EV71) virus‑like particle vaccine against lethal EV71 infection in a neonatal mouse model. Mol Med Rep 2015; 12:2473-80. [PMID: 25936344 PMCID: PMC4464482 DOI: 10.3892/mmr.2015.3680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 03/16/2015] [Indexed: 01/01/2023] Open
Abstract
Enterovirus-71 (EV71) is a viral pathogen that causes severe cases of hand, foot and mouth disease (HFMD) among young children, with significant mortality. Effective vaccines against HFMD are urgently required. Several EV71 virus-like particle (VLP) vaccine candidates were found to be protective in the neonatal mouse EV71 challenge model. However, to what extent the VLP vaccine protects susceptible organs against EV71 infection in vivo has remained elusive. In the present study, the comprehensive immunogenicity of a potential EV71 vaccine candidate based on VLPs was evaluated in a neonatal mouse model. Despite lower levels of neutralizing antibodies to EV71 in the sera of VLP-immunized mice compared with those in mice vaccinated with inactivated EV71, the VLP-based vaccine was shown to be able to induce immunoglobulin (Ig)G and IgA memory-associated cellular immune responses to EV71. Of note, the EV71 VLP vaccine candidate was capable of inhibiting viral proliferation in cardiac muscle, skeletal muscle, lung and intestine of immunized mice and provided effective protection against the pathological damage caused by viral attack. In particular, the VLP vaccine was able to inhibit the transportation of EV71 from the central nervous system to the muscle tissue and greatly protected muscle tissue from infection, along with recovery from the viral infection. This led to nearly 100% immunoprotective efficacy, enabling neonatal mice delivered by VLP-immunized female adult mice to survive and grow with good health. The present study provided valuable additional knowledge of the specific protective efficacy of the EV71 VLP vaccine in vivo, which also indicated that it is a promising potential candidate for being developed into an EV71 vaccine.
Collapse
Affiliation(s)
- Lei Cao
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Fengfeng Mao
- Laboratory Animal Center, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zheng Pang
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Yao Yi
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Feng Qiu
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Ruiguang Tian
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Qingling Meng
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Zhiyuan Jia
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| | - Shengli Bi
- Department of Viral Hepatitis, National Institute for Viral Disease Control and Prevention, China Center for Disease Control and Prevention, Beijing 102206, P.R. China
| |
Collapse
|
113
|
Tsou YL, Lin YW, Shao HY, Yu SL, Wu SR, Lin HY, Liu CC, Huang C, Chong P, Chow YH. Recombinant adeno-vaccine expressing enterovirus 71-like particles against hand, foot, and mouth disease. PLoS Negl Trop Dis 2015; 9:e0003692. [PMID: 25855976 PMCID: PMC4391779 DOI: 10.1371/journal.pntd.0003692] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/10/2015] [Indexed: 12/24/2022] Open
Abstract
Enterovirus 71 (EV71) and coxsackieviruses (CV) are the major causative agents of hand, foot and mouth disease (HFMD). There is not currently a vaccine available against HFMD, even though a newly developed formalin-inactivated EV71 (FI-EV71) vaccine has been tested in clinical trial and has shown efficacy against EV71. We have designed and genetically engineered a recombinant adenovirus Ad-EVVLP with the EV71 P1 and 3CD genes inserted into the E1/E3-deleted adenoviral genome. Ad-EVVLP were produced in HEK-293A cells. In addition to Ad-EVVLP particles, virus-like particles (VLPs) formed from the physical association of EV71 capsid proteins, VP0, VP1, and VP3 expressed from P1 gene products. They were digested by 3CD protease and confirmed to be produced by Ad-EVVLP-producing cells, as determined using transmission electron microscopy and western blotting. Mouse immunogenicity studies showed that Ad-EVVLP-immunized antisera neutralized the EV71 B4 and C2 genotypes. Activation of VLP-specific CD4+ and CD8+/IFN-γ T cells associated with Th1/Th2-balanced IFN-ɣ, IL-17, IL-4, and IL-13 was induced; in contrast, FI-EV71 induced only Th2-mediated neutralizing antibody against EV71 and low VLP-specific CD4+ and CD8+ T cell responses. The antiviral immunity against EV71 was clearly demonstrated in mice vaccinated with Ad-EVVLP in a hSCARB2 transgenic (hSCARB2-Tg) mouse challenge model. Ad-EVVLP-vaccinated mice were 100% protected and demonstrated reduced viral load in both the CNS and muscle tissues. Ad-EVVLP successfully induced anti-CVA16 immunities. Although antisera had no neutralizing activity against CVA16, the 3C-specific CD4+ and CD8+/IFN-γ T cells were identified, which could mediate protection against CVA16 challenge. FI-EV71 did not induce 3C-mediated immunity and had no efficacy against the CVA16 challenge. These results suggest that Ad-EVVLP can enhance neutralizing antibody and protective cellular immune responses to prevent EV71 infection and cellular immune responses against CV infection.
Collapse
Affiliation(s)
- Yueh-Liang Tsou
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Program of Biotechnology in Medicine, Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Wen Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Yun Shao
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Program of Biotechnology in Medicine, Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Shang-Rung Wu
- Institute of Oral Medicine, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Hsiao-Yu Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Chyi Liu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Chieh Huang
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Pele Chong
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
114
|
Rahn J, Hoffmann D, Harder TC, Beer M. Vaccines against influenza A viruses in poultry and swine: Status and future developments. Vaccine 2015; 33:2414-24. [PMID: 25835575 DOI: 10.1016/j.vaccine.2015.03.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/01/2015] [Accepted: 03/18/2015] [Indexed: 12/29/2022]
Abstract
Influenza A viruses are important pathogens with a very broad host spectrum including domestic poultry and swine. For preventing clinical disease and controlling the spread, vaccination is one of the most efficient tools. Classical influenza vaccines for domestic poultry and swine are conventional inactivated preparations. However, a very broad range of novel vaccine types ranging from (i) nucleic acid-based vaccines, (ii) replicon particles, (iii) subunits and virus-like particles, (iv) vectored vaccines, or (v) live-attenuated vaccines has been described, and some of them are now also used in the field. The different novel approaches for vaccines against avian and swine influenza virus infections are reviewed, and additional features like universal vaccines, novel application approaches and the "differentiating infected from vaccinated animals" (DIVA)-strategy are summarized.
Collapse
Affiliation(s)
- J Rahn
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany
| | - D Hoffmann
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany
| | - T C Harder
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany
| | - M Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany.
| |
Collapse
|
115
|
Cho WK, Weeratunga P, Lee BH, Park JS, Kim CJ, Ma JY, Lee JS. Epimedium koreanum Nakai displays broad spectrum of antiviral activity in vitro and in vivo by inducing cellular antiviral state. Viruses 2015; 7:352-77. [PMID: 25609307 PMCID: PMC4306843 DOI: 10.3390/v7010352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/14/2015] [Indexed: 01/15/2023] Open
Abstract
Epimedium koreanum Nakai has been extensively used in traditional Korean and Chinese medicine to treat a variety of diseases. Despite the plant's known immune modulatory potential and chemical make-up, scientific information on its antiviral properties and mode of action have not been completely investigated. In this study, the broad antiviral spectrum and mode of action of an aqueous extract from Epimedium koreanum Nakai was evaluated in vitro, and moreover, the protective effect against divergent influenza A subtypes was determined in BALB/c mice. An effective dose of Epimedium koreanum Nakai markedly reduced the replication of Influenza A Virus (PR8), Vesicular Stomatitis Virus (VSV), Herpes Simplex Virus (HSV) and Newcastle Disease Virus (NDV) in RAW264.7 and HEK293T cells. Mechanically, we found that an aqueous extract from Epimedium koreanum Nakai induced the secretion of type I IFN and pro-inflammatory cytokines and the subsequent stimulation of the antiviral state in cells. Among various components present in the extract, quercetin was confirmed to have striking antiviral properties. The oral administration of Epimedium koreanum Nakai exhibited preventive effects on BALB/c mice against lethal doses of highly pathogenic influenza A subtypes (H1N1, H5N2, H7N3 and H9N2). Therefore, an extract of Epimedium koreanum Nakai and its components play roles as immunomodulators in the innate immune response, and may be potential candidates for prophylactic or therapeutic treatments against diverse viruses in animal and humans.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon 305-764, Korea.
| | - Prasanna Weeratunga
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Byeong-Hoon Lee
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Jun-Seol Park
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Jin Yeul Ma
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon 305-764, Korea.
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| |
Collapse
|
116
|
Ren Z, Ji X, Meng L, Wei Y, Wang T, Feng N, Zheng X, Wang H, Li N, Gao X, Jin H, Zhao Y, Yang S, Qin C, Gao Y, Xia X. H5N1 influenza virus-like particle vaccine protects mice from heterologous virus challenge better than whole inactivated virus. Virus Res 2015; 200:9-18. [PMID: 25599603 DOI: 10.1016/j.virusres.2015.01.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/06/2015] [Accepted: 01/10/2015] [Indexed: 12/20/2022]
Abstract
The highly pathogenic avian influenza (HPAI) H5N1 virus has become highly enzootic since 2003 and has dynamically evolved to undergo substantial evolution. Clades 2.3.2.1 and 2.3.4 have become the most dominant lineage in recent years, and H5N8 avian influenza outbreaks have been reported Asia. The current approach to generate influenza virus vaccines uses embryonated chicken eggs for large-scale production, although such vaccines have been poorly immunogenic to heterologous virus challenge. In the current study, virus-like particles (VLP) based on A/meerkat/Shanghai/SH-1/2012 (clade 2.3.2.1) and comprising hemagglutinin (HA), neuraminidase (NA), and matrix (M1) were produced using a baculovirus expression system to develop effective protection for different H5 HPAI clade challenges. Mice immunized with VLP demonstrated stronger humoral and cellular immune responses than mice immunized with whole influenza virus (WIV), with 20-fold higher IgG antibody titers against A/meerkat/Shanghai/SH-1/2012 after boost. Notably, the WIV vaccine group showed partial protection (80% survival) to homologous challenge, little protection (40% survival) to heterologous challenge, and 20% survival to H5N8 challenge, whereas all mice in the VLP+CFA group survived. These results provide insight for the development of effective prophylactic vaccines based on VLPs with cross-clade protection for the control of current H5 HPAI outbreaks in humans.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/immunology
- Chick Embryo
- Cross Protection
- Female
- Humans
- Influenza A Virus, H5N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A virus/classification
- Influenza A virus/genetics
- Influenza A virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
Collapse
Affiliation(s)
- Zhiguang Ren
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xianliang Ji
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; College of veterinary Medicine, Inner Mongolia Agricultural University, Inner Mongolia Autonomous Region, Huhhot, China
| | - Lingnan Meng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
| | - Yurong Wei
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang Province, China
| | - Tiecheng Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Na Feng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xuexing Zheng
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Hualei Wang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Nan Li
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Xiaolong Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Hongli Jin
- Changchun SR Biological Technology Co., Ltd, Changchun, Jilin Province, China
| | - Yongkun Zhao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China
| | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuwei Gao
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| | - Xianzhu Xia
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu Province, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| |
Collapse
|
117
|
Yusibov V, Kushnir N, Streatfield SJ. Advances and challenges in the development and production of effective plant-based influenza vaccines. Expert Rev Vaccines 2014; 14:519-35. [PMID: 25487788 DOI: 10.1586/14760584.2015.989988] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Influenza infections continue to present a major threat to public health. Traditional modes of influenza vaccine manufacturing are failing to satisfy the global demand because of limited scalability and long production timelines. In contrast, subunit vaccines (SUVs) can be produced in heterologous expression systems in shorter times and at higher quantities. Plants are emerging as a promising platform for SUV production due to time efficiency, scalability, lack of harbored mammalian pathogens and possession of the machinery for eukaryotic post-translational protein modifications. So far, several organizations have utilized plant-based transient expression systems to produce SUVs against influenza, including vaccines based on virus-like particles. Plant-produced influenza SUV candidates have been extensively evaluated in animal models and some have shown safety and immunogenicity in clinical trials. Here, the authors review ongoing efforts and challenges to producing influenza SUV candidates in plants and discuss the likelihood of bringing these products to the market.
Collapse
Affiliation(s)
- Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, 9 Innovation Way, Suite 200, Newark, DE 19711, USA
| | | | | |
Collapse
|
118
|
Masri L, Cremer S. Individual and social immunisation in insects. Trends Immunol 2014; 35:471-82. [DOI: 10.1016/j.it.2014.08.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 01/03/2023]
|
119
|
Additive protection induced by mixed virus-like particles presenting respiratory syncytial virus fusion or attachment glycoproteins. Antiviral Res 2014; 111:129-35. [PMID: 25239522 DOI: 10.1016/j.antiviral.2014.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 01/13/2023]
Abstract
Respiratory syncytial virus (RSV) is the most important pathogen for lower respiratory tract illness in infants and a high priority for vaccine development. We previously reported that RSV virus-like particles (VLPs) expressing either the fusion (F) or attachment (G) glycoprotein could confer protection against RSV challenge in BALB/c mice. Here, we tested the hypothesis that RSV VLP vaccine efficacy can be enhanced by mixing RSV VLP F and RSV VLP G, and we analyzed host responses to these RSV VLPs. Mice were immunized with VLP F, VLP G, or VLP F+VLP G. Lung viral loads in BALB/c mice following RSV strain A2-line19F challenge were lower in mice vaccinated with RSV VLP F+VLP G compared to VLP F- or VLP G-vaccinated mice. Vaccination with VLP F or VLP F+VLP G induced similar levels of neutralizing antibodies. The enhanced protection against RSV challenge induced by vaccination with RSV VLP F+VLP G correlated with CD8 T cells producing T helper type 1 cytokines. VLP G vaccination alone followed by challenge resulted in immunopathology similar to formalin-inactivated RSV vaccination and RSV challenge. Taken together, mixed VLP F+VLP G provided a high level of protection against RSV without vaccine-induced immunopathology, but VLP G vaccination enhanced disease when used alone.
Collapse
|
120
|
O E, Ko EJ, Kim MC, Lee YT, Song JM, Kwon YM, Compans RW, Kang SM. Distinct B-cell populations contribute to vaccine antigen-specific antibody production in a transgenic mouse model. Immunology 2014; 142:624-35. [PMID: 24645831 DOI: 10.1111/imm.12287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 02/19/2014] [Accepted: 02/28/2014] [Indexed: 12/01/2022] Open
Abstract
The generation of memory B cells by vaccination plays a critical role in maintaining antigen-specific antibodies and producing antibody responses upon re-exposure to a pathogen. B-cell populations contributing to antibody production and protection by vaccination remain poorly defined. We used influenza virus-like particle (VLP) vaccine in a transgenic mouse model that would identify germinal centre-derived memory B cells with the expression of yellow fluorescent protein (YFP(+) cells). Immunization with influenza VLP vaccine did not induce significant increases in YFP(+) cells although vaccine antigen-specific antibodies in sera were found to confer protection against a lethal dose of influenza A virus (A/PR8). In addition, CD43(+) B220(-) populations with low YFP(+) cells mainly contributed to the production of vaccine antigen-specific IgG isotype-switched antibodies whereas CD43(-) B220(+) populations with high YFP(+) cells were able to produce vaccine antigen-specific IgM antibodies. Challenge infection of immunized transgenic mice with live influenza A virus resulted in significant increases in YFP(+) cells in the B220(-) populations of spleen and bone marrow cells. These results suggest that CD43(+) B220(-) B cells generated by vaccination are important for producing influenza vaccine antigen-specific antibodies and conferring protection.
Collapse
Affiliation(s)
- Eunju O
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Chimeric influenza-virus-like particles containing the porcine reproductive and respiratory syndrome virus GP5 protein and the influenza virus HA and M1 proteins. Arch Virol 2014; 159:3043-51. [PMID: 25064513 PMCID: PMC7086999 DOI: 10.1007/s00705-014-2178-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/11/2014] [Indexed: 12/03/2022]
Abstract
Both porcine reproductive and respiratory syndrome and swine influenza are acute, highly contagious swine diseases. These diseases pose severe threats for the swine industry and cause heavy economic losses worldwide. In this study, we have developed a chimeric virus-like particle (VLP) vaccine candidate for porcine reproductive and respiratory syndrome virus (PRRSV) and H3N2 influenza virus and investigated its immunogenicity in mice. The HA and M1 proteins from the H3N2 influenza virus and the PRRSV GP5 protein fused to the cytoplasmic and transmembrane domains of the NA protein were both incorporated into the chimeric VLPs. Analysis of the immune responses showed that the chimeric VLPs elicited serum antibodies specific for both PRRSV GP5 and the H3N2 HA protein, and they stimulated cellular immune responses compared to the responses to equivalent amounts of inactivated viruses. Taken together, the results suggested that the chimeric VLP vaccine represents a potential strategy for the development of a safe and effective vaccine to control PRRSV and H3N2 influenza virus.
Collapse
|
122
|
RNA virus reverse genetics and vaccine design. Viruses 2014; 6:2531-50. [PMID: 24967693 PMCID: PMC4113782 DOI: 10.3390/v6072531] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 12/22/2022] Open
Abstract
RNA viruses are capable of rapid spread and severe or potentially lethal disease in both animals and humans. The development of reverse genetics systems for manipulation and study of RNA virus genomes has provided platforms for designing and optimizing viral mutants for vaccine development. Here, we review the impact of RNA virus reverse genetics systems on past and current efforts to design effective and safe viral therapeutics and vaccines.
Collapse
|
123
|
Wohlbold TJ, Krammer F. In the shadow of hemagglutinin: a growing interest in influenza viral neuraminidase and its role as a vaccine antigen. Viruses 2014; 6:2465-94. [PMID: 24960271 PMCID: PMC4074938 DOI: 10.3390/v6062465] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 06/06/2014] [Accepted: 06/13/2014] [Indexed: 01/03/2023] Open
Abstract
Despite the availability of vaccine prophylaxis and antiviral therapeutics, the influenza virus continues to have a significant, annual impact on the morbidity and mortality of human beings, highlighting the continued need for research in the field. Current vaccine strategies predominantly focus on raising a humoral response against hemagglutinin (HA)—the more abundant, immunodominant glycoprotein on the surface of the influenza virus. In fact, anti-HA antibodies are often neutralizing, and are used routinely to assess vaccine immunogenicity. Neuraminidase (NA), the other major glycoprotein on the surface of the influenza virus, has historically served as the target for antiviral drug therapy and is much less studied in the context of humoral immunity. Yet, the quest to discern the exact importance of NA-based protection is decades old. Also, while antibodies against the NA glycoprotein fail to prevent infection of the influenza virus, anti-NA immunity has been shown to lessen the severity of disease, decrease viral lung titers in animal models, and reduce viral shedding. Growing evidence is intimating the possible gains of including the NA antigen in vaccine design, such as expanded strain coverage and increased overall immunogenicity of the vaccine. After giving a tour of general influenza virology, this review aims to discuss the influenza A virus neuraminidase while focusing on both the historical and present literature on the use of NA as a possible vaccine antigen.
Collapse
Affiliation(s)
- Teddy John Wohlbold
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1124, New York, NY 10029, USA.
| |
Collapse
|
124
|
Roles of major histocompatibility complex class II in inducing protective immune responses to influenza vaccination. J Virol 2014; 88:7764-75. [PMID: 24760891 DOI: 10.1128/jvi.00748-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Major histocompatibility complex class II-deficient (MHC-II KO; Aβ(-/-)) mice were used to assess the roles of MHC-II molecules in inducing protective immune responses to vaccination. After vaccination with influenza A/PR8 virus-like particle (VLP) vaccine, in vivo and in vitro vaccine antigen-specific IgG isotype antibodies were not detected in MHC-II KO mice, which is quite different from CD4 T cell-deficient mice that induced vaccine-specific IgG antibodies. The deficiency in MHC-II did not significantly affect the induction of antigen-specific IgM antibody in sera. MHC-II KO mice that were vaccinated with influenza VLP, whole inactivated influenza virus, or live attenuated influenza virus vaccines were not protected against lethal infection with influenza A/PR8 virus. Adoptive transfer of fractionated spleen cells from wild-type mice to MHC-II KO mice indicated that CD43(+) cell populations with MHC-II contributed more significantly to producing vaccine-specific IgG antibodies than CD43(-) B220(+) conventional B cell or CD4 T cell populations, as well as conferring protection against lethal infection. Bone marrow-derived dendritic cells from MHC-II KO mice showed a significant defect in producing interleukin-6 and tumor necrosis factor alpha cytokines. Thus, results indicate that MHC-II molecules play multiple roles in inducing protective immunity to influenza vaccination. Importance: Major histocompatibility complex class II (MHC-II) has been known to activate CD4 T helper immune cells. A deficiency in MHC-II was considered to be equivalent to the lack of CD4 T cells in developing host immune responses to pathogens. However, the roles of MHC-II in inducing protective immune responses to vaccination have not been well understood. In the present study, we demonstrate that MHC-II-deficient mice showed much more significant defects in inducing protective antibody responses to influenza vaccination than CD4 T cell-deficient mice. Further analysis showed that CD43 marker-positive immune cells with MHC-II, as well as an innate immunity-simulating adjuvant, could rescue some defects in inducing protective immune responses in MHC-II-deficient mice. These results have important implications for our understanding of host immunity-inducing mechanisms to vaccination, as well as in developing effective vaccines and adjuvants.
Collapse
|
125
|
Chowdhury MYE, Li R, Kim JH, Park ME, Kim TH, Pathinayake P, Weeratunga P, Song MK, Son HY, Hong SP, Sung MH, Lee JS, Kim CJ. Mucosal vaccination with recombinant Lactobacillus casei-displayed CTA1-conjugated consensus matrix protein-2 (sM2) induces broad protection against divergent influenza subtypes in BALB/c mice. PLoS One 2014; 9:e94051. [PMID: 24714362 PMCID: PMC3979752 DOI: 10.1371/journal.pone.0094051] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/11/2014] [Indexed: 01/31/2023] Open
Abstract
To develop a safe and effective mucosal vaccine against pathogenic influenza viruses, we constructed recombinant Lactobacillus casei strains that express conserved matrix protein 2 with (pgsA-CTA1-sM2/L. casei) or without (pgsA-sM2/L. casei) cholera toxin subunit A1 (CTA1) on the surface. The surface localization of the fusion protein was verified by cellular fractionation analyses, flow cytometry and immunofluorescence microscopy. Oral and nasal inoculations of recombinant L. casei into mice resulted in high levels of serum immunoglobulin G (IgG) and mucosal IgA. However, the conjugation of cholera toxin subunit A1 induced more potent mucosal, humoral and cell-mediated immune responses. In a challenge test with 10 MLD50 of A/EM/Korea/W149/06(H5N1), A/Puerto Rico/8/34(H1N1), A/Aquatic bird /Korea/W81/2005(H5N2), A/Aquatic bird/Korea/W44/2005(H7N3), and A/Chicken/Korea/116/2004(H9N2) viruses, the recombinant pgsA-CTA1-sM2/L. casei provided better protection against lethal challenges than pgsA-sM2/L. casei, pgsA/L. casei and PBS in mice. These results indicate that mucosal immunization with recombinant L. casei expressing CTA1-conjugated sM2 protein on its surface is an effective means of eliciting protective immune responses against diverse influenza subtypes.
Collapse
Affiliation(s)
- Mohammed Y. E. Chowdhury
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
- Faculty of Veterinary Medicine, Chittagong Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Rui Li
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Jae-Hoon Kim
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Min-Eun Park
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Tae-Hwan Kim
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Prabuddha Pathinayake
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Prasanna Weeratunga
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | - Man Ki Song
- Laboratory Science Division, International Vaccine Institute, Seoul, Republic of Korea
| | - Hwa-Young Son
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
| | | | | | - Jong-Soo Lee
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
- * E-mail: (CJK); (JSL)
| | - Chul-Joong Kim
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, Republic of Korea
- * E-mail: (CJK); (JSL)
| |
Collapse
|
126
|
Liu Q, Liu K, Xue C, Zhou J, Li X, Luo D, Zheng J, Xu S, Liu GD, Cao Y. Recombinant influenza H1, H5 and H9 hemagglutinins containing replaced H3 hemagglutinin transmembrane domain showed enhanced heterosubtypic protection in mice. Vaccine 2014; 32:3041-9. [PMID: 24704333 PMCID: PMC7115591 DOI: 10.1016/j.vaccine.2014.03.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/24/2014] [Accepted: 03/13/2014] [Indexed: 11/06/2022]
Abstract
We found H3-WT transmembrane domain is critical for H3 HA-induced hetero-protection. Wild-type H3 showed more hetero-protection than H1, H5 and H9 HAs. Replaced transmembrane domain had no apparent impact on in vitro expression of H1, H5 and H9 HA proteins in Sf9 cells. HAs with H3 transmembrane domain proteins exhibited enhanced heterosubtypic protections.
Influenza A viruses cause annual epidemics and irregular pandemics. A vaccine with heterosubtypic protection (hetero-protection) has been needed. In the present study, various influenza H1, H3, H5, and H9 hemagglutinin (HA) proteins were expressed in insect cells, and then mice were subcutaneously immunized with the expressed HA proteins, and challenged by influenza A viruses (A/Puerto Rico/8/1934 (H1N1) or A/chicken/Guangdong/96 (H9N2)). The results first showed that wild-type H3 hemagglutinin (HA) (H3-WT), but not a transmembrane domain (TM) mutant, had hetero-protection against both H1N1 and H9N2 with survival rates of 17% and 33% respectively, and that wild-type H1 (H1-WT), H5 (H5-WT) and H9 (H9-WT) had no hetero-protection against H1N1 or H9N2 except for H5-WT against H1N1 with a survival rate of 17%. Then the H3-WT TM replaced the TMs of H1-WT, H5-WT and H9-WT to generate recombinant H1-TM, H5-TM and H9-TM respectively, and whether the H3-WT TM-dependent hetero-protection could be transferred to these TM mutants was investigated. The results showed that the H3-WT TM-dependent hetero-protection was transferable. H1-TM against H9N2 and H9-TM against H1N1 were with survival rates of 33% and 17% respectively, and H5-TM against both H1N1 and H9N2 with survival rates of 50% and 17% respectively. Furthermore, higher dosage H5-TM scored 100% hetero-protection against H1N1. These results demonstrated that replacement of the TMs of non-H3 HAs with H3-WT TM could enhance their hetero-protection. These findings would help the development of future influenza vaccines against pandemics such as the recently appeared H7N9 infection.
Collapse
Affiliation(s)
- Qiliang Liu
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Kang Liu
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chunyi Xue
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jianqiang Zhou
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xiaoming Li
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Dongyu Luo
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Zheng
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shun Xu
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China
| | - George Dacai Liu
- Firstline Biopharmaceuticals Corporation, 12050 167th PL NE, Redmond, WA 98052, USA
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, Life Sciences School, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
127
|
Kim MC, Lee YN, Ko EJ, Lee JS, Kwon YM, Hwang HS, Song JM, Song BM, Lee YJ, Choi JG, Kang HM, Quan FS, Compans RW, Kang SM. Supplementation of influenza split vaccines with conserved M2 ectodomains overcomes strain specificity and provides long-term cross protection. Mol Ther 2014; 22:1364-1374. [PMID: 24590045 DOI: 10.1038/mt.2014.33] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 02/20/2014] [Indexed: 11/09/2022] Open
Abstract
Current influenza vaccines do not provide good protection against antigenically different influenza A viruses. As an approach to overcome strain specificity of protection, this study demonstrates significantly improved long-term cross protection by supplementing split vaccines with a conserved molecular target, a repeat of the influenza M2 ectodomain (M2e) expressed on virus-like particles (M2e5x VLPs) in a membrane-anchored form. Intramuscular immunization with H1N1 split vaccine (A/California/07/2009) supplemented with M2e5x VLPs induced M2e-specific humoral and cellular immune responses, and shaped the host responses to the vaccine in the direction of T-helper type 1 responses inducing dominant IgG2a isotype antibodies as well as interferon-γ (IFN-γ) producing cells in systemic and mucosal sites. Upon lethal challenge, M2e5x VLP-supplemented vaccination lowered lung viral loads and induced long-term cross protection against H3N2 or H5N1 subtype influenza viruses over 12 months. M2e antibodies, CD4 T cells, and CD8 T cells were found to contribute to improving heterosubtypic cross protection. In addition, improved cross protection by supplemented vaccination with M2e5x VLPs was mediated via Fc receptors. The results support evidence that supplementation with M2e5x VLPs is a promising approach for overcoming the limitation of strain-specific protection by current influenza vaccination.
Collapse
Affiliation(s)
- Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA; Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jong Seok Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jae-Min Song
- Department of Global medical Science, Sungshin Women's University, Seoul, Korea
| | - Byung-Min Song
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Youn-Jeong Lee
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Jun-Gu Choi
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Hyun-Mi Kang
- Animal and Plant Quarantine Agency, Anyang City, Gyeonggi-do, Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Korea
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
128
|
Subathra M, Santhakumar P, Satyam Naidu S, Lakshmi Narasu M, Senthilkumar TMA, Lal SK. Expression of avian influenza virus (H5N1) hemagglutinin and matrix protein 1 in Pichia pastoris and evaluation of their immunogenicity in mice. Appl Biochem Biotechnol 2014; 172:3635-45. [PMID: 24562978 DOI: 10.1007/s12010-014-0771-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/03/2014] [Indexed: 11/25/2022]
Abstract
The conventional avian influenza vaccines rely on development of neutralizing antibodies against the HA and NA antigens. However, these antigens are highly variable, and hence there is a need for better vaccine candidates which would offer broader protection in animals. The M1 of avian influenza is another major structural protein that has conserved epitopes that are reported to induce CD8+ T cells and can contribute to protection against morbidity and mortality from influenza. Hence in an effort to study the immune response of rM1 either alone or in combination with rHA, the hemagglutinin (HA) and matrix protein (M1) of A/Hatay/2004/H5N1 strain of avian influenza were expressed in Pichia pastoris as his-tagged proteins and purified through Ni-NTA chromatography. The His-tag was removed using TEV protease cleavage site and the immunogenicity of purified rHA and rM1 either alone or in combination was determined in mice. One group of mice was immunized with 5 μg of purified rHA, the other group was immunized with rM1, and a third group of mice were immunized with 5 μg of rHA and rM1. All the animals were boosted twice, once on 28 days postimmunization (dpi) and the second on 42 dpi. The immune response was evaluated by enzyme-linked immunosorbent assay (ELISA) and hemagglutination inhibition (HI) assay. The group of mice immunized with rHA and rM1 together showed significantly higher immune response against rHA and rM1 than mice immunized with either HA or M1 antigens. The addition of rM1 with rHA resulted in increased HI titer in animals immunized with both the antigens. These results suggest that the HA and M1 expressed in P. pastoris can be utilized in combination for the development of faster and cost-effective vaccines for circulating and newer strains of avian influenza and would aid in combating the disease in a pandemic situation, in which production time matters greatly.
Collapse
Affiliation(s)
- M Subathra
- Centre for Biotechnology, Jawaharlal Nehru Technological University, Hyderabad, 500085, India
| | | | | | | | | | | |
Collapse
|
129
|
Wang BZ, Gill HS, He C, Ou C, Wang L, Wang YC, Feng H, Zhang H, Prausnitz MR, Compans RW. Microneedle delivery of an M2e-TLR5 ligand fusion protein to skin confers broadly cross-protective influenza immunity. J Control Release 2014; 178:1-7. [PMID: 24417966 DOI: 10.1016/j.jconrel.2014.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/04/2013] [Accepted: 01/04/2014] [Indexed: 10/25/2022]
Abstract
Influenza vaccines with broad cross-protection are urgently needed to prevent an emerging influenza pandemic. A fusion protein of the Toll-like receptor (TLR) 5-agonist domains from flagellin and multiple repeats of the conserved extracellular domain of the influenza matrix protein 2 (M2e) was constructed, purified and evaluated as such a vaccine. A painless vaccination method suitable for possible self-administration using coated microneedle arrays was investigated for skin-targeted delivery of the fusion protein in a mouse model. The results demonstrate that microneedle immunization induced strong humoral as well as mucosal antibody responses and conferred complete protection against homo- and heterosubtypic lethal virus challenges. Protective efficacy with microneedles was found to be significantly better than that seen with conventional intramuscular injection, and comparable to that observed with intranasal immunization. Because of its advantages for administration, safety and storage, microneedle delivery of M2e-flagellin fusion protein is a promising approach for an easy-to-administer universal influenza vaccine.
Collapse
Affiliation(s)
- Bao-Zhong Wang
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Harvinder S Gill
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Cheng He
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China
| | - Changbo Ou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100094, China
| | - Li Wang
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ying-Chun Wang
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hao Feng
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Han Zhang
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Richard W Compans
- Department of Microbiology and Immunology, and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
130
|
Kapoor S, Dhama K. Prevention and Control of Influenza Viruses. INSIGHT INTO INFLUENZA VIRUSES OF ANIMALS AND HUMANS 2014. [PMCID: PMC7121144 DOI: 10.1007/978-3-319-05512-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The 2003–2004 outbreaks of highly pathogenic avian influenza (HPAI) have proven to be disastrous to the regional poultry industry in Asia, and have raised serious worldwide public health apprehension regarding the steps that should be taken to urgently control HPAI. Control measures must be taken based on the principles of biosecurity and disease management and at the same time making public aware of the precautionary measures at the verge of outbreak. Creation of protection and surveillance zones, various vaccination strategies viz. routine, preventive, emergency, mass and targeted vaccination programmes using live, inactivated and recombinant vaccines are the common strategies adopted in different parts of the globe. The new generation vaccines include recombinant vaccines and recombinant fusion vaccine. The pro-poor disease control programmes, giving compensation and subsidies to the farmers along with effective and efficient Veterinary Services forms integral part of control of HPAI. Following biosecurity principles and vaccination forms integral part of control programme against swine and equine influenza as well. Use of neuraminidase (NA) inhibitors (Zanamivir and Oseltamivir) for the treatment of human influenza has been widely accepted worldwide. The threat of increasing resistance of the flu viruses to these antivirals has evoked interest in the development of novel antiviral drugs for influenza virus such as inhibitors of cellular factors and host signalling cascades, cellular miRNAs, siRNA and innate immune peptides (defensins and cathelicidins). Commercial licensed inactivated vaccines for humans against influenza A and B viruses are available consisting of three influenza viruses: influenza type A subtype H3N2, influenza type A subtype H1N1 (seasonal) virus strain and influenza type B virus strain. As per WHO, use of tetravaccine consisting of antigens of influenza virus serotypes H3N2, H1N1, B and H5 is the most promising method to control influenza pandemic. All healthy children in many countries are required to be vaccinated between 6 and 59 months of age. The seasonal vaccines currently used in humans induce strain-specific humoral immunity as the antibodies. Universal influenza virus vaccines containing the relatively conserved ectodomain of M2 (M2e), M1, HA fusion peptide and stalk domains, NA, NP alone or in combination have been developed which have been shown to induce cross-protection. The T cell-based vaccines are another recent experimental approach that has been shown to elicit broad-spectrum heterosubtypic immunity in the host. As far as HPAI is concerned, various pandemic preparedness strategies have been documented.
Collapse
Affiliation(s)
- Sanjay Kapoor
- Department of Veterinary Microbiology, LLR University of Veterinary and Animal Sciences, Hisar, 125004 Haryana India
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar, Bareilly, 243122 Uttar Pradesh India
| |
Collapse
|
131
|
Inn KS, Lee GJ, Quan FS. A pandemic H1N1 influenza virus-like particle vaccine induces cross-protection in mice. Immunol Invest 2013; 43:236-54. [PMID: 24354853 DOI: 10.3109/08820139.2013.864665] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Influenza virus-like particles (VLPs) represent promising alternative vaccines. However, it is necessary to demonstrate that influenza VLPs confer cross-protection against antigenically distinct viruses. In this study, a VLP vaccine comprising hemagglutinin (HA) and M1 from the A/California/04/2009 (H1N1) were used and its ability to induce cross-protective efficacy against heterologous viruses A/PR/8/34 (H1N1) and A/New Caledonia/20/99 (H1N1) in mice was assessed. Vaccination with 2009 H1 VLPs induced significantly higher levels of IgG cross-reactive with these heterologous viruses after the second boost compared to after the prime or first boost. Lung virus titers also decreased significantly and the lung cross-reactive IgG response after lethal virus challenge was significantly greater in immunized mice compared to naïve mice. Vaccinated mice showed 100% protection against A/PR/8/34 and A/Caledonia/20/99 viruses with only moderate body weight loss and induction of cross-reactive recall, IgG antibody-secreting cell responses. The variations in HA amino acid sequences and antigenic sites were determined and correlated with induction of cross-protective immunity. These results indicate that VLPs can be used as an effective vaccine that confers cross-protection against antigenically distinct viruses.
Collapse
Affiliation(s)
- Kyung-Soo Inn
- Department of Pharmaceutical Science, College of Pharmacy, Kyung Hee University , Seoul , Korea 130-701
| | | | | |
Collapse
|
132
|
Kim YC, Yoo DG, Compans RW, Kang SM, Prausnitz MR. Cross-protection by co-immunization with influenza hemagglutinin DNA and inactivated virus vaccine using coated microneedles. J Control Release 2013; 172:579-88. [PMID: 23643528 PMCID: PMC3815987 DOI: 10.1016/j.jconrel.2013.04.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/16/2013] [Accepted: 04/22/2013] [Indexed: 12/11/2022]
Abstract
The need for annual revaccination against influenza is a burden on the healthcare system, leads to low vaccination rates and makes timely vaccination difficult against pandemic strains, such as during the 2009 H1N1 influenza pandemic. In an effort toward achieving a broadly protective vaccine that provides cross-protection against multiple strains of influenza, this study developed a microneedle patch to co-immunize with A/PR8 influenza hemagglutinin DNA and A/PR8 inactivated virus vaccine. We hypothesize that this dual component vaccination strategy administered to the skin using microneedles will provide cross-protection against other strains of influenza. To test this hypothesis, we developed a novel coating formulation that did not require additional excipients to increase coating solution viscosity by using the DNA vaccine itself to increase viscosity and thereby enable thick coatings of DNA vaccine and inactivated virus vaccine on metal microneedles. Co-immunization in this way not only generated robust antibody responses against A/PR8 influenza but also generated robust heterologous antibody responses against pandemic 2009 H1N1 influenza in mice. Challenge studies showed complete cross-protection against lethal challenge with live pandemic 2009 H1N1 virus. Control experiments using A/PR8 inactivated influenza virus vaccine with placebo DNA coated onto microneedles produced lower antibody titers and provided incomplete protection against challenge. Overall, this is the first study showing DNA solution as a microneedle coating agent and demonstrating cross-protection by co-immunization with inactivated virus and DNA vaccine using coated microneedles.
Collapse
MESH Headings
- Animals
- Antibody Formation
- Drug Delivery Systems/instrumentation
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Humans
- Immunization
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Injections, Intradermal
- Mice
- Mice, Inbred BALB C
- Needles
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Dae-Goon Yoo
- Department of Infectious Disease, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Richard W. Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Sang-Moo Kang
- Center for Inflammation, Immunity, and Infection, Georgia State University, Atlanta, GA30303, USA
| | - Mark R. Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
133
|
The highly conserved HA2 protein of the influenza a virus induces a cross protective immune response. J Virol Methods 2013; 194:280-8. [DOI: 10.1016/j.jviromet.2013.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/23/2013] [Accepted: 08/21/2013] [Indexed: 11/20/2022]
|
134
|
Baculoviral Co-Expression of HA, NA and M1 Proteins of Highly Pathogenic H5N1 Influenza Virus in Insect Cells. Jundishapur J Microbiol 2013. [DOI: 10.5812/jjm.7665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
135
|
Park MK, NGO V, Kwon YM, Lee YT, Yoo S, Cho YH, Hong SM, Hwang HS, Ko EJ, Jung YJ, Moon DW, Jeong EJ, Kim MC, Lee YN, Jang JH, Oh JS, Kim CH, Kang SM. Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS One 2013; 8:e75368. [PMID: 24124485 PMCID: PMC3790790 DOI: 10.1371/journal.pone.0075368] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 08/12/2013] [Indexed: 02/02/2023] Open
Abstract
Lactobacillus plantarum DK119 (DK119) isolated from the fermented Korean cabbage food was used as a probiotic to determine its antiviral effects on influenza virus. DK119 intranasal or oral administration conferred 100% protection against subsequent lethal infection with influenza A viruses, prevented significant weight loss, and lowered lung viral loads in a mouse model. The antiviral protective efficacy was observed in a dose and route dependent manner of DK119 administration. Mice that were treated with DK119 showed high levels of cytokines IL-12 and IFN-γ in bronchoalveolar lavage fluids, and a low degree of inflammation upon infection with influenza virus. Depletion of alveolar macrophage cells in lungs and bronchoalveolar lavages completely abrogated the DK119-mediated protection. Modulating host innate immunity of dendritic and macrophage cells, and cytokine production pattern appeared to be possible mechanisms by which DK119 exhibited antiviral effects on influenza virus infection. These results indicate that DK119 can be developed as a beneficial antiviral probiotic microorganism.
Collapse
Affiliation(s)
- Min-Kyung Park
- Department of Human Nutrition and Food Science, Chungwoon University, Namjang-Ri, Hongsung-Eup, Hongsung-Kun, Chungnam, Korea
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Vu NGO
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Sieun Yoo
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Young-Hee Cho
- Department of Animal Resource Science, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, Korea
| | - Sung-Moon Hong
- Department of Animal Resource Science, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, Korea
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Dae-Won Moon
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, Korea
| | - Eun-Ji Jeong
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, Korea
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
| | - Ji-Hun Jang
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, Korea
| | - Joon-Suk Oh
- Tobico Inc. Chungnam Techno Park, Jiksan-Eup, Seobuk-Gu, Cheonan-Si, Chungnam, Korea
| | - Cheol-Hyun Kim
- Department of Animal Resource Science, Dankook University, Dandae-ro, Dongnam-gu, Cheonan-si, Chungnam, Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
136
|
Shen H, Xue C, Lv L, Wang W, Liu Q, Liu K, Chen X, Zheng J, Li X, Cao Y. Assembly and immunological properties of a bivalent virus-like particle (VLP) for avian influenza and Newcastle disease. Virus Res 2013; 178:430-6. [PMID: 24050994 DOI: 10.1016/j.virusres.2013.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Revised: 07/25/2013] [Accepted: 09/06/2013] [Indexed: 11/15/2022]
Abstract
Avian influenza virus (AIV) and Newcastle disease virus (NDV) are both important pathogens in poultry worldwide. The protection of poultry from avian influenza and Newcastle disease can be achieved through vaccination. We embarked on the development of a bivalent vaccine that would allow for a single immunization against both avian influenza and Newcastle disease. We constructed a chimeric virus-like particle (VLP) that is composed of the M1 protein and HA protein of avian influenza virus and a chimeric protein containing the cytoplasmic and transmembrane domains of AIV neuraminidase protein (NA) and the ectodomain of the NDV hemagglutinin-neuraminidase (HN) protein (NA/HN). The single immunization of chickens with the chimeric VLP vaccine induced both AIV H5- and NDV-specific antibodies. The HI titers and specific antibodies elicited by the chimeric VLPs were statistically similar to those elicited in animals vaccinated with the corresponding commercial monovalent vaccines. Chickens vaccinated with chimeric VLP vaccine and then challenged with the Newcastle disease F48E9 virus displayed complete protection. Overall, the chimeric VLP vaccine elicits strong immunity and can protect against Newcastle disease virus challenge.
Collapse
Affiliation(s)
- Huifang Shen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Fernandes F, Teixeira AP, Carinhas N, Carrondo MJT, Alves PM. Insect cells as a production platform of complex virus-like particles. Expert Rev Vaccines 2013; 12:225-36. [PMID: 23414412 DOI: 10.1586/erv.12.153] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are multiprotein structures that resemble the conformation of native viruses but lack a viral genome, potentiating their application as safer and cheaper vaccines. The production of VLPs has been strongly linked with the use of insect cells and the baculovirus expression vector system, especially those particles composed of two or more structural viral proteins. In fact, this expression platform has been extensively improved over the years to address the challenges of coexpression of multiple proteins and their proper assembly into complexes in the same cell. In this article, the role of insect cell technology in the development and production of complex VLPs is overviewed; recent achievements, current bottlenecks and future trends are also highlighted.
Collapse
Affiliation(s)
- Fabiana Fernandes
- ITQB-Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | | | | | | | | |
Collapse
|
138
|
Lin SC, Liu WC, Lin YF, Huang YH, Liu JH, Wu SC. Heterologous prime-boost immunization regimens using adenovirus vector and virus-like particles induce broadly neutralizing antibodies against H5N1 avian influenza viruses. Biotechnol J 2013; 8:1315-22. [DOI: 10.1002/biot.201300116] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/06/2013] [Accepted: 06/21/2013] [Indexed: 12/20/2022]
|
139
|
Hemann EA, Kang SM, Legge KL. Protective CD8 T cell-mediated immunity against influenza A virus infection following influenza virus-like particle vaccination. THE JOURNAL OF IMMUNOLOGY 2013; 191:2486-94. [PMID: 23885108 DOI: 10.4049/jimmunol.1300954] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of influenza A virus (IAV) vaccines capable of inducing cytotoxic CD8 T cell responses could potentially provide superior, long-term protection against multiple, heterologous strains of IAV. Although prior studies demonstrated the effectiveness of baculovirus-derived virus-like particle (VLP) vaccination in generating Ab-mediated protection, the role that CD8 T cell immunity plays in overall VLP-mediated protection is less-well understood. In this article, we demonstrate that intranasal vaccination of mice with a VLP containing the hemagglutinin and matrix 1 proteins of IAV/PR/8/34 leads to a significant increase in hemagglutinin 533-specific CD8 T cells in the lungs and protection following subsequent homologous challenge with IAV. VLP-mediated protection was significantly reduced by CD8 T cell depletion, indicating a critical role for CD8 T cells in protective immunity. Importantly, our results show that VLP vaccine-induced CD8 T cell-mediated protection is not limited to homologous IAV strains. VLP vaccination leads to an increase in protection following heterosubtypic challenge with a strain of IAV that avoids vaccine-induced neutralizing Abs but contains conserved, immunodominant CD8 T cell epitopes. Overall, our results demonstrate the ability of influenza protein-containing VLPs to prime IAV-specific CD8 T cell responses that contribute to protection from homo- and heterosubtypic IAV infections. These results further suggest that vaccination strategies focused on the development of cross-protective CD8 T cell responses may contribute to the development of "universal" IAV vaccines.
Collapse
Affiliation(s)
- Emily A Hemann
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
140
|
Long-term protective immunity from an influenza virus-like particle vaccine administered with a microneedle patch. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1433-9. [PMID: 23863506 DOI: 10.1128/cvi.00251-13] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Skin vaccination with influenza virus-like particles (VLPs) using microneedles has been shown to induce protection similar to or better than that induced by intramuscular immunization. In this study, we examined the long-term protective efficacy of influenza (H1N1 A/PR/8/34) VLPs after skin vaccination using microneedle patches coated with the vaccine. Microneedle vaccination of mice in the skin induced 100% protection against lethal challenge infection with influenza A/PR/8/34 virus 14 months after a single vaccine dose. Influenza virus-specific total IgG response and hemagglutination inhibition (HAI) titers were maintained at high levels for over 1 year after microneedle vaccination. Microneedle vaccination also induced substantial levels of lung IgG and IgA antibody responses, and antibody-secreting plasma cells from spleen and bone marrow, as well as conferring effective control of lung viral loads, resulting in complete protection 14 months after vaccination. These strong and long-lasting immune responses were enabled in part by stabilization of the vaccine by formulation with trehalose during microneedle patch fabrication. Administration of the stabilized vaccine using microneedles was especially effective at enabling strong recall responses measured 4 days after lethal virus challenge, including increased HAI and antibody-secreting cells in the spleen and reduced viral titer and inflammatory response in the lung. The results in this study indicate that skin vaccination with VLP vaccine using a microneedle patch provides long-term protection against influenza in mice.
Collapse
|
141
|
Kim MC, Lee JS, Kwon YM, O E, Lee YJ, Choi JG, Wang BZ, Compans RW, Kang SM. Multiple heterologous M2 extracellular domains presented on virus-like particles confer broader and stronger M2 immunity than live influenza A virus infection. Antiviral Res 2013; 99:328-35. [PMID: 23811283 DOI: 10.1016/j.antiviral.2013.06.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 05/17/2013] [Accepted: 06/18/2013] [Indexed: 11/20/2022]
Abstract
The influenza M2 ectodomain (M2e) is poorly immunogenic and has some amino acid changes among isolates from different host species. We expressed a tandem repeat construct of heterologous M2e sequences (M2e5x) derived from human, swine, and avian origin influenza A viruses on virus-like particles (M2e5x VLPs) in a membrane-anchored form. Immunization of mice with M2e5x VLPs induced protective antibodies cross-reactive to antigenically different influenza A viruses and conferred cross protection. Anti-M2e antibodies induced by heterologous M2e5x VLPs showed a wider range of cross reactivity to influenza A viruses at higher levels than those by live virus infection, homologous M2e VLPs, or M2e monoclonal antibody 14C2. Fc receptors were found to be important for mediating protection by immune sera from M2e5x VLP vaccination. The present study provides evidence that heterologous recombinant M2e5x VLPs can be more effective in inducing protective M2e immunity than natural virus infection and further supports an approach for developing an effective universal influenza vaccine.
Collapse
Affiliation(s)
- Min-Chul Kim
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA 30303, USA; Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Liu F, Wu X, Li L, Liu Z, Wang Z. Use of baculovirus expression system for generation of virus-like particles: successes and challenges. Protein Expr Purif 2013; 90:104-16. [PMID: 23742819 PMCID: PMC7128112 DOI: 10.1016/j.pep.2013.05.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/13/2013] [Accepted: 05/15/2013] [Indexed: 11/10/2022]
Abstract
A brief overview of principles and applications of BES. Generation of VLPs using BES. Major properties of BES: promoting generation of VLPs. Bioprocess considerations for generation of VLPs.
The baculovirus expression system (BES) has been one of the versatile platforms for the production of recombinant proteins requiring multiple post-translational modifications, such as folding, oligomerization, phosphorylation, glycosylation, acylation, disulfide bond formation and proteolytic cleavage. Advances in recombinant DNA technology have facilitated application of the BES, and made it possible to express multiple proteins simultaneously in a single infection and to produce multimeric proteins sharing functional similarity with their natural analogs. Therefore, the BES has been used for the production of recombinant proteins and the construction of virus-like particles (VLPs), as well as for the development of subunit vaccines, including VLP-based vaccines. The VLP, which consists of one or more structural proteins but no viral genome, resembles the authentic virion but cannot replicate in cells. The high-quality recombinant protein expression and post-translational modifications obtained with the BES, along with its capacity to produce multiple proteins, imply that it is ideally suited to VLP production. In this article, we critically review the pros and cons of using the BES as a platform to produce both enveloped and non-enveloped VLPs.
Collapse
Affiliation(s)
- Fuxiao Liu
- National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, Shandong 266032, China
| | | | | | | | | |
Collapse
|
143
|
Thompson CM, Petiot E, Lennaertz A, Henry O, Kamen AA. Analytical technologies for influenza virus-like particle candidate vaccines: challenges and emerging approaches. Virol J 2013; 10:141. [PMID: 23642219 PMCID: PMC3655918 DOI: 10.1186/1743-422x-10-141] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/29/2013] [Indexed: 02/08/2023] Open
Abstract
Influenza virus-like particle vaccines are one of the most promising ways to respond to the threat of future influenza pandemics. VLPs are composed of viral antigens but lack nucleic acids making them non-infectious which limit the risk of recombination with wild-type strains. By taking advantage of the advancements in cell culture technologies, the process from strain identification to manufacturing has the potential to be completed rapidly and easily at large scales. After closely reviewing the current research done on influenza VLPs, it is evident that the development of quantification methods has been consistently overlooked. VLP quantification at all stages of the production process has been left to rely on current influenza quantification methods (i.e. Hemagglutination assay (HA), Single Radial Immunodiffusion assay (SRID), NA enzymatic activity assays, Western blot, Electron Microscopy). These are analytical methods developed decades ago for influenza virions and final bulk influenza vaccines. Although these methods are time-consuming and cumbersome they have been sufficient for the characterization of final purified material. Nevertheless, these analytical methods are impractical for in-line process monitoring because VLP concentration in crude samples generally falls out of the range of detection for these methods. This consequently impedes the development of robust influenza-VLP production and purification processes. Thus, development of functional process analytical techniques, applicable at every stage during production, that are compatible with different production platforms is in great need to assess, optimize and exploit the full potential of novel manufacturing platforms.
Collapse
Affiliation(s)
- Christine M Thompson
- National Research Council Canada, Vaccine Program – Human Health therapeutics Portfolio, 6100 Royalmount Avenue, Montreal, Québec H4P 2R2, Canada
- École Polytechnique de Montréal, C.P. 6079, succ. Centre-ville, Montréal, Québec H3C 3A7, Canada
| | - Emma Petiot
- National Research Council Canada, Vaccine Program – Human Health therapeutics Portfolio, 6100 Royalmount Avenue, Montreal, Québec H4P 2R2, Canada
| | - Alexandre Lennaertz
- National Research Council Canada, Vaccine Program – Human Health therapeutics Portfolio, 6100 Royalmount Avenue, Montreal, Québec H4P 2R2, Canada
- École Polytechnique de Montréal, C.P. 6079, succ. Centre-ville, Montréal, Québec H3C 3A7, Canada
| | - Olivier Henry
- École Polytechnique de Montréal, C.P. 6079, succ. Centre-ville, Montréal, Québec H3C 3A7, Canada
| | - Amine A Kamen
- National Research Council Canada, Vaccine Program – Human Health therapeutics Portfolio, 6100 Royalmount Avenue, Montreal, Québec H4P 2R2, Canada
- École Polytechnique de Montréal, C.P. 6079, succ. Centre-ville, Montréal, Québec H3C 3A7, Canada
| |
Collapse
|
144
|
Liu F, Wu X, Li L, Ge S, Liu Z, Wang Z. Virus-like particles: promising platforms with characteristics of DIVA for veterinary vaccine design. Comp Immunol Microbiol Infect Dis 2013; 36:343-52. [PMID: 23561290 DOI: 10.1016/j.cimid.2013.02.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 02/13/2013] [Accepted: 02/25/2013] [Indexed: 11/27/2022]
Abstract
In general, it is difficult to differentiate infected from vaccinated animals through vaccination with conventional vaccines, thereby impeding the serological surveillance of animal diseases. DIVA (differentiating infected from vaccinated animals) vaccine, originally known as marker vaccine, usually based on the absence of at least one immunogenic protein in the vaccine strain, allows DIVA in conjunction with a diagnostic test that detects antibodies against the antigens lacking in the vaccine strain. Virus-like particles (VLPs), composed of one or more structural proteins but no genomes of native viruses, mimic the organization and conformation of authentic virions but have no ability to self-replicate in cells, potentially yielding safer vaccine candidates. Since VLPs containing either monovalent or multivalent antigen can be produced in compliance with the requirements for serological surveillance, the use of VLP-based vaccines plays a promising role in DIVA vaccination strategies against animal diseases. Here, we critically reviewed VLPs and companion diagnostics with properties of DIVA for veterinary vaccine design, and three different VLPs as promising platforms for DIVA vaccination strategies in animals.
Collapse
Affiliation(s)
- Fuxiao Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin 130062, China
| | | | | | | | | | | |
Collapse
|
145
|
Gregory AE, Titball R, Williamson D. Vaccine delivery using nanoparticles. Front Cell Infect Microbiol 2013; 3:13. [PMID: 23532930 PMCID: PMC3607064 DOI: 10.3389/fcimb.2013.00013] [Citation(s) in RCA: 306] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 03/05/2013] [Indexed: 12/16/2022] Open
Abstract
Vaccination has had a major impact on the control of infectious diseases. However, there are still many infectious diseases for which the development of an effective vaccine has been elusive. In many cases the failure to devise vaccines is a consequence of the inability of vaccine candidates to evoke appropriate immune responses. This is especially true where cellular immunity is required for protective immunity and this problem is compounded by the move toward devising sub-unit vaccines. Over the past decade nanoscale size (<1000 nm) materials such as virus-like particles, liposomes, ISCOMs, polymeric, and non-degradable nanospheres have received attention as potential delivery vehicles for vaccine antigens which can both stabilize vaccine antigens and act as adjuvants. Importantly, some of these nanoparticles (NPs) are able to enter antigen-presenting cells by different pathways, thereby modulating the immune response to the antigen. This may be critical for the induction of protective Th1-type immune responses to intracellular pathogens. Their properties also make them suitable for the delivery of antigens at mucosal surfaces and for intradermal administration. In this review we compare the utilities of different NP systems for the delivery of sub-unit vaccines and evaluate the potential of these delivery systems for the development of new vaccines against a range of pathogens.
Collapse
Affiliation(s)
- Anthony E Gregory
- College of Life and Environmental Sciences, University of Exeter Exeter, UK.
| | | | | |
Collapse
|
146
|
Kang SM, Kim MC, Compans RW. Virus-like particles as universal influenza vaccines. Expert Rev Vaccines 2013; 11:995-1007. [PMID: 23002980 DOI: 10.1586/erv.12.70] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Current influenza vaccines are primarily targeted to induce immunity to the influenza virus strain-specific hemagglutinin antigen and are not effective in controlling outbreaks of new pandemic viruses. An approach for developing universal vaccines is to present highly conserved antigenic epitopes in an immunogenic conformation such as virus-like particles (VLPs) together with an adjuvant to enhance the vaccine immunogenicity. In this review, the authors focus on conserved antigenic targets and molecular adjuvants that were presented in VLPs. Conserved antigenic targets that include the hemagglutinin stalk domain, the external domain of influenza M2 and neuraminidase are discussed in addition to molecular adjuvants that are engineered to be incorporated into VLPs in a membrane-anchored form.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| | | | | |
Collapse
|
147
|
Rezaei F, Mirshafiey A, Shahmahmoodi S, Shoja Z, Ghavami N, Mokhtari-Azad T. Influenza Virus-like Particle Containing Two Different Subtypes of Hemagglutinin Confers Protection in Mice Against Lethal Challenge With A/PR8 (H1N1) and A/HK (H3N2) Viruses. IRANIAN RED CRESCENT MEDICAL JOURNAL 2013; 15:75-82. [PMID: 23487492 PMCID: PMC3589784 DOI: 10.5812/ircmj.6252] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/27/2012] [Accepted: 09/24/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND Preventing the seasonal or pandemic outbreak of influenza can be powerful and cost-effective. OBJECTIVES In this study, we constructed a novel virus-like particle (VLP) platform that contains two hemagglutinin (HA) subtypes and evaluated immunogenicity of constructed VLP in mice. MATERIALS AND METHODS This recombinant candidate vaccine model resulted in the expression of two HAs of H1N1 and H3N2 subtypes co-localized within a VLP. Following infection of insect cells with recombinant baculovirus co-expressing H1, H3 and M1 proteins, VLPs with size of 80-120 nm were self-assembled, budding, and released into the insect culture medium. The resulting VLPs which contained two different subtypes of hemagglutinin were purified by ultracentrifugation. The immunogenicity of VLPs was evaluated in mice following immunization. RESULTS Our data showed that vaccination using VLPs elicited robust levels of serum IgG, and viral neutralizing antibodies against A/PR8 (H1N1) and A/HK (H3N2) viruses. Following challenge with lethal dose of A/PR8 (H1N1) and A/HK (H3N2, vaccinated mice were protected, displaying no sign of weight loss and mortality compared to non-vaccinated control mice. CONCLUSIONS VLPs can serve as a promising vaccination strategy to control influenza virus.
Collapse
Affiliation(s)
- Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Shohreh Shahmahmoodi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Zabihollah Shoja
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Nastaran Ghavami
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Talat Mokhtari-Azad, Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, IR Iran. Tel.: +98-2188962343, Fax: +98-2188962343, E-mail:
| |
Collapse
|
148
|
Fast Single-Use VLP Vaccine Productions Based on Insect Cells and the Baculovirus Expression Vector System: Influenza as Case Study. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2013; 138:99-125. [DOI: 10.1007/10_2013_186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
149
|
Abstract
Vaccination has had a major impact on the control of infectious diseases. However, there are still many infectious diseases for which the development of an effective vaccine has been elusive. In many cases the failure to devise vaccines is a consequence of the inability of vaccine candidates to evoke appropriate immune responses. This is especially true where cellular immunity is required for protective immunity and this problem is compounded by the move toward devising sub-unit vaccines. Over the past decade nanoscale size (<1000 nm) materials such as virus-like particles, liposomes, ISCOMs, polymeric, and non-degradable nanospheres have received attention as potential delivery vehicles for vaccine antigens which can both stabilize vaccine antigens and act as adjuvants. Importantly, some of these nanoparticles (NPs) are able to enter antigen-presenting cells by different pathways, thereby modulating the immune response to the antigen. This may be critical for the induction of protective Th1-type immune responses to intracellular pathogens. Their properties also make them suitable for the delivery of antigens at mucosal surfaces and for intradermal administration. In this review we compare the utilities of different NP systems for the delivery of sub-unit vaccines and evaluate the potential of these delivery systems for the development of new vaccines against a range of pathogens.
Collapse
Affiliation(s)
- Anthony E Gregory
- College of Life and Environmental Sciences, University of Exeter Exeter, UK.
| | | | | |
Collapse
|
150
|
Pushko P, Pumpens P, Grens E. Development of Virus-Like Particle Technology from Small Highly Symmetric to Large Complex Virus-Like Particle Structures. Intervirology 2013; 56:141-65. [DOI: 10.1159/000346773] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|