1
|
Hu H, Zhang C. Conjugation of Multiple Proteins Onto the Surface of PLGA/Lipid Hybrid Nanoparticles. J Biomed Mater Res A 2024. [PMID: 39420678 DOI: 10.1002/jbm.a.37807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024]
Abstract
Nanoparticles are increasingly being used in the development of vaccines for disease prevention or treatment. Recent research has demonstrated that conjugating a protein onto the surface of nanoparticles can significantly increase its immunogenicity. Considering various pathogens that threaten human health, multivalent vaccines are often desirable. Up to now, nanoparticle-based vaccines are mostly limited to one protein per nanoparticle. No research has been conducted to explore the possibility of conjugating more than one protein onto the surface of a nanoparticle. Here we developed a specific conjugation strategy to conjugate multiple proteins to the PLGA/lipid hybrid nanoparticle surface. The maleimide-thiol Michael addition, Aizde-DBCO (Dibenzocyclooctyne), and TCO (trans-cycloctene)-Tetrazine click chemistry were employed to conjugate three different proteins, subunit keyhole limpet hemocyanin (sKLH), Ovalbumin (OVA), and cross-reactive material 197 (CRM197), to the surface of PLGA/lipid hybrid nanoparticles (hNPs). The successful results of this study pave the way for developing multivalent vaccines against different pathogens.
Collapse
Affiliation(s)
- He Hu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, Virginia, USA
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
2
|
Chen YA, Shen YS, Fang CY, Chan TT, Wu SR, Wang JR, Wu SC, Liu CC. Enhanced production of recombinant coxsackievirus A16 using a serum-free HEK293A suspension culture system for bivalent enterovirus vaccine development. Vaccine X 2024; 20:100559. [PMID: 39364390 PMCID: PMC11447303 DOI: 10.1016/j.jvacx.2024.100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
Coxsackievirus A16 (CVA16) is one of the primary pathogens that causes hand, foot, and mouth disease (HFMD) in young children. In previous studies, CVA16 vaccine development has encountered several challenges, such as inefficient replication of the CVA16 virus in present culture systems, the induction of only mild neutralizing antibody titers, and neutralizing antibodies induced by certain vaccine candidates that are unable to protect against CVA16 viral challenge. In this study, we constructed a DNA-launched CVA16 infectious clone (CVA16ic) based on the genomic sequence of the CVA16 N5079 strain to minimize interference from viral quasispecies. The biochemical properties of this CVA16ic strain were similar to those of its parental strain. Serum-free HEK293A suspension cells, which produced higher virus titers than Vero cells, were demonstrated to improve CVA16 production yields. In addition, our study showed that inactivated EV-A71 antigens could enhance the immunogenicity of inactivated CVA16 mature/full particles (F-particles), suggesting that a bivalent CVA16 and EV-A71 vaccine may be an effective strategy for CVA16 vaccine development. These findings are expected to provide novel strategies and accelerate the development of bivalent HFMD vaccines.
Collapse
Affiliation(s)
- Yi-An Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Yu-Sheng Shen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Chih-Yeu Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Ting-Ting Chan
- School of Dentistry & Institute of Oral Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Rung Wu
- School of Dentistry & Institute of Oral Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chyi Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| |
Collapse
|
3
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
4
|
Hou Y, Chen M, Bian Y, Hu Y, Chuan J, Zhong L, Zhu Y, Tong R. Insights into vaccines for elderly individuals: from the impacts of immunosenescence to delivery strategies. NPJ Vaccines 2024; 9:77. [PMID: 38600250 PMCID: PMC11006855 DOI: 10.1038/s41541-024-00874-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Immunosenescence increases the risk and severity of diseases in elderly individuals and leads to impaired vaccine-induced immunity. With aging of the global population and the emerging risk of epidemics, developing adjuvants and vaccines for elderly individuals to improve their immune protection is pivotal for healthy aging worldwide. Deepening our understanding of the role of immunosenescence in vaccine efficacy could accelerate research focused on optimizing vaccine delivery for elderly individuals. In this review, we analyzed the characteristics of immunosenescence at the cellular and molecular levels. Strategies to improve vaccination potency in elderly individuals are summarized, including increasing the antigen dose, preparing multivalent antigen vaccines, adding appropriate adjuvants, inhibiting chronic inflammation, and inhibiting immunosenescence. We hope that this review can provide a review of new findings with regards to the impacts of immunosenescence on vaccine-mediated protection and inspire the development of individualized vaccines for elderly individuals.
Collapse
Affiliation(s)
- Yingying Hou
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Min Chen
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuan Bian
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yuan Hu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Junlan Chuan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Zhong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Yuxuan Zhu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
5
|
Ga E, Kang JA, Hwang J, Moon S, Choi J, Bae E, Seol H, Mun Y, Song D, Jeong DG, Na W. Assessment of the immune interference effects of multivalent vaccine for influenza epidemic strain in 2022-2023 and evaluation of its efficacy. Heliyon 2024; 10:e28326. [PMID: 38532995 PMCID: PMC10963641 DOI: 10.1016/j.heliyon.2024.e28326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
The various strains of influenza virus cause respiratory symptoms in humans every year and annual vaccinations are recommended. Due to its RNA-type genes and segmented state, it belongs to a virus that mutates frequently with antigenic drift and shift, giving rise to various strains. Each year, the World Health Organization identifies the epidemic strains and operates a global surveillance system to suggest the viral composition for the influenza vaccine. Influenza viruses, which have multiple viral strains, are produced in the format of multivalent vaccine. However, the multivalent vaccine has a possibility of causing immune interference by introducing multiple strain-specific antigens in a single injection. Therefore, evaluating immune interference phenomena is essential when assessing multivalent vaccines. In this study, the protective ability and immunogenicity of multivalent and monovalent vaccines were evaluated in mice to assess immune interference in the multivalent vaccine. Monovalent and multivalent vaccines were manufactured using the latest strain of the 2022-2023 seasonal influenza virus selected by the World Health Organization. The protective abilities of both types of vaccines were tested through hemagglutination inhibition test. The immunogenicity of multivalent and monovalent vaccines were tested through enzyme-linked immunosorbent assay to measure the cellular and humoral immunity expression rates. As a result of the protective ability and immunogenicity test, higher level of virus neutralizing ability and greater amount of antibodies in both IgG1 and IgG2 were confirmed in the multivalent vaccine. No immune interference was found to affect the protective capacity and immune responses of the multivalent vaccines.
Collapse
Affiliation(s)
- Eulhae Ga
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Jung-Ah Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Jaehyun Hwang
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Suyun Moon
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Jaeseok Choi
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Eunseo Bae
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, South Korea
- College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, South Korea
| | - Hyein Seol
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Yubin Mun
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Daesub Song
- College of Veterinary Medicine, Seoul National University, Gwanak-ro, Seoul, 08826, South Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, South Korea
| | - Woonsung Na
- Department of Oral Microbiology and Immunology and Dental Research Institute, School of Dentistry, Seoul National University, Daehak-ro 101 (Yeongeon-dong), Jongno-gu, Seoul, 03080, South Korea
| |
Collapse
|
6
|
Clever S, Limpinsel L, Meyer zu Natrup C, Schünemann LM, Beythien G, Rosiak M, Hülskötter K, Gregor KM, Tuchel T, Kalodimou G, Freudenstein A, Kumar S, Baumgärtner W, Sutter G, Tscherne A, Volz A. Single MVA-SARS-2-ST/N Vaccination Rapidly Protects K18-hACE2 Mice against a Lethal SARS-CoV-2 Challenge Infection. Viruses 2024; 16:417. [PMID: 38543782 PMCID: PMC10974247 DOI: 10.3390/v16030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
The sudden emergence of SARS-CoV-2 demonstrates the need for new vaccines that rapidly protect in the case of an emergency. In this study, we developed a recombinant MVA vaccine co-expressing SARS-CoV-2 prefusion-stabilized spike protein (ST) and SARS-CoV-2 nucleoprotein (N, MVA-SARS-2-ST/N) as an approach to further improve vaccine-induced immunogenicity and efficacy. Single MVA-SARS-2-ST/N vaccination in K18-hACE2 mice induced robust protection against lethal respiratory SARS-CoV-2 challenge infection 28 days later. The protective outcome of MVA-SARS-2-ST/N vaccination correlated with the activation of SARS-CoV-2-neutralizing antibodies (nABs) and substantial amounts of SARS-CoV-2-specific T cells especially in the lung of MVA-SARS-2-ST/N-vaccinated mice. Emergency vaccination with MVA-SARS-2-ST/N just 2 days before lethal SARS-CoV-2 challenge infection resulted in a delayed onset of clinical disease outcome in these mice and increased titers of nAB or SARS-CoV-2-specific T cells in the spleen and lung. These data highlight the potential of a multivalent COVID-19 vaccine co-expressing S- and N-protein, which further contributes to the development of rapidly protective vaccination strategies against emerging pathogens.
Collapse
Affiliation(s)
- Sabrina Clever
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Leonard Limpinsel
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Christian Meyer zu Natrup
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Lisa-Marie Schünemann
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Malgorzata Rosiak
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Katharina Manuela Gregor
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Tamara Tuchel
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| | - Georgia Kalodimou
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Astrid Freudenstein
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Satendra Kumar
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (G.B.); (M.R.); (K.H.); (K.M.G.); (W.B.)
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Alina Tscherne
- Division of Virology, Department of Veterinary Sciences, LMU Munich, 85764 Oberschleißheim, Germany; (L.L.); (G.K.); (A.F.); (S.K.); (G.S.); (A.T.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hanover, Germany; (S.C.); (C.M.z.N.); (L.-M.S.)
| |
Collapse
|
7
|
Moriel DG, Piccioli D, Raso MM, Pizza M. The overlooked bacterial pandemic. Semin Immunopathol 2024; 45:481-491. [PMID: 38078911 PMCID: PMC11136754 DOI: 10.1007/s00281-023-00997-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/28/2023] [Indexed: 05/30/2024]
Abstract
The COVID-19 pandemic had a significant economic and health impact worldwide. It also reinforced the misperception that only viruses can pose a threat to human existence, overlooking that bacteria (e.g., plague and cholera) have severely haunted and shaped the course of human civilization. While the world is preparing for the next viral pandemic, it is again overlooking a silent one: antimicrobial resistance (AMR). This review proposes to show the impact of bacterial infections on civilization to remind the pandemic potential. The work will also discuss a few examples of how bacteria can mutate risking global spread and devastating outcomes, the effect on the global burden, and the prophylactic and therapeutic measures. Indeed, AMR is dramatically increasing and if the trend is not reversed, it has the potential to quickly turn into the most important health problem worldwide.
Collapse
Affiliation(s)
- Danilo Gomes Moriel
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy
| | - Diego Piccioli
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy
| | | | | |
Collapse
|
8
|
Xue W, Li T, Gu Y, Li S, Xia N. Molecular engineering tools for the development of vaccines against infectious diseases: current status and future directions. Expert Rev Vaccines 2023. [PMID: 37339445 DOI: 10.1080/14760584.2023.2227699] [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: 04/17/2023] [Accepted: 06/16/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION The escalating global changes have fostered conditions for the expansion and transmission of diverse biological factors, leading to the rise of emerging and reemerging infectious diseases. Complex viral infections, such as COVID-19, influenza, HIV, and Ebola, continue to surface, necessitating the development of effective vaccine technologies. AREAS COVERED This review article highlights recent advancements in molecular biology, virology, and genomics that have propelled the design and development of innovative molecular tools. These tools have promoted new vaccine research platforms and directly improved vaccine efficacy. The review summarizes the cutting-edge molecular engineering tools used in creating novel vaccines and explores the rapidly expanding molecular tools landscape and potential directions for future vaccine development. EXPERT OPINION The strategic application of advanced molecular engineering tools can address conventional vaccine limitations, enhance the overall efficacy of vaccine products, promote diversification in vaccine platforms, and form the foundation for future vaccine development. Prioritizing safety considerations of these novel molecular tools during vaccine development is crucial.
Collapse
Affiliation(s)
- Wenhui Xue
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen, China
| | - Tingting Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen, China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen, China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Life Sciences, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Xiamen University, Xiamen, China
- Xiang an Biomedicine Laboratory, Xiamen, China
- The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen, China
| |
Collapse
|
9
|
Rando HM, Lordan R, Kolla L, Sell E, Lee AJ, Wellhausen N, Naik A, Kamil JP, Gitter A, Greene CS. The Coming of Age of Nucleic Acid Vaccines during COVID-19. mSystems 2023; 8:e0092822. [PMID: 36861992 PMCID: PMC10134841 DOI: 10.1128/msystems.00928-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics: the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within 2 weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last 2 decades in particular and suggest a new era in vaccines against emerging pathogens. IMPORTANCE The SARS-CoV-2 pandemic has caused untold damage globally, presenting unusual demands on but also unique opportunities for vaccine development. The development, production, and distribution of vaccines are imperative to saving lives, preventing severe illness, and reducing the economic and social burdens caused by the COVID-19 pandemic. Although vaccine technologies that provide the DNA or RNA sequence of an antigen had never previously been approved for use in humans, they have played a major role in the management of SARS-CoV-2. In this review, we discuss the history of these vaccines and how they have been applied to SARS-CoV-2. Additionally, given that the evolution of new SARS-CoV-2 variants continues to present a significant challenge in 2022, these vaccines remain an important and evolving tool in the biomedical response to the pandemic.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
| | - Ronan Lordan
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Likhitha Kolla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amruta Naik
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jeremy P. Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| | - COVID-19 Review Consortium
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
DEC-205 receptor targeted poly(lactic-co-glycolic acid) nanoparticles containing Eucommia ulmoides polysaccharide enhances the immune response of foot-and-mouth disease vaccine in mice. Int J Biol Macromol 2023; 227:576-589. [PMID: 36549613 DOI: 10.1016/j.ijbiomac.2022.12.158] [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: 06/08/2022] [Revised: 11/24/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
Nanoparticles targeting the DEC-205 receptor were found to induce antigen-specific protective immune response. When the delivery system carries both antigens and immunomodulators, it can maximize the expected therapeutic effect of the drug and induce effective humoral and cellular immune responses to antigens.In this study, we encapsulated the Eucommia ulmoides Oliv. polysaccharides (EUPS) into PLGA nanoparticles (NPs) and conjugated it with anti-CD205 monoclonal Ab (MAb) to produce a DEC-205 receptor targeted PLGA nanoparticles (anti-DEC-205-EUPS-PLGA NPs). The physicochemical characteristics and adjuvant activity of the above NPs were evaluated in vitro and in vivo. In the in vitro setting, 200 μg·mL-1 anti-DEC-205-EUPS-PLGA could improve the proliferation of DCs and promote their antigen up-take activity. In the in vivo setting, anti-DEC-205-EUPS-PLGA NPs remarkably controlled the release of drug and antigen to induce sustained immune responses and up-regulated the levels of FMDV-specific IgG antibodies, promoted the cytotoxic activity of CTLs and NK cells, and improved the proliferation of splenocytes. Moreover, the anti-DEC-205-EUPS-PLGA NPs facilitated the maturation of DCs. The above data indicated that anti-DEC-205-EUPS-PLGA NPs employed as an targeted adjuvant induced the humoral and cellular immune activity by promoting the maturation of DCs. These findings may provide a new insight onto the development of vaccine adjuvants.
Collapse
|
11
|
Rando HM, Lordan R, Kolla L, Sell E, Lee AJ, Wellhausen N, Naik A, Kamil JP. The Coming of Age of Nucleic Acid Vaccines during COVID-19. ARXIV 2023:arXiv:2210.07247v2. [PMID: 36263086 PMCID: PMC9580386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In the 21st century, several emergent viruses have posed a global threat. Each pathogen has emphasized the value of rapid and scalable vaccine development programs. The ongoing SARS-CoV-2 pandemic has made the importance of such efforts especially clear. New biotechnological advances in vaccinology allow for recent advances that provide only the nucleic acid building blocks of an antigen, eliminating many safety concerns. During the COVID-19 pandemic, these DNA and RNA vaccines have facilitated the development and deployment of vaccines at an unprecedented pace. This success was attributable at least in part to broader shifts in scientific research relative to prior epidemics; the genome of SARS-CoV-2 was available as early as January 2020, facilitating global efforts in the development of DNA and RNA vaccines within two weeks of the international community becoming aware of the new viral threat. Additionally, these technologies that were previously only theoretical are not only safe but also highly efficacious. Although historically a slow process, the rapid development of vaccines during the COVID-19 crisis reveals a major shift in vaccine technologies. Here, we provide historical context for the emergence of these paradigm-shifting vaccines. We describe several DNA and RNA vaccines and in terms of their efficacy, safety, and approval status. We also discuss patterns in worldwide distribution. The advances made since early 2020 provide an exceptional illustration of how rapidly vaccine development technology has advanced in the last two decades in particular and suggest a new era in vaccines against emerging pathogens.
Collapse
Affiliation(s)
- Halie M Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America; Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Center for Health AI, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America; Department of Biomedical Informatics, University of Colorado Anschutz School of Medicine, Aurora, Colorado, United States of America
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5158, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA
| | - Likhitha Kolla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elizabeth Sell
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alexandra J Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Amruta Naik
- Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Jeremy P Kamil
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
12
|
Cooper A, Sidaway A, Chandrashekar A, Latta E, Chakraborty K, Yu J, McMahan K, Giffin V, Manickam C, Kroll K, Mosher M, Reeves RK, Gam R, Arthofer E, Choudhry M, Henley T, Barouch DH. A genetically engineered, stem-cell-derived cellular vaccine. Cell Rep Med 2022; 3:100843. [PMID: 36480934 PMCID: PMC9727836 DOI: 10.1016/j.xcrm.2022.100843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/19/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022]
Abstract
Despite rapid clinical translation of COVID-19 vaccines in response to the global pandemic, an opportunity remains for vaccine technology innovation to address current limitations and meet challenges of inevitable future pandemics. We describe a universal vaccine cell (UVC) genetically engineered to mimic natural physiological immunity induced upon viral infection of host cells. Cells engineered to express the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike as a representative viral antigen induce robust neutralizing antibodies in immunized non-human primates. Similar titers generated in this established non-human primate (NHP) model have translated into protective human neutralizing antibody levels in SARS-CoV-2-vaccinated individuals. Animals vaccinated with ancestral spike antigens and subsequently challenged with SARS-CoV-2 Delta variant in a heterologous challenge have an approximately 3 log decrease in viral subgenomic RNA in the lungs. This cellular vaccine is designed as a scalable cell line with a modular poly-antigenic payload, allowing for rapid, large-scale clinical manufacturing and use in an evolving viral variant environment.
Collapse
Affiliation(s)
| | | | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | | | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Victoria Giffin
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kyle Kroll
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew Mosher
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Rihab Gam
- Intima Bioscience, Inc., New York, NY, USA
| | | | - Modassir Choudhry
- Praesidium Bioscience, Inc., New York, NY, USA; Intima Bioscience, Inc., New York, NY, USA
| | - Tom Henley
- Praesidium Bioscience, Inc., New York, NY, USA; Intima Bioscience, Inc., New York, NY, USA.
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
13
|
Chen J, Deng JC, Goldstein DR. How aging impacts vaccine efficacy: known molecular and cellular mechanisms and future directions. Trends Mol Med 2022; 28:1100-1111. [PMID: 36216643 PMCID: PMC9691569 DOI: 10.1016/j.molmed.2022.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 01/26/2023]
Abstract
Aging leads to a gradual dysregulation of immune functions, one consequence of which is reduced vaccine efficacy. In this review, we discuss several key contributing factors to the age-related decline in vaccine efficacy, such as alterations within the lymph nodes where germinal center (GC) reactions take place, alterations in the B cell compartment, alterations in the T cell compartment, and dysregulation of innate immune pathways. Additionally, we discuss several methods currently used in vaccine development to bolster vaccine efficacy in older adults. This review highlights the multifactorial defects that impair vaccine responses with aging.
Collapse
Affiliation(s)
- Judy Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jane C Deng
- Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA; Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Daniel R Goldstein
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA; Program in Immunology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
14
|
Cheong HC, Cheok YY, Chan YT, Sulaiman S, Looi CY, Alshanon AF, Hassan J, Abubakar S, Wong WF. Zika Virus Vaccine: The Current State of Affairs and Challenges Posed by Antibody-Dependent Enhancement Reaction. Viral Immunol 2022; 35:586-596. [PMID: 36301533 DOI: 10.1089/vim.2022.0082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Heng Choon Cheong
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yee Teng Chan
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sofiah Sulaiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Ahmed F. Alshanon
- Center of Biotechnology Researches, University of Al-Nahrain, Baghdad, Iraq
| | - Jamiyah Hassan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sazaly Abubakar
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Tropical Infectious Diseases Research and Educational Center (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Bockstal V, Shukarev G, McLean C, Goldstein N, Bart S, Gaddah A, Anumenden D, Stoop JN, Marit de Groot A, Pau MG, Hendriks J, De Rosa SC, Cohen KW, McElrath MJ, Callendret B, Luhn K, Douoguih M, Robinson C. First-in-human study to evaluate safety, tolerability, and immunogenicity of heterologous regimens using the multivalent filovirus vaccines Ad26.Filo and MVA-BN-Filo administered in different sequences and schedules: A randomized, controlled study. PLoS One 2022; 17:e0274906. [PMID: 36197845 PMCID: PMC9534391 DOI: 10.1371/journal.pone.0274906] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/22/2022] [Indexed: 11/05/2022] Open
Abstract
Background Though clinically similar, Ebola virus disease and Marburg virus disease are caused by different viruses. Of the 30 documented outbreaks of these diseases in sub-Saharan Africa, eight were major outbreaks (≥200 cases; five caused by Zaire ebolavirus [EBOV], two by Sudan ebolavirus [SUDV], and one by Marburg virus [MARV]). Our purpose is to develop a multivalent vaccine regimen protecting against each of these filoviruses. This first-in-human study assessed the safety and immunogenicity of several multivalent two-dose vaccine regimens that contain Ad26.Filo and MVA-BN-Filo. Methods Ad26.Filo combines three vaccines encoding the glycoprotein (GP) of EBOV, SUDV, and MARV. MVA-BN-Filo is a multivalent vector encoding EBOV, SUDV, and MARV GPs, and Taï Forest nucleoprotein. This Phase 1, randomized, double-blind, placebo-controlled study enrolled healthy adults (18–50 years) into four groups, randomized 5:1 (active:placebo), to assess different Ad26.Filo and MVA-BN-Filo vaccine directionality and administration intervals. The primary endpoint was safety; immune responses against EBOV, SUDV, and MARV GPs were also assessed. Results Seventy-two participants were randomized, and 60 (83.3%) completed the study. All regimens were well tolerated with no deaths or vaccine-related serious adverse events (AEs). The most frequently reported solicited local AE was injection site pain/tenderness. Solicited systemic AEs most frequently reported were headache, fatigue, chills, and myalgia; most solicited AEs were Grade 1–2. Solicited/unsolicited AE profiles were similar between regimens. Twenty-one days post-dose 2, 100% of participants on active regimen responded to vaccination and exhibited binding antibodies against EBOV, SUDV, and MARV GPs; neutralizing antibody responses were robust against EBOV (85.7–100%), but lower against SUDV (35.7–100%) and MARV (0–57.1%) GPs. An Ad26.Filo booster induced a rapid further increase in humoral responses. Conclusion This study demonstrates that heterologous two-dose vaccine regimens with Ad26.Filo and MVA-BN-Filo are well tolerated and immunogenic in healthy adults. ClinicalTrials.gov NCT02860650.
Collapse
Affiliation(s)
- Viki Bockstal
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Georgi Shukarev
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Chelsea McLean
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
- * E-mail:
| | - Neil Goldstein
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Stephan Bart
- Optimal Research, LLC, Rockville, Maryland, United States of America
| | - Auguste Gaddah
- Janssen Infectious Diseases and Vaccines, Beerse, Belgium
| | | | - Jeroen N. Stoop
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | | | - Maria G. Pau
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Jenny Hendriks
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kristen W. Cohen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | | | - Kerstin Luhn
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Macaya Douoguih
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| | - Cynthia Robinson
- Janssen Infectious Diseases and Vaccines, Leiden, The Netherlands
| |
Collapse
|
16
|
Kumar S, Basu M, Ghosh P, Ansari A, Ghosh MK. COVID-19: Clinical status of vaccine development to date. Br J Clin Pharmacol 2022; 89:114-149. [PMID: 36184710 PMCID: PMC9538545 DOI: 10.1111/bcp.15552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/08/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-induced COVID-19 is a complicated disease. Clinicians are continuously facing difficulties to treat infected patients using the principle of repurposing of drugs as no specific drugs are available to treat COVID-19. To minimize the severity and mortality, global vaccination is the only hope as a potential preventive measure. After a year-long global research and clinical struggle, 165 vaccine candidates have been developed and some are currently still in the pipeline. A total of 28 candidate vaccines have been approved for use and the remainder are in different phases of clinical trials. In this comprehensive report, the authors aim to demonstrate, classify and provide up-to-date clinical trial status of all the vaccines discovered to date and specifically focus on the approved candidates. Finally, the authors specifically focused on the vaccination of different types of medically distinct populations.
Collapse
Affiliation(s)
- Sunny Kumar
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| | - Malini Basu
- Department of MicrobiologyDhruba Chand Halder CollegeIndia
| | - Pratyasha Ghosh
- Department of Economics, Bethune CollegeUniversity of CalcuttaKolkataIndia
| | - Aafreen Ansari
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder DivisionCouncil of Scientific and Industrial Research‐Indian Institute of Chemical Biology (CSIR‐IICB), TRUE CampusKolkataIndia
| |
Collapse
|
17
|
Kumar V, Kumar S, Sharma PC. Recent advances in the vaccine development for the prophylaxis of SARS Covid-19. Int Immunopharmacol 2022; 111:109175. [PMID: 35994853 PMCID: PMC9381430 DOI: 10.1016/j.intimp.2022.109175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-caused Coronavirus Disease 2019 (COVID-19) is currently a global pandemic that has wreaked havoc on public health, lives, and the global economy. The present COVID-19 outbreak has put pressure on the scientific community to develop medications and vaccinations to combat COVID-19. However, according to highly optimistic forecasts, we could not have a COVID-19 vaccine until September 2020. This is due to the fact that a successful COVID-19 vaccine will necessitate a careful validation of effectiveness and adverse reactivity given that the target vaccine population includes high-risk people over 60, particularly those with severe co-morbid conditions, frontline healthcare professionals, and those involved in essential industrial sectors. For passive immunization, which is being considered for Covid-19, there are several platforms for vaccine development, each with its own advantages and disadvantages. The COVID-19 pandemic, which is arguably the deadliest in the last 100 years after the Spanish flu, necessitates a swift assessment of the various approaches for their ability to incite protective immunity and safety to prevent unintended immune potentiation, which is crucial to the pathogenesis of this virus. Considering the pandemic's high fatality rate and rapid spread, an efficient vaccination is critical for its management. As a result, academia, industry, and government are collaborating in unprecedented ways to create and test a wide range of vaccinations. In this review, we summarize the Covid-19 vaccine development initiatives, recent trends, difficulties, comparison between traditional vaccines development and Covid-19 vaccines development also listed the approved/authorized, phase-3 and pre-clinical trials Covid-19 vaccines in different countries.
Collapse
Affiliation(s)
- Vipul Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Sahil Kumar
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India.
| | - Prabodh Chander Sharma
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| |
Collapse
|
18
|
Deng S, Liang H, Chen P, Li Y, Li Z, Fan S, Wu K, Li X, Chen W, Qin Y, Yi L, Chen J. Viral Vector Vaccine Development and Application during the COVID-19 Pandemic. Microorganisms 2022; 10:microorganisms10071450. [PMID: 35889169 PMCID: PMC9317404 DOI: 10.3390/microorganisms10071450] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/08/2022] [Accepted: 07/15/2022] [Indexed: 12/14/2022] Open
Abstract
With the accumulation of mutations in SARS-CoV-2 and the continuous emergence of new variants, the importance of developing safer and effective vaccines has become more prominent in combating the COVID-19 pandemic. Both traditional and genetically engineered vaccines have contributed to the prevention and control of the pandemic. However, in recent years, the trend of vaccination research has gradually transitioned from traditional to genetically engineered vaccines, with the development of viral vector vaccines attracting increasing attention. Viral vector vaccines have several unique advantages compared to other vaccine platforms. The spread of Omicron has also made the development of intranasal viral vector vaccines more urgent, as the infection site of Omicron is more prominent in the upper respiratory tract. Therefore, the present review focuses on the development of viral vector vaccines and their application during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Shaofeng Deng
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China;
- State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hui Liang
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
| | - Pin Chen
- Oriental Fortune Capital Post-Doctoral Innovation Center, Shenzhen 518055, China;
- Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuwan Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Zhaoyao Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Yuwei Qin
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (L.Y.); (J.C.)
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, No. 483 Wushan Road, Tianhe District, Guangzhou 510642, China; (H.L.); (Y.L.); (Z.L.); (S.F.); (K.W.); (X.L.); (W.C.); (Y.Q.)
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangzhou 510642, China
- Correspondence: (L.Y.); (J.C.)
| |
Collapse
|
19
|
Mahroum N, Lavine N, Ohayon A, Seida R, Alwani A, Alrais M, Zoubi M, Bragazzi NL. COVID-19 Vaccination and the Rate of Immune and Autoimmune Adverse Events Following Immunization: Insights From a Narrative Literature Review. Front Immunol 2022; 13:872683. [PMID: 35865539 PMCID: PMC9294236 DOI: 10.3389/fimmu.2022.872683] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
Despite their proven efficacy and huge contribution to the health of humankind, vaccines continue to be a source of concern for some individuals around the world. Vaccinations against COVID-19 increased the number of distressed people and intensified their distrust, particularly as the pandemic was still emerging and the populations were encouraged to be vaccinated under various slogans like "back to normal life" and "stop coronavirus", goals which are still to be achieved. As fear of vaccination-related adverse events following immunization (AEFIs) is the main reason for vaccine hesitancy, we reviewed immune and autoimmune AEFIs in particular, though very rare, as the most worrisome aspect of the vaccines. Among others, autoimmune AEFIs of the most commonly administered COVID-19 vaccines include neurological ones such as Guillain-Barre syndrome, transverse myelitis, and Bell's palsy, as well as myocarditis. In addition, the newly introduced notion related to COVID-19 vaccines, "vaccine-induced immune thrombotic thrombocytopenia/vaccine-induced prothrombotic immune thrombotic thrombocytopenia" (VITT/VIPITT)", is of importance as well. Overviewing recent medical literature while focusing on the major immune and autoimmune AEFIs, demonstrating their rate of occurrence, presenting the cases reported, and their link to the specific type of COVID-19 vaccines represented the main aim of our work. In this narrative review, we illustrate the different vaccine types in current use, their associated immune and autoimmune AEFIs, with a focus on the 3 main COVID-19 vaccines (BNT162b2, mRNA-1273, and ChAdOx1). While the rate of AEFIs is extremely low, addressing the issue in this manner, in our opinion, is the best strategy for coping with vaccine hesitancy.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel
| | - Noy Lavine
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel
- St. George School of Medicine, University of London, London, United Kingdom
| | - Aviran Ohayon
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel
- St. George School of Medicine, University of London, London, United Kingdom
| | - Ravend Seida
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Abdulkarim Alwani
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mahmoud Alrais
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Magdi Zoubi
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| |
Collapse
|
20
|
Jasrotia R, Dhanjal DS, Bhardwaj S, Sharma P, Chopra C, Singh R, Kumar A, Mubayi A, Kumar D, Kumar R, Goyal A. Nanotechnology based vaccines: Cervical cancer management and perspectives. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
21
|
Kim NY, Son WR, Lee MH, Choi HS, Choi JY, Song YJ, Yu CH, Song DH, Hur GH, Jeong ST, Hong SY, Shin YK, Shin S. A multipathogen DNA vaccine elicits protective immune responses against two class A bioterrorism agents, anthrax and botulism. Appl Microbiol Biotechnol 2022; 106:1531-1542. [PMID: 35141866 PMCID: PMC8979915 DOI: 10.1007/s00253-022-11812-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/17/2022]
Abstract
Abstract
The potential use of biological agents has become a major public health concern worldwide. According to the CDC classification, Bacillus anthracis and Clostridium botulinum, the bacterial pathogens that cause anthrax and botulism, respectively, are considered to be the most dangerous potential biological agents. Currently, there is no licensed vaccine that is well suited for mass immunization in the event of an anthrax or botulism epidemic. In the present study, we developed a dual-expression system-based multipathogen DNA vaccine that encodes the PA-D4 gene of B. anthracis and the HCt gene of C. botulinum. When the multipathogen DNA vaccine was administered to mice and guinea pigs, high level antibody responses were elicited against both PA-D4 and HCt. Analysis of the serum IgG subtype implied a combined Th1/Th2 response to both antigens, but one that was Th2 skewed. In addition, immunization with the multipathogen DNA vaccine induced effective neutralizing antibody activity against both PA-D4 and HCt. Finally, the protection efficiency of the multipathogen DNA vaccine was determined by sequential challenge with 10 LD50 of B. anthracis spores and 10 LD50 of botulinum toxin, or vice versa, and the multipathogen DNA vaccine provided higher than 50% protection against lethal challenge with both high-risk biothreat agents. Our studies suggest the strategy used for this anthrax-botulinum multipathogen DNA vaccine as a prospective approach for developing emergency vaccines that can be immediately distributed on a massive scale in response to a biothreat emergency or infectious disease outbreak.
Key points • A novel multipathogen DNA vaccine was constructed against anthrax and botulism. • Robust immune responses were induced following vaccination. • Suggests a potential vaccine development strategy against biothreat agents. |
Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00253-022-11812-6.
Collapse
Affiliation(s)
- Na Young Kim
- R&D Center, ABION Inc., Seoul, Republic of Korea
| | - Won Rak Son
- R&D Center, ABION Inc., Seoul, Republic of Korea
| | - Min Hoon Lee
- R&D Center, ABION Inc., Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | | | | | - Young Jo Song
- The 4th R&D Institute, Agency for Defense Development, Daejeon, Republic of Korea
| | - Chi Ho Yu
- The 4th R&D Institute, Agency for Defense Development, Daejeon, Republic of Korea
| | - Dong Hyun Song
- The 4th R&D Institute, Agency for Defense Development, Daejeon, Republic of Korea
| | - Gyeung Haeng Hur
- The 4th R&D Institute, Agency for Defense Development, Daejeon, Republic of Korea
| | - Seong Tae Jeong
- The 4th R&D Institute, Agency for Defense Development, Daejeon, Republic of Korea
| | - Sung Youl Hong
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young Kee Shin
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sungho Shin
- Bio-MAX/N-Bio, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
MCMV-based vaccine vectors expressing full-length viral proteins provide long-term humoral immune protection upon a single-shot vaccination. Cell Mol Immunol 2022; 19:234-244. [PMID: 34992275 PMCID: PMC8739032 DOI: 10.1038/s41423-021-00814-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 11/25/2021] [Indexed: 11/15/2022] Open
Abstract
Global pandemics caused by influenza or coronaviruses cause severe disruptions to public health and lead to high morbidity and mortality. There remains a medical need for vaccines against these pathogens. CMV (cytomegalovirus) is a β-herpesvirus that induces uniquely robust immune responses in which remarkably large populations of antigen-specific CD8+ T cells are maintained for a lifetime. Hence, CMV has been proposed and investigated as a novel vaccine vector for expressing antigenic peptides or proteins to elicit protective cellular immune responses against numerous pathogens. We generated two recombinant murine CMV (MCMV) vaccine vectors expressing hemagglutinin (HA) of influenza A virus (MCMVHA) or the spike protein of severe acute respiratory syndrome coronavirus 2 (MCMVS). A single injection of MCMVs expressing either viral protein induced potent neutralizing antibody responses, which strengthened over time. Importantly, MCMVHA-vaccinated mice were protected from illness following challenge with the influenza virus, and we excluded that this protection was due to the effects of memory T cells. Conclusively, we show here that MCMV vectors induce not only long-term cellular immunity but also humoral responses that provide long-term immune protection against clinically relevant respiratory pathogens.
Collapse
|
23
|
Koda S, Zhu XQ, Zheng KY, Yan C. Molecular Mechanisms of Clonorchis sinensis-Host Interactions and Implications for Vaccine Development. Front Cell Dev Biol 2022; 9:781768. [PMID: 35118069 PMCID: PMC8804234 DOI: 10.3389/fcell.2021.781768] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Infections caused by Clonorchis sinensis remain a significant public health challenge for both humans and animals, causing pyogenic cholangitis, cholelithiasis, cholecystitis, biliary fibrosis, and even cholangiocarcinoma. However, the strategies used by the parasite and the immunological mechanisms used by the host have not yet been fully understood. With the advances in technologies and the accumulated knowledge of host-parasite interactions, many vaccine candidates against liver flukes have been investigated using different strategies. In this review, we explore and analyze in-depth the immunological mechanisms involved in the pathogenicity of C. sinensis. We highlight the different mechanisms by which the parasite interacts with its host to induce immune responses. All together, these data will allow us to have a better understanding of molecular mechansism of host-parasite interactions, which may shed lights on the development of an effective vaccine against C. sinensis.
Collapse
Affiliation(s)
- Stephane Koda
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Kui-Yang Zheng, ; Chao Yan,
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Kui-Yang Zheng, ; Chao Yan,
| |
Collapse
|
24
|
Malburet C, Leclercq L, Cotte JF, Thiebaud J, Cottet H. Separation of three strains of polio virus by capillary zone electrophoresis and study of their interaction with aluminum oxyhydroxide. J Chromatogr A 2022; 1667:462838. [DOI: 10.1016/j.chroma.2022.462838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 10/19/2022]
|
25
|
Lee JC, Janda KD. Development of effective therapeutics for polysubstance use disorders. Curr Opin Chem Biol 2021; 66:102105. [PMID: 34936944 DOI: 10.1016/j.cbpa.2021.102105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/11/2021] [Accepted: 11/18/2021] [Indexed: 11/18/2022]
Abstract
Traditional pharmacotherapies for substance use disorders have focused on mono-substance abuse. However, recent epidemiological studies have found polysubstance use disorders (PUD) are becoming more prevalent and the abuse of adulterated drugs has led to increasing unintentional overdose deaths. Unfortunately, there are no approved pharmacological agents for PUD. Hence, a therapeutic model of interest to address this growing epidemic is immunopharmacotherapy, where individuals are inoculated with conjugate vaccines formulated with haptens that mimic the drug of abuse. These conjugate vaccines have demonstrated significant therapeutic potential against mono-substance abuse, thus recent studies have applied this model to address PUD. This review presents immunopharmacotherapeutic advancements against polysubstance abuse and discusses necessary developments for conjugate vaccines in order to effectively treat this unaddressed epidemic.
Collapse
Affiliation(s)
- Jinny Claire Lee
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States
| | - Kim D Janda
- Department of Chemistry, Department of Immunology and Microbial Science, The Skaggs Institute for Chemical Biology, The WIRM Institute for Research & Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, United States.
| |
Collapse
|
26
|
Ortega-Rivera O, Shin MD, Chen A, Beiss V, Moreno-Gonzalez MA, Lopez-Ramirez MA, Reynoso M, Wang H, Hurst BL, Wang J, Pokorski JK, Steinmetz NF. Trivalent Subunit Vaccine Candidates for COVID-19 and Their Delivery Devices. J Am Chem Soc 2021; 143:14748-14765. [PMID: 34490778 PMCID: PMC8442557 DOI: 10.1021/jacs.1c06600] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 02/06/2023]
Abstract
The COVID-19 pandemic highlights the need for platform technologies enabling rapid development of vaccines for emerging viral diseases. The current vaccines target the SARS-CoV-2 spike (S) protein and thus far have shown tremendous efficacy. However, the need for cold-chain distribution, a prime-boost administration schedule, and the emergence of variants of concern (VOCs) call for diligence in novel SARS-CoV-2 vaccine approaches. We studied 13 peptide epitopes from SARS-CoV-2 and identified three neutralizing epitopes that are highly conserved among the VOCs. Monovalent and trivalent COVID-19 vaccine candidates were formulated by chemical conjugation of the peptide epitopes to cowpea mosaic virus (CPMV) nanoparticles and virus-like particles (VLPs) derived from bacteriophage Qβ. Efficacy of this approach was validated first using soluble vaccine candidates as solo or trivalent mixtures and subcutaneous prime-boost injection. The high thermal stability of our vaccine candidates allowed for formulation into single-dose injectable slow-release polymer implants, manufactured by melt extrusion, as well as microneedle (MN) patches, obtained through casting into micromolds, for prime-boost self-administration. Immunization of mice yielded high titers of antibodies against the target epitope and S protein, and data confirms that antibodies block receptor binding and neutralize SARS-CoV and SARS-CoV-2 against infection of human cells. We present a nanotechnology vaccine platform that is stable outside the cold-chain and can be formulated into delivery devices enabling single administration or self-administration. CPMV or Qβ VLPs could be stockpiled, and epitopes exchanged to target new mutants or emergent diseases as the need arises.
Collapse
Affiliation(s)
- Oscar
A. Ortega-Rivera
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Matthew D. Shin
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Angela Chen
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Veronique Beiss
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Miguel A. Moreno-Gonzalez
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Miguel A. Lopez-Ramirez
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Maria Reynoso
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322, United States
| | - Hong Wang
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Brett L. Hurst
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
- Institute
for Antiviral Research, Utah State University, Logan, Utah 84322, United States
| | - Joseph Wang
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Jonathan K. Pokorski
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| | - Nicole F. Steinmetz
- Department
of NanoEngineering, Center for Nano-ImmunoEngineering, Institute for Materials
Discovery and Design, Department of Bioengineering, Department of Radiology, and Moores Cancer Center, University of California−San Diego, La Jolla, California 92039, United States
| |
Collapse
|
27
|
Benazzato C, Russo FB, Beltrão-Braga PCB. An update on preclinical pregnancy models of Zika virus infection for drug and vaccine discovery. Expert Opin Drug Discov 2021; 17:19-25. [PMID: 34461793 DOI: 10.1080/17460441.2021.1973999] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Congenital Zika syndrome is caused by Zika virus (ZIKV) infection during pregnancy and can culminate in structural and neurological defects in the fetus, including a spectrum of symptoms such as brain calcifications, hydrocephalus, holoprosencephaly, lissencephaly, ventriculomegaly, and microcephaly. Using animal models to study ZIKV infection during pregnancy represents a critical tool for understanding ZIKV pathophysiology, drug testing, vaccine development, and prevention of vertical transmission. AREAS COVERED In this review, the authors cover state-of-the-art preclinical pregnancy models of ZIKV infection for drug discovery and vaccine development to prevent vertical transmission. EXPERT OPINION The discovery of drugs against ZIKV infection represents an urgent necessity, and until now, no effective drug that can prevent the effects of vertical transmission has been tested in humans. Even after six years of the ZIKV outbreak in Brazil, no drugs or vaccines have been approved for use in humans. In part, this failure could be related to the lack of translatability from available preclinical models to humans.
Collapse
Affiliation(s)
- Cecilia Benazzato
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo, Brazil
| | - Fabiele Baldino Russo
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo, Brazil.,Scientific Plataform Pasteur/USP, São Paulo, Brazil
| | - Patricia Cristina Baleeiro Beltrão-Braga
- Disease Modeling Laboratory at Department of Microbiology, Institute of Biomedical Sciences, São Paulo, Brazil.,Scientific Plataform Pasteur/USP, São Paulo, Brazil
| |
Collapse
|
28
|
Barreat JGN, Katzourakis A. Paleovirology of the DNA viruses of eukaryotes. Trends Microbiol 2021; 30:281-292. [PMID: 34483047 DOI: 10.1016/j.tim.2021.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022]
Abstract
Paleovirology is the study of ancient viruses and how they have coevolved with their hosts. An increasingly detailed understanding of the diversity, origins, and evolution of the DNA viruses of eukaryotes has been obtained through the lens of paleovirology in recent years. Members of multiple viral families have been found integrated in the genomes of eukaryotes, providing a rich fossil record to study. These elements have extended our knowledge of exogenous viral diversity, host ranges, and the timing of viral evolution, and are revealing the existence of entire new families of eukaryotic integrating dsDNA viruses and transposons. Future work in paleovirology will continue to provide insights into antiviral immunity, viral diversity, and potential applications, and reveal other secrets of the viral world.
Collapse
Affiliation(s)
| | - Aris Katzourakis
- Department of Zoology, University of Oxford, Oxford, OX1 3SY, UK.
| |
Collapse
|
29
|
Safini N, Bamouh Z, Hamdi J, Jazouli M, Tadlaoui KO, El Harrak M. In-vitro and in-vivo study of the interference between Rift Valley fever virus (clone 13) and Sheeppox/Limpy Skin disease viruses. Sci Rep 2021; 11:12395. [PMID: 34117312 PMCID: PMC8196192 DOI: 10.1038/s41598-021-91926-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 05/17/2021] [Indexed: 11/09/2022] Open
Abstract
Viral interference is a common occurrence that has been reported in cell culture in many cases. In the present study, viral interference between two capripox viruses (sheeppox SPPV and lumpy skin disease virus LSDV in cattle) with Rift Valley fever virus (RVFV) was investigated in vitro and in their natural hosts, sheep and cattle. A combination of SPPV/RVFV and LSDV/RVFV was used to co-infect susceptible cells and animals to detect potential competition. In-vitro interference was evaluated by estimating viral infectivity and copies of viral RNA by a qPCR during three serial passages in cell cultures, whereas in-vivo interference was assessed through antibody responses to vaccination. When lamb testis primary cells were infected with the mixture of capripox and RVFV, the replication of both SPPV and LSDV was inhibited by RVFV. In animals, SPPV/RVFV or LSDV/RVFV combinations inhibited the replication SPPV and LSDV and the antibody response following vaccination. The combined SPPV/RVFV did not protect sheep after challenging with the virulent strain of SPPV and the LSDV/RVFV did not induce interferon Gamma to LSDV, while immunological response to RVFV remain unaffected. Our goal was to assess this interference response to RVFV/capripoxviruses’ coinfection in order to develop effective combined live-attenuated vaccines as a control strategy for RVF and SPP/LSD diseases. Our findings indicated that this approach was not suitable for developing a combined SPPV/LSDV/RVFV vaccine candidate because of interference of replication and the immune response among these viruses.
Collapse
Affiliation(s)
- N Safini
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco.
| | - Z Bamouh
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco
| | - J Hamdi
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco
| | - M Jazouli
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco
| | - K O Tadlaoui
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco
| | - M El Harrak
- R&D Virology, MCI Santé Animale, Lot. 157, Z I, Sud-Ouest (ERAC), B.P. 278, 28810, Mohammedia, Morocco
| |
Collapse
|
30
|
Sharma A, Ahmed S, Kaur J, Chawla R, Rejeeth C. Exploring status of emergency drugs and vaccine development in Covid-19 pandemic: an update. Virusdisease 2021; 32:198-210. [PMID: 33969152 PMCID: PMC8096892 DOI: 10.1007/s13337-021-00684-5] [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: 10/16/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
COVID-19 outburst initiated from the city of Wuhan in China in December 2019 and has been declared as a public health emergency of international concern. This pandemic portrays a spectrum of clinical complications, primarily affecting the respiratory system reported to be caused by a pathogen SARS-CoV-2 belonging to the family of beta coronavirus. Currently, the main objective of the government authorities of all affected countries and research organizations is to find a potential solution in the form of a pharmacological intervention or a vaccination to eradicate the disease and to have a long-term solution to deal with the pandemic. A multitude of anti-viral regimens is proposed based on the repurposing of currently available drugs for other issues or the compassionate use of drugs to immediately control and optimize the healthcare facilities. Meanwhile, a number of agencies are proposing new drug candidates to recreate the possibility of treating the disease. Similarly, a lot of research work is going on worldwide for the development of vaccination. Currently, a good number of candidates has finally reached the borders of Clinical Trials and are expected to be launched as soon as possible. However, the regulatory framework and authorization of these candidates is the most significant aspect of the whole scenario, which needs a specific set of time for validation purposes. The present work is widely focused on the general aspects of COVID-19 and the regulatory landscape for the emergency authorization of repurposed drug candidates, compassionate use of drugs, investigational entities, and vaccine development worldwide. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13337-021-00684-5.
Collapse
Affiliation(s)
- Alok Sharma
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab 142001 India
| | - Suhail Ahmed
- Department of Pharmacognosy, ISF College of Pharmacy, Moga, Punjab 142001 India
| | - Jasleen Kaur
- Department of Pharmacology and Toxicology, NIPER-Kolkata-700054, Kolkata, India
| | - Rakesh Chawla
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab 151203 India
| | - Chandrababu Rejeeth
- Laboratory of Bio-Process and Engineering, Department of Biochemistry, School of Bioscience, Periyar University, Salem, 636011 India
| |
Collapse
|
31
|
Zhan W, Muhuri M, Tai PWL, Gao G. Vectored Immunotherapeutics for Infectious Diseases: Can rAAVs Be The Game Changers for Fighting Transmissible Pathogens? Front Immunol 2021; 12:673699. [PMID: 34046041 PMCID: PMC8144494 DOI: 10.3389/fimmu.2021.673699] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
Conventional vaccinations and immunotherapies have encountered major roadblocks in preventing infectious diseases like HIV, influenza, and malaria. These challenges are due to the high genomic variation and immunomodulatory mechanisms inherent to these diseases. Passive transfer of broadly neutralizing antibodies may offer partial protection, but these treatments require repeated dosing. Some recombinant viral vectors, such as those based on lentiviruses and adeno-associated viruses (AAVs), can confer long-term transgene expression in the host after a single dose. Particularly, recombinant (r)AAVs have emerged as favorable vectors, given their high in vivo transduction efficiency, proven clinical efficacy, and low immunogenicity profiles. Hence, rAAVs are being explored to deliver recombinant antibodies to confer immunity against infections or to diminish the severity of disease. When used as a vaccination vector for the delivery of antigens, rAAVs enable de novo synthesis of foreign proteins with the conformation and topology that resemble those of natural pathogens. However, technical hurdles like pre-existing immunity to the rAAV capsid and production of anti-drug antibodies can reduce the efficacy of rAAV-vectored immunotherapies. This review summarizes rAAV-based prophylactic and therapeutic strategies developed against infectious diseases that are currently being tested in pre-clinical and clinical studies. Technical challenges and potential solutions will also be discussed.
Collapse
Affiliation(s)
- Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Manish Muhuri
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Phillip W. L. Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States
- VIDE Program, University of Massachusetts Medical School, Worcester, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
32
|
Structure-Based and Rational Design of a Hepatitis C Virus Vaccine. Viruses 2021; 13:v13050837. [PMID: 34063143 PMCID: PMC8148096 DOI: 10.3390/v13050837] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/11/2022] Open
Abstract
A hepatitis C virus (HCV) vaccine is a critical yet unfulfilled step in addressing the global disease burden of HCV. While decades of research have led to numerous clinical and pre-clinical vaccine candidates, these efforts have been hindered by factors including HCV antigenic variability and immune evasion. Structure-based and rational vaccine design approaches have capitalized on insights regarding the immune response to HCV and the structures of antibody-bound envelope glycoproteins. Despite successes with other viruses, designing an immunogen based on HCV glycoproteins that can elicit broadly protective immunity against HCV infection is an ongoing challenge. Here, we describe HCV vaccine design approaches where immunogens were selected and optimized through analysis of available structures, identification of conserved epitopes targeted by neutralizing antibodies, or both. Several designs have elicited immune responses against HCV in vivo, revealing correlates of HCV antigen immunogenicity and breadth of induced responses. Recent studies have elucidated the functional, dynamic and immunological features of key regions of the viral envelope glycoproteins, which can inform next-generation immunogen design efforts. These insights and design strategies represent promising pathways to HCV vaccine development, which can be further informed by successful immunogen designs generated for other viruses.
Collapse
|
33
|
Butkovich N, Li E, Ramirez A, Burkhardt AM, Wang SW. Advancements in protein nanoparticle vaccine platforms to combat infectious disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1681. [PMID: 33164326 PMCID: PMC8052270 DOI: 10.1002/wnan.1681] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
Infectious diseases are a major threat to global human health, yet prophylactic treatment options can be limited, as safe and efficacious vaccines exist only for a fraction of all diseases. Notably, devastating diseases such as acquired immunodeficiency syndrome (AIDS) and coronavirus disease of 2019 (COVID-19) currently do not have vaccine therapies. Conventional vaccine platforms, such as live attenuated vaccines and whole inactivated vaccines, can be difficult to manufacture, may cause severe side effects, and can potentially induce severe infection. Subunit vaccines carry far fewer safety concerns due to their inability to cause vaccine-based infections. The applicability of protein nanoparticles (NPs) as vaccine scaffolds is promising to prevent infectious diseases, and they have been explored for a number of viral, bacterial, fungal, and parasitic diseases. Many types of protein NPs exist, including self-assembling NPs, bacteriophage-derived NPs, plant virus-derived NPs, and human virus-based vectors, and these particular categories will be covered in this review. These vaccines can elicit strong humoral and cellular immune responses against specific pathogens, as well as provide protection against infection in a number of animal models. Furthermore, published clinical trials demonstrate the promise of applying these NP vaccine platforms, which include bacteriophage-derived NPs, in addition to multiple viral vectors that are currently used in the clinic. The continued investigations of protein NP vaccine platforms are critical to generate safer alternatives to current vaccines, advance vaccines for diseases that currently lack effective prophylactic therapies, and prepare for the rapid development of new vaccines against emerging infectious diseases. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Nina Butkovich
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Enya Li
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Aaron Ramirez
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
| | - Amanda M. Burkhardt
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089 USA
| | - Szu-Wen Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA 92697 USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92697 USA
| |
Collapse
|
34
|
Bezbaruah R, Borah P, Kakoti BB, Al-Shar’I NA, Chandrasekaran B, Jaradat DMM, Al-Zeer MA, Abu-Romman S. Developmental Landscape of Potential Vaccine Candidates Based on Viral Vector for Prophylaxis of COVID-19. Front Mol Biosci 2021; 8:635337. [PMID: 33937326 PMCID: PMC8082173 DOI: 10.3389/fmolb.2021.635337] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2, SARS-CoV-2, arose at the end of 2019 as a zoonotic virus, which is the causative agent of the novel coronavirus outbreak COVID-19. Without any clear indications of abatement, the disease has become a major healthcare threat across the globe, owing to prolonged incubation period, high prevalence, and absence of existing drugs or vaccines. Development of COVID-19 vaccine is being considered as the most efficient strategy to curtail the ongoing pandemic. Following publication of genetic sequence of SARS-CoV-2, globally extensive research and development work has been in progress to develop a vaccine against the disease. The use of genetic engineering, recombinant technologies, and other computational tools has led to the expansion of several promising vaccine candidates. The range of technology platforms being evaluated, including virus-like particles, peptides, nucleic acid (DNA and RNA), recombinant proteins, inactivated virus, live attenuated viruses, and viral vectors (replicating and non-replicating) approaches, are striking features of the vaccine development strategies. Viral vectors, the next-generation vaccine platforms, provide a convenient method for delivering vaccine antigens into the host cell to induce antigenic proteins which can be tailored to arouse an assortment of immune responses, as evident from the success of smallpox vaccine and Ervebo vaccine against Ebola virus. As per the World Health Organization, till January 22, 2021, 14 viral vector vaccine candidates are under clinical development including 10 nonreplicating and four replicating types. Moreover, another 39 candidates based on viral vector platform are under preclinical evaluation. This review will outline the current developmental landscape and discuss issues that remain critical to the success or failure of viral vector vaccine candidates against COVID-19.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Pobitra Borah
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Bibhuti Bhushan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | - Nizar A. Al-Shar’I
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Da’san M. M. Jaradat
- Department of Chemistry, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Munir A. Al-Zeer
- Department of Applied Biochemistry, Institute of Biotechnology, Technical University of Berlin, Berlin, Germany
| | - Saeid Abu-Romman
- Department of Biotechnology, Faculty of Agricultural Technology, Al-Balqa Applied University, Al-Salt, Jordan
| |
Collapse
|
35
|
Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, Cohen JD, Singh PA, Baldi A, Bajwa N, Kumar R, Vora LK, Patel TA, Oleynikov MD, Soni D, Yeapuri P, Mukadam I, Chakraborty R, Saksena CG, Herskovitz J, Hasan M, Oupicky D, Das S, Donnelly RF, Hettie KS, Chang L, Gendelman HE, Kevadiya BD. Nanocarrier vaccines for SARS-CoV-2. Adv Drug Deliv Rev 2021; 171:215-239. [PMID: 33428995 PMCID: PMC7794055 DOI: 10.1016/j.addr.2021.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/18/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Srijanee Das
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Preet Amol Singh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Ashish Baldi
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Neha Bajwa
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Tapan A Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology (CHARUSAT), Changa, Anand 388421, Gujarat, India
| | - Maxim D Oleynikov
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Rajashree Chakraborty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Caroline G Saksena
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Suvarthi Das
- Department of Medicine, Stanford Medical School, Stanford University, Palo Alto, CA 94304, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University, Palo Alto, CA 94304, USA
| | - Linda Chang
- Departments of Diagnostic Radiology & Nuclear Medicine, and Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| |
Collapse
|
36
|
Pal N, Mavi AK, Kumar S, Kumar U, Joshi MD, Saluja R. Current updates on adaptive immune response by B cell and T cell stimulation and therapeutic strategies for novel coronavirus disease 2019 (COVID-19) treatment. Heliyon 2021; 7:e06894. [PMID: 33937545 PMCID: PMC8076978 DOI: 10.1016/j.heliyon.2021.e06894] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/09/2021] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
The prevalence of COVID-19 continues to rise with more than 114,315,846 million confirmed cases and 2,539,427 deaths worldwide by 3 March 2021 and this number kept on increasing day by day. There is no clear therapeutic treatment or vaccine available for COVID-19 till date and by seeing such a high rise in the cases of COVID-19 on daily basis, it would have been necessary to implement precautions and hygienic measures to monitor and reduce human-to-human transmission of SARS-CoV-2 before there is any successful intervention/treatment available. Currently, several studies demonstrated the important improvements in both the innate and adaptive immune systems of COVID-19 patients. In particular, pre-existing research, on immune response to B cell and T cells are highlighting that pre-existing immunity exists in about 90% of the general population because of previous exposure to CoVs and having immunity against these CoVs. Although it is not clear from, the current studies on COVID-19 but it assumed that, it might have implication to COVID-19 severity and could play an important role in treatment or vaccine development against COVID-19. This review summarizes the information from occurrence of SARS-CoV-2 to its pathogenesis, transmission, adaptive immune response with respect to T cell and B cell stimulation and therapeutic interventions/treatment against COVID-19.
Collapse
Affiliation(s)
- Neeraj Pal
- Department of Molecular Biology and Genetic Engineering, GB Pant University of Agriculture and Technology, Pantnagar, 263145, India
| | - Anil Kumar Mavi
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, 110007, India
| | - Sundip Kumar
- Department of Molecular Biology and Genetic Engineering, GB Pant University of Agriculture and Technology, Pantnagar, 263145, India
| | - Umesh Kumar
- School of Biosciences, IMS Ghaziabad University Courses Campus, Uttar Pradesh, 201015, India
| | - Maya Datt Joshi
- Department of Biotechnology, Shobhit Institute of Engineering & Technology (Deemed to be University), Meerut, 250110, India
| | - Rohit Saluja
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Hyderabad, Telangana, 508126, India
| |
Collapse
|
37
|
An Overview of Nanocarrier-Based Adjuvants for Vaccine Delivery. Pharmaceutics 2021; 13:pharmaceutics13040455. [PMID: 33801614 PMCID: PMC8066039 DOI: 10.3390/pharmaceutics13040455] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The development of vaccines is one of the most significant medical accomplishments which has helped to eradicate a large number of diseases. It has undergone an evolutionary process from live attenuated pathogen vaccine to killed whole organisms or inactivated toxins (toxoids), each of them having its own advantages and disadvantages. The crucial parameters in vaccination are the generation of memory response and protection against infection, while an important aspect is the effective delivery of antigen in an intelligent manner to evoke a robust immune response. In this regard, nanotechnology is greatly contributing to developing efficient vaccine adjuvants and delivery systems. These can protect the encapsulated antigen from the host’s in-vivo environment and releasing it in a sustained manner to induce a long-lasting immunostimulatory effect. In view of this, the present review article summarizes nanoscale-based adjuvants and delivery vehicles such as viral vectors, virus-like particles and virosomes; non-viral vectors namely nanoemulsions, lipid nanocarriers, biodegradable and non-degradable nanoparticles, calcium phosphate nanoparticles, colloidally stable nanoparticles, proteosomes; and pattern recognition receptors covering c-type lectin receptors and toll-like receptors.
Collapse
|
38
|
Kurup D, Malherbe DC, Wirblich C, Lambert R, Ronk AJ, Zabihi Diba L, Bukreyev A, Schnell MJ. Inactivated rabies virus vectored SARS-CoV-2 vaccine prevents disease in a Syrian hamster model. PLoS Pathog 2021; 17:e1009383. [PMID: 33765062 PMCID: PMC8023494 DOI: 10.1371/journal.ppat.1009383] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/06/2021] [Accepted: 02/12/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emergent coronavirus that has caused a worldwide pandemic. Although human disease is often asymptomatic, some develop severe illnesses such as pneumonia, respiratory failure, and death. There is an urgent need for a vaccine to prevent its rapid spread as asymptomatic infections accounting for up to 40% of transmission events. Here we further evaluated an inactivated rabies vectored SARS-CoV-2 S1 vaccine CORAVAX in a Syrian hamster model. CORAVAX adjuvanted with MPLA-AddaVax, a TRL4 agonist, induced high levels of neutralizing antibodies and generated a strong Th1-biased immune response. Vaccinated hamsters were protected from weight loss and viral replication in the lungs and nasal turbinates three days after challenge with SARS-CoV-2. CORAVAX also prevented lung disease, as indicated by the significant reduction in lung pathology. This study highlights CORAVAX as a safe, immunogenic, and efficacious vaccine that warrants further assessment in human trials.
Collapse
Affiliation(s)
- Drishya Kurup
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Delphine C. Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rachael Lambert
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Adam J. Ronk
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Leila Zabihi Diba
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Matthias J. Schnell
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Jefferson Vaccine Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
39
|
Farmers' preference and willingness to pay for a multivalent lumpy skin disease and Rift Valley fever novel vaccine: A discrete choice experiment in the Free State province, South Africa. Prev Vet Med 2021; 189:105293. [PMID: 33631510 DOI: 10.1016/j.prevetmed.2021.105293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 11/20/2022]
Abstract
Rift Valley fever and lumpy skin disease are infectious ruminant diseases that are endemic in most African countries. The most cost-effective method of prevention and control is through annual vaccination. However, unlike lumpy skin disease, annual vaccination against Rift Valley fever is not practiced by many farmers due to its sporadic occurrences and shortcoming of the existing vaccines. This necessitates development of novel vaccines that would provide dual protection against a Rift Valley fever and a more prevalent disease. In this study, a discrete choice experiment was undertaken to guide vaccine development by examining the value smallholder livestock farmers place on different vaccine attributes and related attribute levels. The attributes considered are target-species, thermotolerance, nature of the vaccine, efficacy and price. The study was carried out with 164 smallholder livestock farmers in the Free State province. Results indicate that thermostaility is not a major deciding attribute to smallholder farmer's choice of vaccine. Farmers prefer multivalent vaccines, which are highly efficacious with about 90-100 % efficacy levels. Farmers were found to be heterogeneous in preference. The heterogeneity is explained by socio-economic factors such as type of livestock owned, income level, gender and perceived disease risk. Farmers were also willing to pay for preferred attribute levels. However, for less favourable levels such as multispecies, female farmers were willing to accept a lower compensation than males. These findings present a favourable potential for development of a novel multivalent vaccine and also provide vaccine research and development scientists with evidence based knowledge for development of vaccines that cater for the needs of smallholder farmers.
Collapse
|
40
|
Future perspectives on swine viral vaccines: where are we headed? Porcine Health Manag 2021; 7:1. [PMID: 33397477 PMCID: PMC7780603 DOI: 10.1186/s40813-020-00179-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/27/2020] [Indexed: 12/18/2022] Open
Abstract
Deliberate infection of humans with smallpox, also known as variolation, was a common practice in Asia and dates back to the fifteenth century. The world's first human vaccination was administered in 1796 by Edward Jenner, a British physician. One of the first pig vaccines, which targeted the bacterium Erysipelothrix rhusiopathiae, was introduced in 1883 in France by Louis Pasteur. Since then vaccination has become an essential part of pig production, and viral vaccines in particular are essential tools for pig producers and veterinarians to manage pig herd health. Traditionally, viral vaccines for pigs are either based on attenuated-live virus strains or inactivated viral antigens. With the advent of genomic sequencing and molecular engineering, novel vaccine strategies and tools, including subunit and nucleic acid vaccines, became available and are being increasingly used in pigs. This review aims to summarize recent trends and technologies available for the production and use of vaccines targeting pig viruses.
Collapse
|
41
|
Awadasseid A, Wu Y, Tanaka Y, Zhang W. Current advances in the development of SARS-CoV-2 vaccines. Int J Biol Sci 2021; 17:8-19. [PMID: 33390829 PMCID: PMC7757035 DOI: 10.7150/ijbs.52569] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/22/2020] [Indexed: 01/01/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now a global pandemic that has wreaked havoc globally, which has put a heavy toll on public health, lives, and the world economy. Vaccination is considered as one of the greatest successes in medical history. Based on prior experience with the development of SARS-CoV vaccines, all COVID-19 vaccines must be subjected to the tests for protective effects and harmful risks derived from antibody-dependent enhancement that may contribute to augmented infectivity and/or eosinophilic infiltration. The SARS-CoV-2 vaccine is now being developed urgently in several different ways. China is regarded as one of the world's leading countries in SARS-CoV-2 vaccine development, up to date the last inactivated vaccine international clinical (Phase III) trial was launched in the United Arab Emirates by Sinopharm China National Biotec Group (CNBG). In this review, we outline the current status of vaccine development against clinically relevant SARS-CoV-2 strains, anticipating that such attempts would help create efficacious and sage SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Annoor Awadasseid
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
- Department of Biochemistry & Food Sciences, University of Kordofan, El-Obeid, 51111, Sudan
| | - Yanling Wu
- Lab of Molecular Immunology, Virus Inspection Department, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China
| |
Collapse
|
42
|
Layman NC, Tuschhoff BM, Nuismer SL. Designing transmissible viral vaccines for evolutionary robustness and maximum efficiency. Virus Evol 2021; 7:veab002. [PMID: 33680502 PMCID: PMC7920745 DOI: 10.1093/ve/veab002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The danger posed by emerging infectious diseases necessitates the development of new tools that can mitigate the risk of animal pathogens spilling over into the human population. One promising approach is the development of recombinant viral vaccines that are transmissible, and thus capable of self-dissemination through hard to reach populations of wild animals. Indeed, mathematical models demonstrate that transmissible vaccines can greatly reduce the effort required to control the spread of zoonotic pathogens in their animal reservoirs, thereby limiting the chances of human infection. A key challenge facing these new vaccines, however, is the inevitability of evolutionary change resulting from their ability to self-replicate and generate extended chains of transmission. Further, carrying immunogenic transgenes is often costly, in terms of metabolic burden, increased competition with the pathogen, or due to unintended interactions with the viral host regulatory network. As a result, natural selection is expected to favor vaccine strains that down-regulate or delete these transgenes resulting in increased rates of transmission and reduced efficacy against the target pathogen. In addition, efficacy and evolutionary stability will often be at odds; as when longer, more efficacious antigens experience faster rates of evolutionary decay. Here, we ask how such trade-offs influence the overall performance of transmissible vaccines. We find that evolutionary instability can substantially reduce performance, even for vaccine candidates with the ideal combination of efficacy and transmission. However, we find that, at least in some cases, vaccine stability and overall performance can be improved by the inclusion of a second, redundant antigen. Overall, our results suggest that the successful application of recombinant transmissible vaccines will require consideration of evolutionary dynamics and epistatic effects, as well as basic measurements of epidemiological features.
Collapse
Affiliation(s)
| | - Beth M Tuschhoff
- Department of Mathematics, University of Idaho, 875 Perimeter Drive, Moscow, ID 83844, USA
| | | |
Collapse
|
43
|
Viral Vector Vaccines against Bluetongue Virus. Microorganisms 2020; 9:microorganisms9010042. [PMID: 33375723 PMCID: PMC7823852 DOI: 10.3390/microorganisms9010042] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/20/2022] Open
Abstract
Bluetongue virus (BTV), the prototype member of the genus Orbivirus (family Reoviridae), is the causative agent of an important livestock disease, bluetongue (BT), which is transmitted via biting midges of the genus Culicoides. To date, up to 29 serotypes of BTV have been described, which are classified as classical (BTV 1–24) or atypical (serotypes 25–27), and its distribution has been expanding since 1998, with important outbreaks in the Mediterranean Basin and devastating incursions in Northern and Western Europe. Classical vaccine approaches, such as live-attenuated and inactivated vaccines, have been used as prophylactic measures to control BT through the years. However, these vaccine approaches fail to address important matters like vaccine safety profile, effectiveness, induction of a cross-protective immune response among serotypes, and implementation of a DIVA (differentiation of infected from vaccinated animals) strategy. In this context, a wide range of recombinant vaccine prototypes against BTV, ranging from subunit vaccines to recombinant viral vector vaccines, have been investigated. This article offers a comprehensive outline of the live viral vectors used against BTV.
Collapse
|
44
|
Van der Weken H, Cox E, Devriendt B. Advances in Oral Subunit Vaccine Design. Vaccines (Basel) 2020; 9:1. [PMID: 33375151 PMCID: PMC7822154 DOI: 10.3390/vaccines9010001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/17/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023] Open
Abstract
Many pathogens invade the host at the intestinal surface. To protect against these enteropathogens, the induction of intestinal secretory IgA (SIgA) responses is paramount. While systemic vaccination provides strong systemic immune responses, oral vaccination is the most efficient way to trigger protective SIgA responses. However, the development of oral vaccines, especially oral subunit vaccines, is challenging due to mechanisms inherent to the gut. Oral vaccines need to survive the harsh environment in the gastrointestinal tract, characterized by low pH and intestinal proteases and need to reach the gut-associated lymphoid tissues, which are protected by chemical and physical barriers that prevent efficient uptake. Furthermore, they need to surmount default tolerogenic responses present in the gut, resulting in suppression of immunity or tolerance. Several strategies have been developed to tackle these hurdles, such as delivery systems that protect vaccine antigens from degradation, strong mucosal adjuvants that induce robust immune responses and targeting approaches that aim to selectively deliver vaccine antigens towards specific immune cell populations. In this review, we discuss recent advances in oral vaccine design to enable the induction of robust gut immunity and highlight that the development of next generation oral subunit vaccines will require approaches that combines these solutions.
Collapse
Affiliation(s)
| | | | - Bert Devriendt
- Department of Virology, Parasitology and Immunology, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (H.V.d.W.); (E.C.)
| |
Collapse
|
45
|
Almond J, Hacker J, Harwood C, Pizza M, Rappuoli R, Ron EZ, Sansonetti P, Vanderslott S, Wieler LH. Development of vaccines at the time of COVID-19. ACTA ACUST UNITED AC 2020; 1:uqaa003. [PMID: 34235437 PMCID: PMC7798935 DOI: 10.1093/femsml/uqaa003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
In December 2019, a working group of the European Academy of Microbiology assembled to discuss various aspects of vaccines and vaccinations. The meeting was organised by Jörg Hacker and Eliora Z. Ron and took place in the offices of the Leopoldina (German National Academy of Sciences Leopoldina). Several important issues were addressed and a major part of the discussion focused on the need to develop new vaccines, especially to protect against pathogens that constitute a pandemic threat. Following the rapid and unpredicted spread of COVID-19 in the first seven months of 2020, the need to develop vaccines for pandemic viruses rapidly has been clearly established. Thus, this paper will concentrate on points that were highlighted by the recent COVID-19 pandemic and lessons learnt therefrom.
Collapse
Affiliation(s)
- Jeffrey Almond
- The Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Jörg Hacker
- German National Academy of Science Leopoldina, Jägerberg 1, 06108 Halle, Germany
| | - Colin Harwood
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Baddiley-Clark Building, Newcastle upon Tyne NE2 4AX, UK
| | | | - Rino Rappuoli
- GSK Vaccines, Via Fiorentina, 1, 53100 Siena SI, Italy
| | - Eliora Z Ron
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, PO Box 39040, Tel Aviv 6997801, Israel
| | | | | | | |
Collapse
|
46
|
Iankov ID, Kurokawa C, Viker K, Robinson SI, Ammayappan A, Panagioti E, Federspiel MJ, Galanis E. Live Attenuated Measles Virus Vaccine Expressing Helicobacter pylori Heat Shock Protein A. MOLECULAR THERAPY-ONCOLYTICS 2020; 19:136-148. [PMID: 33145397 PMCID: PMC7585873 DOI: 10.1016/j.omto.2020.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022]
Abstract
Measles virus (MV) Edmonston derivative strains are attractive vector platforms in vaccine development and oncolytic virotherapy. Helicobacter pylori heat shock protein A (HspA) is a bacterial heat shock chaperone with essential function as a Ni-ion scavenging protein. We generated and characterized the immunogenicity of an attenuated MV strain encoding the HspA transgene (MV-HspA). MV-HspA showed faster replication within 48 h of infection with >10-fold higher titers and faster accumulation of the MV proteins. It also demonstrated a superior tumor-killing effect in vitro against a variety of human solid tumor cell lines, including sarcoma, ovarian and breast cancer. Two intraperitoneal (i.p.) doses of 106 50% tissue culture infectious dose (TCID50) MV-HspA significantly improved survival in an ovarian cancer xenograft model: 63.5 days versus 27 days for the control group. The HspA transgene induced a humoral immune response in measles-permissive Ifnarko-CD46Ge transgenic mice. Eight of nine animals developed a long-term anti-HspA antibody response with titers of 1:400 to 1:12,800 without any negative impact on development of protective anti-MV immune memory. MV-HspA triggered an immunogenic cytopathic effect as measured by an HMGB1 assay. The absence of significant elevation of PD-L1 expression indicated that vector-encoded HspA could act as an immunomodulator on the immune check point axis. These data demonstrate that MV-HspA is a potent oncolytic agent and vaccine candidate for clinical translation in cancer treatment and immunoprophylaxis against H. pylori.
Collapse
Affiliation(s)
- Ianko D Iankov
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Cheyne Kurokawa
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Kimberly Viker
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Steven I Robinson
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Arun Ammayappan
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Eleni Panagioti
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Mark J Federspiel
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
47
|
Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and Applications of Viral Vectored Vaccines to Combat Zoonotic and Emerging Public Health Threats. Vaccines (Basel) 2020; 8:E680. [PMID: 33202961 PMCID: PMC7712223 DOI: 10.3390/vaccines8040680] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Vaccination is arguably the most cost-effective preventative measure against infectious diseases. While vaccines have been successfully developed against certain viruses (e.g., yellow fever virus, polio virus, and human papilloma virus HPV), those against a number of other important public health threats, such as HIV-1, hepatitis C, and respiratory syncytial virus (RSV), have so far had very limited success. The global pandemic of COVID-19, caused by the SARS-CoV-2 virus, highlights the urgency of vaccine development against this and other constant threats of zoonotic infection. While some traditional methods of producing vaccines have proven to be successful, new concepts have emerged in recent years to produce more cost-effective and less time-consuming vaccines that rely on viral vectors to deliver the desired immunogens. This review discusses the advantages and disadvantages of different viral vaccine vectors and their general strategies and applications in both human and veterinary medicines. A careful review of these issues is necessary as they can provide important insights into how some of these viral vaccine vectors can induce robust and long-lasting immune responses in order to provide protective efficacy against a variety of infectious disease threats to humans and animals, including those with zoonotic potential to cause global pandemics.
Collapse
Affiliation(s)
- Sophia M. Vrba
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Natalie M. Kirk
- Comparative Molecular Biosciences Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Morgan E. Brisse
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA;
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, St. Paul, MN 55108, USA; (S.M.V.); (Y.L.)
| |
Collapse
|
48
|
Soltani S, Zandi M, Shiri Aghbash P, Rezaei M, Mohammadzadeh N, Afsharifar A, Poortahmasebi V. A review of COVID-19 vaccines and major considerations for diabetic patients. Biotechnol Appl Biochem 2020; 69:30-40. [PMID: 33179788 DOI: 10.1002/bab.2076] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/05/2020] [Indexed: 01/08/2023]
Abstract
The necessity and impact of SARS-CoV2 on the world's health have led to developing and producing practical and useful vaccines for this deadly respiratory virus. Since April 2020, a vaccine for the virus has been developed. Given that comorbidities such as diabetes, hypertension, and cardiovascular disease are more prone to viruses and the risk of infection, vaccines should be designed to protect against high-risk respiratory illnesses. Including SARS, MERS, influenza, and the SARS-CoV-2 provide a safe immune response. Here, we review the information and studies that have been done to help develop strategies and perspectives for producing a safe and ideal vaccine to prevent COVID-19 in normal people, especially at high-risk groups such as diabetes patients.
Collapse
Affiliation(s)
- Saber Soltani
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Zandi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Parisa Shiri Aghbash
- Department of Bacteriology and Virology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Rezaei
- Department of Plant Protection, Shiraz University, Shiraz, Iran
| | - Nader Mohammadzadeh
- Health Reference Laboratory, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
49
|
Ye T, Zhong Z, García‐Sastre A, Schotsaert M, De Geest BG. Current Status of COVID-19 (Pre)Clinical Vaccine Development. Angew Chem Int Ed Engl 2020; 59:18885-18897. [PMID: 32663348 PMCID: PMC7405471 DOI: 10.1002/anie.202008319] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Indexed: 12/29/2022]
Abstract
The current COVID-19 pandemic has a tremendous impact on daily life world-wide. Despite the ability to dampen the spread of SARS-CoV-2, the causative agent of the diseases, through restrictive interventions, it is believed that only effective vaccines will provide sufficient control over the disease and revert societal live back to normal. At present, a double-digit number of efforts are devoted to the development of a vaccine against COVID-19. Here, we provide an overview of these (pre)clinical efforts and provide background information on the technologies behind these vaccines. In addition, we discuss potential hurdles that need to be addressed prior to mass scale clinical translation of successful vaccine candidates.
Collapse
Affiliation(s)
- Tingting Ye
- Department of PharmaceuticsGhent UniversityBelgium
| | - Zifu Zhong
- Department of PharmaceuticsGhent UniversityBelgium
| | - Adolfo García‐Sastre
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of MedicineDivision of Infectious DiseasesIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- The Tisch Cancer InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Michael Schotsaert
- Department of MicrobiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Global Health and Emerging Pathogens InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | | |
Collapse
|
50
|
Brown S, Brown T, Cederna PS, Rohrich RJ. The Race for a COVID-19 Vaccine: Current Trials, Novel Technologies, and Future Directions. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e3206. [PMID: 33173705 PMCID: PMC7647601 DOI: 10.1097/gox.0000000000003206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic has presented a major threat to public health worldwide alongside unprecedented global economic and social implications. In the absence of a "gold standard" treatment, the rapid development of a safe and effective vaccine is considered the most promising way to control the pandemic. In recent years, traditional vaccine technologies have seemed insufficient to provide global protection against the rapid spread of emerging pandemics. Therefore, the establishment of novel approaches that are independent of whole pathogen cultivation, cost-effective, and able to be rapidly developed and produced on a large scale are of paramount importance for global health. This article summarizes the current efforts to develop a COVID-19 vaccine, including the ongoing and future anticipated clinical trials. We also provide plastic and reconstructive surgeons with insight into the novel technologies currently utilized for COVID-19 vaccine development, focusing on the very promising viral-vector-based and gene-based vaccine technologies. Each platform has its own advantages and disadvantages related to its efficacy and ability to induce certain immune responses, manufacturing capacity, and safety for human use. Once the fundamental key challenges have been addressed for viral-vector-based and gene-based vaccines, these novel technologies may become helpful in winning the fight against COVID-19 and transforming the future of health care.
Collapse
Affiliation(s)
- Stav Brown
- From the Sackler School of Medicine at Tel Aviv University, Tel Aviv, Israel
| | - Tal Brown
- From the Sackler School of Medicine at Tel Aviv University, Tel Aviv, Israel
| | - Paul S. Cederna
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Mich
- Section of Plastic Surgery, University of Michigan, Ann Arbor, Mich
| | | |
Collapse
|