1
|
Cadena-Cruz C, Villarreal Camacho JL, De Ávila-Arias M, Hurtado-Gomez L, Rodriguez A, San-Juan-Vergara H. Respiratory syncytial virus entry mechanism in host cells: A general overview. Mol Microbiol 2023; 120:341-350. [PMID: 37537859 DOI: 10.1111/mmi.15133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 08/05/2023]
Abstract
Respiratory syncytial virus (RSV) is a virus that causes acute respiratory infections in neonates and older adults. To infect host cells, the attachment glycoprotein (G) interacts with a cell surface receptor. This interaction determines the specific cell types that are susceptible to infection. RSV possesses a type I fusion protein F. Type I fusion proteins are metastable when rearrangement of the prefusion F occurs; the fusion peptide is exposed transforming the protein into postfusion form. The transition between the prefusion form and its postfusion form facilitates the viral envelope and the host cell membrane to fuse, enabling the virus to enter the host cell. Understanding the entry mechanism employed by RSV is crucial for developing effective antiviral therapies. In this review, we will discuss the various types of viral fusion proteins and explore the potential entry mechanisms utilized by RSV. A deeper understanding of these mechanisms will provide valuable insights for the development of novel approaches to treat RSV infections.
Collapse
Affiliation(s)
- C Cadena-Cruz
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - J L Villarreal Camacho
- Facultad de Ciencias de la Salud, Programa de Medicina, Universidad Libre Seccional Barranquilla, Barranquilla, Colombia
| | - Marcio De Ávila-Arias
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Leidy Hurtado-Gomez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | - Alexander Rodriguez
- División Ciencias de la Salud, Universidad del Norte Barranquilla, Barranquilla, Colombia
| | | |
Collapse
|
2
|
Blanco JCG, Cullen LM, Kamali A, Sylla FYD, Boukhvalova MS, Morrison TG. Development of Respiratory Syncytial Virus Vaccine Candidates for the Elderly. Viruses 2023; 15:1305. [PMID: 37376605 PMCID: PMC10304043 DOI: 10.3390/v15061305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a significant threat to elderly populations and repeated infections that occur throughout life are poorly protective. To assess the role of prior RSV infections as well as elderly immune senescence on vaccine efficacy, we compared immune responses after virus-like particle (VLP) immunization of elderly cotton rats and young cotton rats, both previously RSV infected, in order to mimic the human population. We show that immunization of RSV-experienced young or elderly animals resulted in the same levels of anti-pre-F IgG, anti-G IgG, neutralizing antibody titers, and protection from challenge indicating that the delivery of F and G proteins in a VLP is equally effective in activation of protective responses in both elderly and young populations. Our results suggest that F and G protein-containing VLPs induce anti-RSV memory established in prior RSV infections equally well in both young and elderly animals and thus can be an effective vaccine for the elderly.
Collapse
Affiliation(s)
| | - Lori M. Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Arash Kamali
- Sigmovir Biosystems Inc., Rockville, MD 20850, USA
| | | | | | - Trudy G. Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
3
|
Virus-like Particle Vaccine Expressing the Respiratory Syncytial Virus Pre-Fusion and G Proteins Confers Protection against RSV Challenge Infection. Pharmaceutics 2023; 15:pharmaceutics15030782. [PMID: 36986643 PMCID: PMC10051362 DOI: 10.3390/pharmaceutics15030782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Respiratory syncytial virus (RSV) causes severe lower respiratory tract disease in children and the elderly. However, there are no effective antiviral drugs or licensed vaccines available for RSV infection. Here, RSV virus-like particle (VLP) vaccines expressing Pre-F, G, or Pre-F and G proteins on the surface of influenza virus matrix protein 1 (M1) were produced using the baculovirus expression system, and their protective efficacy was evaluated in mice. The morphology and successful assembly of VLPs were confirmed by transmission electron microscope (TEM) and Western blot. High levels of serum IgG antibody response were detected in VLP-immunized mice, and significantly higher levels of IgG2a and IgG2b were found in the Pre-F+G VLP immunization group compared to the unimmunized control. Serum-neutralizing activity was higher in the VLP immunization groups compared to the naïve group, with Pre-F+G VLPs demonstrating superior neutralizing activity to the single antigen-expressing VLP groups. Pulmonary IgA and IgG responses were generally comparable across the immunization groups, with VLPs expressing the Pre-F antigen eliciting higher IFN-γ in spleens. The frequencies of eosinophils and IL-4-producing CD4+ T cell populations were substantially lower in the lungs of VLP-immunized mice, with the PreF+G vaccine inducing a significant increase in CD4+ and CD8+ T cells. VLP immunization significantly decreased the viral titer and inflammation in the lungs of mice, with Pre-F+G VLPs conferring the best protection. In conclusion, our present study suggests that the Pre-F+G VLPs could be a potential vaccine candidate against RSV infection.
Collapse
|
4
|
McGinnes Cullen L, Luo B, Wen Z, Zhang L, Durr E, Morrison TG. The Respiratory Syncytial Virus (RSV) G Protein Enhances the Immune Responses to the RSV F Protein in an Enveloped Virus-Like Particle Vaccine Candidate. J Virol 2023; 97:e0190022. [PMID: 36602367 PMCID: PMC9888267 DOI: 10.1128/jvi.01900-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a serious human respiratory pathogen, but no RSV vaccine has been licensed. Many vaccine candidates are focused on the viral F protein since the F protein is more conserved than the viral G protein across RSV strains and serotypes; thus, the F protein is thought more likely to induce a broader range of protection from infection. However, it is the G protein that binds the likely receptor, CX3CR1, in lung ciliated epithelial cells, raising the question of the importance of the G protein in vaccine candidates. Using virus-like particle (VLP) vaccine candidates, we have directly compared VLPs containing only the prefusion F protein (pre-F), only the G protein, or both glycoproteins. We report that VLPs containing both glycoproteins bind to anti-F-protein-specific monoclonal antibodies differently than do VLPs containing only the prefusion F protein. In RSV-naive cotton rats, VLPs assembled with only the pre-F protein stimulated extremely weak neutralizing antibody (NAb) titers, as did VLPs assembled with G protein. However, VLPs assembled with both glycoproteins stimulated quite robust neutralizing antibody titers, induced improved protection of the animals from RSV challenge compared to pre-F VLPs, and induced significantly higher levels of antibodies specific for F protein antigenic site 0, site III, and the AM14 binding site than did VLPs containing only the pre-F protein. These results indicate that assembly of pre-F protein with G protein in VLPs further stabilized the prefusion conformation or otherwise altered the conformation of the F protein, increasing the induction of protective antibodies. IMPORTANCE Respiratory syncytial virus (RSV) results in significant disease in infants, young children, and the elderly. Thus, development of an effective vaccine for these populations is a priority. Most ongoing efforts in RSV vaccine development have focused on the viral fusion (F) protein; however, the importance of the inclusion of G in vaccine candidates is unclear. Here, using virus-like particles (VLPs) assembled with only the F protein, only the G protein, or both glycoproteins, we show that VLPs assembled with both glycoproteins are a far superior vaccine in a cotton rat model compared with VLPs containing only F protein or only G protein. The results show that the presence of G protein in the VLPs influences the conformation of the F protein and the immune responses to F protein, resulting in significantly higher neutralizing antibody titers and better protection from RSV challenge. These results suggest that inclusion of G protein in a vaccine candidate may improve its effectiveness.
Collapse
Affiliation(s)
- Lori McGinnes Cullen
- Department of Microbiology and Physiological Systems, Program in Immunology and Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Bin Luo
- Pharmacology, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Zhiyun Wen
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Lan Zhang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Eberhard Durr
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Trudy G. Morrison
- Department of Microbiology and Physiological Systems, Program in Immunology and Microbiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
5
|
Chen Y, Ding P, Li M, Liu S, Chang Z, Ren D, Li R, Zhang N, Sun X, Zhang G. Spy&IAC enables specific capture of SpyTagged proteins for rapid assembly of plug-and-display nanoparticle vaccines. Int J Biol Macromol 2023; 226:240-253. [PMID: 36509200 DOI: 10.1016/j.ijbiomac.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/29/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
From modular vaccine production to protein assembly on nanoparticles, the SpyCatcher/SpyTag system provides a convenient plug-and-display procedure. Here, we established a general-purpose immunoaffinity chromatography (IAC) method for SpyTagged proteins (Spy&IAC). SpyTags are displayed on the surface of nanoparticles to induce high-affinity monoclonal antibodies, allowing the specific capture of the target protein. Taking the key core antigenic regions of two coronaviruses that are currently more threatened in the field of human and animal diseases, the nucleocapsid (N) protein of SARS-CoV-2 and the COE protein of porcine epidemic diarrhea virus (PEDV) as model proteins, a purification model with SpyTag at the N-terminal or C-terminal expressed in E. coli or mammalian cells was constructed. After the efficient elution of Spy&IAC, the final yield of several proteins is about 3.5-15 mg/L culture, and the protein purity is above 90 %. Purification also preserves the assembly function and immunogenicity of the protein to support subsequent modular assembly and immunization programs. This strategy provides a general tool for the efficient purification of SpyTagged proteins from different expression sources and different tag positions, enabling the production of modular vaccines at lower cost and in a shorter time, which will prepare the public health field for potential pandemic threats.
Collapse
Affiliation(s)
- Yilan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Peiyang Ding
- College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Minghui Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Siyuan Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Zejie Chang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Dongna Ren
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Ruiqi Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Ning Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | - Xueke Sun
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China; College of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
6
|
Chu KB, Quan FS. Respiratory Viruses and Virus-like Particle Vaccine Development: How Far Have We Advanced? Viruses 2023; 15:v15020392. [PMID: 36851606 PMCID: PMC9965150 DOI: 10.3390/v15020392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
With technological advancements enabling globalization, the intercontinental transmission of pathogens has become much easier. Respiratory viruses are one such group of pathogens that require constant monitoring since their outbreak leads to massive public health crises, as exemplified by the influenza virus, respiratory syncytial virus (RSV), and the recent coronavirus disease 2019 (COVID-19) outbreak caused by the SARS-CoV-2. To prevent the transmission of these highly contagious viruses, developing prophylactic tools, such as vaccines, is of considerable interest to the scientific community. Virus-like particles (VLPs) are highly sought after as vaccine platforms for their safety and immunogenicity profiles. Although several VLP-based vaccines against hepatitis B and human papillomavirus have been approved for clinical use by the United States Food and Drug Administration, VLP vaccines against the three aforementioned respiratory viruses are lacking. Here, we summarize the most recent progress in pre-clinical and clinical VLP vaccine development. We also outline various strategies that contributed to improving the efficacy of vaccines against each virus and briefly discuss the stability aspect of VLPs that makes it a highly desired vaccine platform.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence:
| |
Collapse
|
7
|
Su C, Zhong Y, Zhao G, Hou J, Zhang S, Wang B. RSV pre-fusion F protein enhances the G protein antibody and anti-infectious responses. NPJ Vaccines 2022; 7:168. [PMID: 36535957 PMCID: PMC9762623 DOI: 10.1038/s41541-022-00591-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection in children is the most common viral respiratory infection and can cause severe lung damage or death. There is no licensed vaccine for preventing RSV infection. Previously we demonstrated that an RSV vaccine, BARS13, consisting of recombinant G protein from E. coli plus cyclosporine A (CsA) as an immune-modulator, can protect animals from RSV challenge without inducing vaccine-enhanced disease (VED). To maximize the efficacy of such a vaccine, we introduced RSV pre-fusion F protein (pre-F) to form a new vaccine comprised of the pre-F and G proteins with the CsA. Two intramuscular immunizations with the vaccine induced a higher level of neutralizing antibodies against RSV and protected mice from RSV challenge without incurring VED. Interestingly, the addition of the pre-F to the vaccine facilitated anti-G antibody production and protection from RSV infection mainly via induction of antibodies against the central conserved domain (CCD) of the G protein which correlated with blocking the CX3C-CX3CR1 interaction. A 15 amino acid sequence (FP4) within the F2 region of pre-F served as a CD4+ Th epitope to facilitate the anti-G antibody response. Collectively, such a combination of the FP4 peptide with the G protein and CsA provides a novel strategy for developing a safe and maximally effective recombinant G protein-containing RSV vaccine.
Collapse
Affiliation(s)
- Caixia Su
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China
| | - Yiwei Zhong
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China ,Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
| | - Gan Zhao
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou, Jiangsu Province China
| | - Jiawang Hou
- Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou, Jiangsu Province China
| | - Shuren Zhang
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China ,Present Address: Shenzhen Pregene Biopharma Company LTD, Shenzhen, China
| | - Bin Wang
- grid.8547.e0000 0001 0125 2443Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, China ,grid.8547.e0000 0001 0125 2443Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China ,Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China ,Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou, Jiangsu Province China ,grid.411405.50000 0004 1757 8861National Clinical Research Center for Geriatric Diseases, Huashan Hospital, Shanghai, China ,grid.411333.70000 0004 0407 2968Children’s Hospital of Fudan University, Shanghai, China
| |
Collapse
|
8
|
Luo J, Qin H, Lei L, Lou W, Li R, Pan Z. Virus-like particles containing a prefusion-stabilized F protein induce a balanced immune response and confer protection against respiratory syncytial virus infection in mice. Front Immunol 2022; 13:1054005. [PMID: 36578490 PMCID: PMC9792133 DOI: 10.3389/fimmu.2022.1054005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a serious respiratory pathogen in infants and young children worldwide. Currently, no licensed RSV vaccines are available. In this study, we explored stable prefusion conformation virus-like particles (Pre-F VLPs) as RSV vaccine candidates. RSV fusion (F) protein mutants were constructed to form stabilized Pre-F or postfusion (Post-F) configurations. VLPs containing Pre-F or Post-F protein were generated using a recombinant baculovirus (rBV)-insect cell expression system. The assembly and immunological properties of Pre-F or Post-F VLPs were investigated. Pre-F and Post-F VLPs contained antigenic sites Ø and I of pre- and postfusion conformations, respectively. Compared with Post-F VLPs, immunization with Pre-F VLPs elicited upregulation of IFN-γ, IL-2 and IL-10 and downregulation of IL-4 and IL-5 cytokine production in mice. A high percentage of CD25+ Foxp3+ cells or a low percentage of IL-17A-producing cells among CD4+ T cells was observed in the lungs of mice vaccinated with Pre-F VLPs. Importantly, immunization with Pre-F VLPs induced a high level of RSV neutralizing antibody and a balanced immune response, which protected mice against RSV infection without evidence of immunopathology. Our results suggested that Pre-F VLPs generated from rBV-insect cells represent promising RSV vaccine candidates.
Collapse
|
9
|
Chu KB, Lee SH, Kim MJ, Kim AR, Moon EK, Quan FS. Virus-like particles coexpressing the PreF and Gt antigens of respiratory syncytial virus confer protection in mice. Nanomedicine (Lond) 2022; 17:1159-1171. [DOI: 10.2217/nnm-2022-0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: The purpose of this study was to assess the protective efficacy of virus-like particles (VLPs) co-expressing the pre-fusogenic (PreF) and G protein with tandem repeats (Gt) antigens of respiratory syncytial virus (RSV) in mice. Materials & methods: VLP constructs expressing PreF, Gt or both were used to immunize mice, and the protective efficacies were evaluated using antibody responses, neutralizing antibody titers, T-cell responses, histopathological assessment and plaque assay. Results: PreF+Gt VLP immunization elicited strong RSV-specific antibody responses and pulmonary T-cell responses that contributed to lessening virus titer and inflammation. Conclusion: Our findings suggest that coexpressing PreF and Gt antigens elicits better protection than either one alone. This combinatorial approach could assist in future RSV vaccine development.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Su-Hwa Lee
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ah-Ra Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species & Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
10
|
Rama A, Pai A, Rosa Barreto D, Kumar Kannan S, Naha A. Virus-Like particles as a Novel Targeted Drug Delivery Platform for Biomedical Applications. RESEARCH JOURNAL OF PHARMACY AND TECHNOLOGY 2022:2801-2808. [DOI: 10.52711/0974-360x.2022.00468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Virus-Like Particles (VLP) mimics virions immunologically which induces high titers of neutralizing antibodies to conformational epitopes due to the high-density display of epitopes, present multiple proteins which are optimal for uptake by dendritic cells and are assembled in vivo. VLP triggers the immune response of the body against the diseases and is broadly two types like non enveloped VLP’s and Enveloped VLP’s. The present review discusses the production, analysis, and mechanism of action of virus-like particles. Various applications, the Indian Scenario of VLP, Limitations, and future scopes are briefly reviewed and discussed. VLPs imitate authentic viruses in antigenic morphology and offer a stable alternative to attenuated and inactivated viruses in the production of vaccines. It can effectively deliver foreign nucleic acids, proteins, or conjugated compounds to the system, or even to particular types of cells, due to their transducing properties. It retains the ability to infiltrate and render cells useful for a wide range of applications. Used as a tool to increase the immunogenicity of poorly immunogenic antigens, VLP therapeutics can be developed and manufactured in a way that would be sufficiently cheap to be seen globally in many countries. The ability to mass-produce them cost-effectively improves their possibility of being introduced to undeveloped countries.
Collapse
Affiliation(s)
- Annamalai Rama
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Anuja Pai
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Divya Rosa Barreto
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Siva Kumar Kannan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Anup Naha
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
11
|
D’Sa S, Braz Gomes K, Allotey-Babington GL, Boyoglu C, Kang SM, D’Souza MJ. Transdermal Immunization with Microparticulate RSV-F Virus-like Particles Elicits Robust Immunity. Vaccines (Basel) 2022; 10:vaccines10040584. [PMID: 35455333 PMCID: PMC9030121 DOI: 10.3390/vaccines10040584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 01/09/2023] Open
Abstract
No approved vaccines against respiratory syncytial virus (RSV) infections exist to date, due to challenges arising during vaccine development. There is an unmet need to explore novel approaches and a universal strategy to prevent RSV infections. Previous studies have proven the immune efficacy of virus-like particles (VLPs) consisting of RSV fusion (F) protein, yielding a highly immunogenic RSV-F VLP subunit vaccine. In this study, RSV-F VLP (with or without MPL®) was added to a polymer mix and spray-dried, forming microparticles. The formulations were transdermally administered in C57BL/6 mice to evaluate vaccine efficacy. The transdermal delivery of RSV-F VLP + MPL® was more effective in clearing lung viral loads and preventing weight loss after RSV challenge. At the cellular level, MPL® augmented the vaccine response in microparticulate form, which was evidenced by higher serum and lung antibody titers, and lower lung viral titers in the vaccinated groups. These preliminary results validate the effectiveness of the RSV-F VLP microparticulate vaccine via the transdermal route due to its potential to trigger robust immune responses.
Collapse
Affiliation(s)
- Sucheta D’Sa
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (K.B.G.); (G.L.A.-B.); (C.B.)
| | - Kimberly Braz Gomes
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (K.B.G.); (G.L.A.-B.); (C.B.)
| | - Grace Lovia Allotey-Babington
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (K.B.G.); (G.L.A.-B.); (C.B.)
| | - Cemil Boyoglu
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (K.B.G.); (G.L.A.-B.); (C.B.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA;
| | - Martin J. D’Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, Atlanta, GA 30341, USA; (S.D.); (K.B.G.); (G.L.A.-B.); (C.B.)
- Correspondence:
| |
Collapse
|
12
|
Abstract
Human respiratory syncytial virus (RSV) is a negative sense single-stranded RNA virus that can result in epidemics of seasonal respiratory infections. Generally, one of the two genotypes (A and B) predominates in a single season and alternate annually with regional variation. RSV is a known cause of disease and death at both extremes of ages in the pediatric and elderly, as well as immunocompromised populations. The clinical impact of RSV on the hospitalized adults has been recently clarified with the expanded use of multiplex molecular assays. Among adults, RSV can produce a wide range of clinical symptoms due to upper respiratory tract infections potentially leading to severe lower respiratory tract infections, as well as exacerbations of underlying cardiac and lung diseases. While supportive care is the mainstay of therapy, there are currently multiple therapeutic and preventative options under development.
Collapse
Affiliation(s)
- Hannah H Nam
- Department of Infectious Diseases, University of California, Irvine, Orange, California
| | - Michael G Ison
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
13
|
Stephens LM, Varga SM. Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population. Vaccines (Basel) 2021; 9:vaccines9060624. [PMID: 34207770 PMCID: PMC8228432 DOI: 10.3390/vaccines9060624] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is most commonly associated with acute lower respiratory tract infections in infants and children. However, RSV also causes a high disease burden in the elderly that is often under recognized. Adults >65 years of age account for an estimated 80,000 RSV-associated hospitalizations and 14,000 deaths in the United States annually. RSV infection in aged individuals can result in more severe disease symptoms including pneumonia and bronchiolitis. Given the large disease burden caused by RSV in the aged, this population remains an important target for vaccine development. Aging results in lowered immune responsiveness characterized by impairments in both innate and adaptive immunity. This immune senescence poses a challenge when developing a vaccine targeting elderly individuals. An RSV vaccine tailored towards an elderly population will need to maximize the immune response elicited in order to overcome age-related defects in the immune system. In this article, we review the hurdles that must be overcome to successfully develop an RSV vaccine for use in the elderly, and discuss the vaccine candidates currently being tested in this highly susceptible population.
Collapse
Affiliation(s)
- Laura M. Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven M. Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
14
|
Cicconi P, Jones C, Sarkar E, Silva-Reyes L, Klenerman P, de Lara C, Hutchings C, Moris P, Janssens M, Fissette LA, Picciolato M, Leach A, Gonzalez-Lopez A, Dieussaert I, Snape MD. First-in-Human Randomized Study to Assess the Safety and Immunogenicity of an Investigational Respiratory Syncytial Virus (RSV) Vaccine Based on Chimpanzee-Adenovirus-155 Viral Vector-Expressing RSV Fusion, Nucleocapsid, and Antitermination Viral Proteins in Healthy Adults. Clin Infect Dis 2021; 70:2073-2081. [PMID: 31340042 PMCID: PMC7201425 DOI: 10.1093/cid/ciz653] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/19/2019] [Indexed: 11/17/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) disease is a major cause of infant morbidity and mortality. This Phase I, randomized, observer-blind, placebo- and active-controlled study evaluated an investigational vaccine against RSV (ChAd155-RSV) using the viral vector chimpanzee-adenovirus-155, encoding RSV fusion (F), nucleocapsid, and transcription antitermination proteins. Methods Healthy 18–45-year-old adults received ChAd155-RSV, a placebo, or an active control (Bexsero) at Days (D) 0 and 30. An escalation from a low dose (5 × 109 viral particles) to a high dose (5 × 1010 viral particles) occurred after the first 16 participants. Endpoints were solicited/unsolicited and serious adverse events (SAEs), biochemical/hematological parameters, cell-mediated immunogenicity by enzyme-linked immunospot, functional neutralizing antibodies, anti RSV-F immunoglobin (Ig) G, and ChAd155 neutralizing antibodies. Results There were 7 participants who received the ChAd155-RSV low dose, 31 who received the ChAd155-RSV high dose, 19 who received the placebo, and 15 who received the active control. No dose-related toxicity or attributable SAEs at the 1-year follow-up were observed. The RSV-A neutralizing antibodies geometric mean titer ratios (post/pre-immunization) following a high dose were 2.6 (D30) and 2.3 (D60). The ratio of the fold-rise (D0 to D30) in anti-F IgG over the fold-rise in RSV-A–neutralizing antibodies was 1.01. At D7 after the high dose of the study vaccine, the median frequencies of circulating B-cells secreting anti-F antibodies were 133.3/106 (IgG) and 16.7/106 (IgA) in peripheral blood mononuclear cells (PBMCs). The median frequency of RSV-F–specific interferon γ–secreting T-cells after a ChAd155-RSV high dose was 108.3/106 PBMCs at D30, with no increase after the second dose. Conclusions In adults previously naturally exposed to RSV, ChAd155-RSV generated increases in specific humoral and cellular immune responses without raising significant safety concerns. Clinical Trials Registration NCT02491463.
Collapse
Affiliation(s)
- Paola Cicconi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | - Esha Sarkar
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Catherine de Lara
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | - Claire Hutchings
- Nuffield Department of Medicine, University of Oxford, United Kingdom
| | | | | | | | | | | | | | | | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, United Kingdom.,National Institute for Health Research Oxford Biomedical Centre, United Kingdom
| |
Collapse
|
15
|
Kotomina T, Isakova-Sivak I, Stepanova E, Mezhenskaya D, Matyushenko V, Prokopenko P, Sivak K, Kiseleva I, Rudenko L. Neutralizing epitope of the Fusion Protein of Respiratory Syncytial Virus Embedded in the HA Molecule of LAIV Virus is not Sufficient to Prevent RS Virus Pulmonary Replication but Ameliorates Lung Pathology following RSV Infection in Mice. Open Microbiol J 2020. [DOI: 10.2174/1874285802014010147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aims:
To develop experimental bivalent vaccines against influenza and RSV using a cold-adapted LAIV backbone.
Background:
Respiratory syncytial virus (RSV) is a causative agent of bronchiolitis and pneumonia in young children, elderly and immunocompromised adults. No vaccine against RSV has been licensed to date for various reasons. One of the promising platforms for designing RSV vaccine is the use of live attenuated influenza vaccine (LAIV) viruses to deliver RSV epitopes to the respiratory mucosa.
Objective:
To generate recombinant LAIV viruses encoding a neutralizing epitope of the RSV fusion protein and assess their protective potential against both influenza and RSV infections in a mouse model.
Methods:
Reverse genetics methods were used to rescue recombinant LAIV+HA/RSV viruses expressing chimeric hemagglutinins encoding the RSV-F epitope at its N-terminus using two different flexible linkers. BALB/c mice were intranasally immunized with two doses of the recombinant viruses and then challenged with the influenza virus or RSV. The LAIV viral vector and formalin-inactivated RSV (FI-RSV) were included as control vaccines. Protection was assessed by the reduction of virus pulmonary titers. In addition, RSV-induced lung pathology was evaluated by histopathology studies.
Results:
Two rescued chimeric LAIV+HA/RSV viruses were identical to the LAIV vector in terms of replication capacity in vitro and in vivo. The RSV-F neutralizing epitope was successfully expressed only if inserted into the HA molecule via G-linker, but not A-linker. Both chimeric viruses induced high influenza-specific antibody levels and fully protected mice against a lethal influenza challenge virus. However, they induced weak anti-RSV antibody responses which did not prevent RS virus replication upon challenge, and only LAIV-HA+G-RSV variant protected mice against RSV-induced lung pathology.
Conclusion:
Although the designed LAIV-RSV chimeric viruses were unable to neutralize the RS virus pulmonary replication, the LAIV-HA+G-RSV reduced RSV-induced lung pathology and can be considered a promising bivalent vaccine against influenza and RSV infections and warrants its further development.
Collapse
|
16
|
Comparisons of Antibody Populations in Different Pre-Fusion F VLP-Immunized Cotton Rat Dams and Their Offspring. Vaccines (Basel) 2020; 8:vaccines8010133. [PMID: 32197348 PMCID: PMC7157610 DOI: 10.3390/vaccines8010133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/12/2020] [Accepted: 03/14/2020] [Indexed: 01/16/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection poses a significant risk for infants. Since the direct vaccination of infants is problematic, maternal vaccination may provide a safer, more effective approach to their protection. In the cotton rat (CR) model, we have compared the immunization of pregnant CR dams with virus-like particles assembled with the prototype mutation stabilized pre-fusion F protein, DS-Cav1, as well two alternative mutation stabilized pre-fusion proteins (UC-2 F, UC-3 F) and showed that the alternative pre-fusion F VLPs protected the offspring of immunized dams significantly better than DS-Cav1 F VLPs (Blanco, et al. J. Virol. 93: e00914). Here, we have addressed the reasons for this increased protection by characterizing the specificities of antibodies in the sera of both immunized dams and their offspring. The approach was to measure the levels of total anti-pre-F IgG serum antibodies that would block the binding of representative pre-fusion specific monoclonal antibodies to soluble pre-fusion F protein targets. Strikingly, we found that the sera in most offspring of DS-Cav1 F VLP-immunized dams had no mAb D25-blocking antibodies, although their dams had robust levels. In contrast, all offspring of UC-3 F VLP-immunized dams had robust levels of these D25-blocking antibodies. Both sets of pup sera had significant levels of mAb AM14-blocking antibodies, indicating that all pups received maternal antibodies. A lack of mAb D25-blocking antibodies in the offspring of DS-Cav1 F VLP-immunized dams may account for the lower protection of their pups from challenge compared to the offspring of UC-3 F VLP-immunized dams.
Collapse
|
17
|
Conformational Flexibility in Respiratory Syncytial Virus G Neutralizing Epitopes. J Virol 2020; 94:JVI.01879-19. [PMID: 31852779 DOI: 10.1128/jvi.01879-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/10/2019] [Indexed: 02/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a top cause of severe lower respiratory tract disease and mortality in infants and the elderly. Currently, no vaccine or effective treatment exists for RSV. The RSV G glycoprotein mediates viral attachment to cells and contributes to pathogenesis by modulating host immunity through interactions with the human chemokine receptor CX3CR1. Antibodies targeting the RSV G central conserved domain are protective in both prophylactic and postinfection animal models. Here, we describe the crystal structure of the broadly neutralizing human monoclonal antibody 3G12 bound to the RSV G central conserved domain. Antibody 3G12 binds to a conformational epitope composed of highly conserved residues, explaining its broad neutralization activity. Surprisingly, RSV G complexed with 3G12 adopts a distinct conformation not observed in previously described RSV G-antibody structures. Comparison to other structures reveals that the RSV G central conserved domain is flexible and can adopt multiple conformations in the regions flanking the cysteine noose. We also show that restriction of RSV G flexibility with a proline mutation abolishes binding to antibody 3G12 but not antibody 3D3, which recognizes a different conformation of RSV G. Our studies provide new insights for rational vaccine design, indicating the importance of preserving both the global structural integrity of antigens and local conformational flexibility at antigenic sites, which may elicit a more diverse antibody response and broader protection against infection and disease.IMPORTANCE Respiratory syncytial virus (RSV) causes severe respiratory infections in infants, young children, and the elderly, and currently, no licensed vaccine exists. In this study, we describe the crystal structure of the RSV surface glycoprotein G in complex with a broadly neutralizing human monoclonal antibody. The antibody binds to RSV G at a highly conserved region stabilized by two disulfide bonds, but it captures RSV G in a conformation not previously observed, revealing that this region is both structured and flexible. Importantly, our findings provide insight for the design of vaccines that elicit diverse antibodies, which may provide broad protection from infection and disease.
Collapse
|
18
|
Blanco JCG, Fernando LR, Zhang W, Kamali A, Boukhvalova MS, McGinnes-Cullen L, Morrison TG. Alternative Virus-Like Particle-Associated Prefusion F Proteins as Maternal Vaccines for Respiratory Syncytial Virus. J Virol 2019; 93:e00914-19. [PMID: 31511382 PMCID: PMC6854499 DOI: 10.1128/jvi.00914-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/03/2019] [Indexed: 01/09/2023] Open
Abstract
Maternal vaccination may be the most effective and safest approach to the protection of infants from respiratory syncytial virus (RSV) infection, a severe acute lower respiratory tract disease in infants and young children worldwide. We previously compared five different virus-like particle (VLP)-associated, mutation-stabilized prefusion F (pre-F) proteins, including the prototype DS-Cav1 F VLPs. We showed that alternative versions of prefusion F proteins have different conformations and induce different populations of anti-F protein antibodies. Two of these alternative pre-F VLPs, the UC-2 F and UC-3 F VLPs, stimulated in mice higher titers of neutralizing antibodies than DS-Cav1 F VLPs (M. L. Cullen, R. M. Schmidt, M. G. Torres, A. A. Capoferri, et al., Vaccines 7:21-41, 2019, https://doi.org/10.3390/vaccines7010021). Here we describe a comparison of these two pre-F VLPs with DS-Cav1 F VLPs as maternal vaccines in cotton rats and report that UC-3 F VLPs significantly increased the neutralizing antibody (NAb) titers in pregnant dams compared to DS-Cav1 F VLPs. The neutralizing antibody titers in the sera of the offspring of the dams immunized with UC-3 F VLPs were significantly higher than those in the sera of the offspring of dams immunized with DS-Cav1 VLPs. This increase in serum NAb titers translated to a 6- to 40-fold lower virus titer in the lungs of the RSV-challenged offspring of dams immunized with UC-3 F VLPs than in the lungs of the RSV-challenged offspring of dams immunized with DS-Cav1 F VLPs. Importantly, the offspring of UC-3 F VLP-immunized dams showed significant protection from lung pathology and from induction of inflammatory lung cytokine mRNA expression after RSV challenge. Immunization with UC-3 F VLPs also induced durable levels of high-titer neutralizing antibodies in dams.IMPORTANCE Respiratory syncytial virus (RSV) is a significant human pathogen severely impacting neonates and young children, but no vaccine exists to protect this vulnerable population. Furthermore, direct vaccination of neonates is likely ineffective due to the immaturity of their immune system, and neonate immunization is potentially unsafe. Maternal vaccination may be the best and safest approach to the protection of neonates through the passive transfer of maternal neutralizing antibodies in utero to the fetus after maternal immunization. Here we report that immunization of pregnant cotton rats, a surrogate model for human maternal immunization, with novel RSV virus-like particle (VLP) vaccine candidates containing stabilized prefusion RSV F proteins provides significant levels of protection of the offspring of immunized dams from RSV challenge. We also found that antibodies induced by VLPs containing different versions of the prefusion F protein varied by 40-fold in the extent of protection provided to the offspring of vaccinated dams upon RSV challenge.
Collapse
Affiliation(s)
| | | | - Wei Zhang
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | - Arash Kamali
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | | | - Lori McGinnes-Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Trudy G Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
- Program in Microbiology and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
19
|
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. IMMUNITY & AGEING 2019; 16:25. [PMID: 31528180 PMCID: PMC6743147 DOI: 10.1186/s12979-019-0164-9] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
The age-related dysregulation and decline of the immune system-collectively termed "immunosenescence"-has been generally associated with an increased susceptibility to infectious pathogens and poor vaccine responses in older adults. While numerous studies have reported on the clinical outcomes of infected or vaccinated individuals, our understanding of the mechanisms governing the onset of immunosenescence and its effects on adaptive immunity remains incomplete. Age-dependent differences in T and B lymphocyte populations and functions have been well-defined, yet studies that demonstrate direct associations between immune cell function and clinical outcomes in older individuals are lacking. Despite these knowledge gaps, research has progressed in the development of vaccine and adjuvant formulations tailored for older adults in order to boost protective immunity and overcome immunosenescence. In this review, we will discuss the development of vaccines for older adults in light of our current understanding-or lack thereof-of the aging immune system. We highlight the functional changes that are known to occur in the adaptive immune system with age, followed by a discussion of current, clinically relevant pathogens that disproportionately affect older adults and are the central focus of vaccine research efforts for the aging population. We conclude with an outlook on personalized vaccine development for older adults and areas in need of further study in order to improve our fundamental understanding of adaptive immunosenescence.
Collapse
Affiliation(s)
- Stephen N Crooke
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
20
|
Abstract
Human respiratory syncytial virus (RSV) belongs to the recently defined Pneumoviridae family, Orthopneumovirus genus. It is a negative sense, single stranded RNA virus that results in epidemics of respiratory infections that typically peak in the winter in temperate climates and during the rainy season in tropical climates. Generally, one of the two genotypes (A and B) predominates in a single season, alternating annually, although regional variation occurs. RSV is a cause of disease and death in children, older people, and immunocompromised patients, and its clinical effect on adults admitted to hospital is clarified with expanded use of multiplex molecular assays. Among adults, RSV produces a wide range of clinical symptoms including upper respiratory tract infections, severe lower respiratory tract infections, and exacerbations of underlying disease. Here we discuss the latest evidence on the burden of RSV related disease in adults, especially in those with immunocompromise or other comorbidities. We review current therapeutic and prevention options, as well as those in development.
Collapse
Affiliation(s)
- Hannah H Nam
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Michael G Ison
- Division of Infectious Diseases and Organ Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
21
|
Beugeling M, De Zee J, Woerdenbag HJ, Frijlink HW, Wilschut JC, Hinrichs WLJ. Respiratory syncytial virus subunit vaccines based on the viral envelope glycoproteins intended for pregnant women and the elderly. Expert Rev Vaccines 2019; 18:935-950. [PMID: 31446807 DOI: 10.1080/14760584.2019.1657013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Respiratory syncytial virus (RSV) causes high morbidity and mortality rates among infants, young children, and the elderly worldwide. Unfortunately, a safe and effective vaccine is still unavailable. In 1966, a formalin-inactivated RSV vaccine failed and resulted in the death of two young children. This failure shifted research toward the development of subunit-based vaccines for pregnant women (to passively vaccinate infants) and the elderly. Among these subunit-based vaccines, the viral envelope glycoproteins show great potential as antigens. Areas covered: In this review, progress in the development of safe and effective subunit RSV vaccines based on the viral envelope glycoproteins and intended for pregnant women and the elderly, are reviewed and discussed. Studies published in the period 2012-2018 were included. Expert opinion: Researchers are close to bringing safe and effective subunit-based RSV vaccines to the market using the viral envelope glycoproteins as antigens. However, it remains a major challenge to elicit protective immunity, with a formulation that has sufficient (storage) stability. These issues may be overcome by using the RSV fusion protein in its pre-fusion conformation, and by formulating this protein as a dry powder. It may further be convenient to administer this powder via the pulmonary route.
Collapse
Affiliation(s)
- Max Beugeling
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Jildou De Zee
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Herman J Woerdenbag
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Jan C Wilschut
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
22
|
Effect of Previous Respiratory Syncytial Virus Infection on Murine Immune Responses to F and G Protein-Containing Virus-Like Particles. J Virol 2019; 93:JVI.00087-19. [PMID: 30760576 DOI: 10.1128/jvi.00087-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Most individuals are infected with respiratory syncytial virus (RSV) by age two, but infection does not result in long-term protective immunity to subsequent infections. Previous RSV infection may, however, impact responses to an RSV vaccine. The goal of these studies was to explore the effect of previous RSV infection on murine antibody responses to RSV F and G protein-containing virus-like particles (VLP), comparing responses to those resulting from VLP immunization of RSV-naive animals. These studies showed that after RSV infection, immunization with a single dose of VLPs containing a conformation-stabilized prefusion F protein stimulated high titers of neutralizing antibodies (NA), while an immunization with post-F-containing VLPs or a second RSV infection only weakly stimulated NA, even though total anti-F protein IgG antibody levels in both VLP-immunized animals were similar. Furthermore, single pre-F or post-F VLP immunization of animals previously infected (primed) with RSV resulted in total anti-F antibody titers that were 10- to 12-fold higher than titers after a VLP prime and boost of RSV-naive animals or after two consecutive RSV infections. The avidities of serum antibodies as well as numbers of splenic B cells and bone marrow cells after different immunization protocols were also assessed. The combined results show that RSV infection can quite effectively prime animals for the production of protective antibodies that can be efficiently activated by a pre-F VLP boost but not by a post-F VLP boost or a second RSV infection.IMPORTANCE Humans may experience repeated infections caused by the same serotype of respiratory syncytial virus (RSV), in contrast to infections with most other viruses, indicating that immune memory responses to RSV are defective. However, the effects of any residual but nonprotective immunity on responses to RSV vaccines are not clear. This study demonstrates that a VLP vaccine candidate containing a stabilized prefusion F protein can robustly stimulate protective immunity in animals previously infected with RSV, while a second RSV infection or a postfusion F-containing VLP cannot. This result shows that a properly constructed immunogen can be an effective vaccine in animals previously infected with RSV. The results also suggest that the defect in RSV memory is not in the induction of that memory but rather in its activation by a subsequent RSV infection.
Collapse
|
23
|
Cullen LM, Schmidt MR, Torres GM, Capoferri AA, Morrison TG. Comparison of Immune Responses to Different Versions of VLP Associated Stabilized RSV Pre-Fusion F Protein. Vaccines (Basel) 2019; 7:vaccines7010021. [PMID: 30769923 PMCID: PMC6466353 DOI: 10.3390/vaccines7010021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 11/16/2022] Open
Abstract
Efforts to develop a vaccine for respiratory syncytial virus (RSV) have primarily focused on the RSV fusion protein. The pre-fusion conformation of this protein induces the most potent neutralizing antibodies and is the focus of recent efforts in vaccine development. Following the first identification of mutations in the RSV F protein (DS-Cav1 mutant protein) that stabilized the pre-fusion conformation, other mutant stabilized pre-fusion F proteins have been described. To determine if there are differences in alternate versions of stabilized pre-fusion F proteins, we explored the use, as vaccine candidates, of virus-like particles (VLPs) containing five different pre-fusion F proteins, including the DS-Cav1 protein. The expression of these five pre-F proteins, their assembly into VLPs, their pre-fusion conformation stability in VLPs, their reactivity with anti-F monoclonal antibodies, and their induction of immune responses after the immunization of mice, were characterized, comparing VLPs containing the DS-Cav1 pre-F protein with VLPs containing four alternative pre-fusion F proteins. The concentrations of anti-F IgG induced by each VLP that blocked the binding of prototype monoclonal antibodies using two different soluble pre-fusion F proteins as targets were measured. Our results indicate that both the conformation and immunogenicity of alternative VLP associated stabilized pre-fusion RSV F proteins are different from those of DS-Cav1 VLPs.
Collapse
Affiliation(s)
- Lori M Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Madelyn R Schmidt
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Gretel M Torres
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Adam A Capoferri
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | - Trudy G Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01655, USA.
- Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
24
|
Ibrahim A, Odon V, Kormelink R. Plant Viruses in Plant Molecular Pharming: Toward the Use of Enveloped Viruses. FRONTIERS IN PLANT SCIENCE 2019; 10:803. [PMID: 31275344 PMCID: PMC6594412 DOI: 10.3389/fpls.2019.00803] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/04/2019] [Indexed: 05/03/2023]
Abstract
Plant molecular pharming has emerged as a reliable platform for recombinant protein expression providing a safe and low-cost alternative to bacterial and mammalian cells-based systems. Simultaneously, plant viruses have evolved from pathogens to molecular tools for recombinant protein expression, chimaeric viral vaccine production, and lately, as nanoagents for drug delivery. This review summarizes the genesis of viral vectors and agroinfection, the development of non-enveloped viruses for various biotechnological applications, and the on-going research on enveloped plant viruses.
Collapse
|
25
|
Brune KD, Howarth M. New Routes and Opportunities for Modular Construction of Particulate Vaccines: Stick, Click, and Glue. Front Immunol 2018; 9:1432. [PMID: 29997617 PMCID: PMC6028521 DOI: 10.3389/fimmu.2018.01432] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/11/2018] [Indexed: 02/02/2023] Open
Abstract
Vaccines based on virus-like particles (VLPs) can induce potent B cell responses. Some non-chimeric VLP-based vaccines are highly successful licensed products (e.g., hepatitis B surface antigen VLPs as a hepatitis B virus vaccine). Chimeric VLPs are designed to take advantage of the VLP framework by decorating the VLP with a different antigen. Despite decades of effort, there have been few licensed chimeric VLP vaccines. Classic approaches to create chimeric VLPs are either genetic fusion or chemical conjugation, using cross-linkers from lysine on the VLP to cysteine on the antigen. We describe the principles that make these classic approaches challenging, in particular for complex, full-length antigens bearing multiple post-translational modifications. We then review recent advances in conjugation approaches for protein-based non-enveloped VLPs or nanoparticles, to overcome such challenges. This includes the use of strong non-covalent assembly methods (stick), unnatural amino acids for bio-orthogonal chemistry (click), and spontaneous isopeptide bond formation by SpyTag/SpyCatcher (glue). Existing applications of these methods are outlined and we critically consider the key practical issues, with particular insight on Tag/Catcher plug-and-display decoration. Finally, we highlight the potential for modular particle decoration to accelerate vaccine generation and prepare for pandemic threats in human and veterinary realms.
Collapse
Affiliation(s)
- Karl D Brune
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Mark Howarth
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
26
|
Blanco JCG, Pletneva LM, McGinnes-Cullen L, Otoa RO, Patel MC, Fernando LR, Boukhvalova MS, Morrison TG. Efficacy of a respiratory syncytial virus vaccine candidate in a maternal immunization model. Nat Commun 2018; 9:1904. [PMID: 29765035 PMCID: PMC5953919 DOI: 10.1038/s41467-018-04216-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 04/12/2018] [Indexed: 01/10/2023] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of bronchiolitis in infants. Maternal immunization is an option to increase maternal antibody levels and protect infants from infection. Here we assess the efficacy of virus-like particle (VLP) vaccine candidates containing stabilized pre-fusion (pre-F) or post-fusion (post-F) conformations of the RSV F protein and the attachment RSV G protein in a maternal immunization model using cotton rats. VLP vaccines containing RSV F and G proteins strongly boost pre-existing RSV immunity in dams preventing their perinatal drop in immunity. Boosting is stronger for the pre-F VLP than for the post-F VLP or purified subunit F protein vaccines, giving an advantage on mothers’ protection. VLP immunization of dams provides significant protection to pups from RSV challenge and reduced pulmonary inflammation. Collectively, our results show that a VLP vaccine with RSV F and G proteins is safe and effective for maternal and adult vaccination. RSV infection is a major cause of bronchiolitis in infants and maternal vaccination is a potential preventive option. Here, Blanco et al. show efficacy of a Newcastle disease virus-based virus-like particle vaccine candidate in naive and pre-exposed cotton rat dams and their offspring.
Collapse
Affiliation(s)
- Jorge C G Blanco
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA.
| | - Lioubov M Pletneva
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Lori McGinnes-Cullen
- Department of Microbiology and Physiological Systems, Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Raymonde O Otoa
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Mira C Patel
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Lurds R Fernando
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Marina S Boukhvalova
- Sigmovir Biosystems Inc., 9610 Medical Center Drive, Suite 100, Rockville, MD, 20850, USA
| | - Trudy G Morrison
- Department of Microbiology and Physiological Systems, Program of Immunology and Microbiology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| |
Collapse
|
27
|
Clark CM, Guerrero-Plata A. Respiratory Syncytial Virus Vaccine Approaches: a Current Overview. CURRENT CLINICAL MICROBIOLOGY REPORTS 2017; 4:202-207. [PMID: 30009126 PMCID: PMC6040676 DOI: 10.1007/s40588-017-0074-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE OF REVIEW Respiratory syncytial virus (RSV) is a global human pathogen responsible for lower respiratory tract infections (LRTI). While RSV infection is innocuous in healthy adults, it is the leading cause of infant hospitalization for respiratory tract infection. Nearly everyone shows evidence of an RSV infection by the age of 3. However, there is still not a vaccine commercially available. This review will provide an update on the clinical and preclinical vaccine studies and different approaches to prevent RSV infection. RECENT FINDINGS Novel vaccine approaches that induce protection against RSV without enhancement of respiratory tract disease. SUMMARY Recent technological approaches have led to generation of different strategies to prevent RSV infection, including live attenuated, chimeric, and subunit vaccines, virus-like particles, and nanoparticles. These vaccine approaches represent promising candidates towards an efficient RSV vaccine that effectively protects infants, children, and adults.
Collapse
Affiliation(s)
- Carolyn M Clark
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|