1
|
Saravanan V, Chagaleti BK, Narayanan PL, Anandan VB, Manoharan H, Anjana GV, Peraman R, Namasivayam SKR, Kavisri M, Arockiaraj J, Muthu Kumaradoss K, Moovendhan M. Discovery and development of COVID-19 vaccine from laboratory to clinic. Chem Biol Drug Des 2024; 103:e14383. [PMID: 37953736 DOI: 10.1111/cbdd.14383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/01/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023]
Abstract
The world has recently experienced one of the biggest and most severe public health disasters with severe acute respiratory syndrome coronavirus (SARS-CoV-2). SARS-CoV-2 is responsible for the coronavirus disease of 2019 (COVID-19) which is one of the most widespread and powerful infections affecting human lungs. Current figures show that the epidemic had reached 216 nations, where it had killed about 6,438,926 individuals and infected 590,405,710. WHO proclaimed the outbreak of the Ebola virus disease (EVD), in 2014 that killed hundreds of people in West Africa. The development of vaccines for SARS-CoV-2 becomes more difficult due to the viral mutation in its non-structural proteins (NSPs) especially NSP2 and NSP3, S protein, and RNA-dependent RNA polymerase (RdRp). Continuous monitoring of SARS-CoV-2, dynamics of the genomic sequence, and spike protein mutations are very important for the successful development of vaccines with good efficacy. Hence, the vaccine development for SARS-CoV-2 faces specific challenges starting from viral mutation. The requirement of long-term immunity development, safety, efficacy, stability, vaccine allocation, distribution, and finally, its cost is discussed in detail. Currently, 169 vaccines are in the clinical development stage, while 198 vaccines are in the preclinical development stage. The majority of these vaccines belong to the Ps-Protein subunit type which has 54, and the minor BacAg-SPV (Bacterial antigen-spore expression vector) type, at least 1 vaccination. The use of computational methods and models for vaccine development has revolutionized the traditional methods of vaccine development. Further, this updated review highlights the upcoming vaccine development strategies in response to the current pandemic and post-pandemic era, in the field of vaccine development.
Collapse
Affiliation(s)
- Venkatesan Saravanan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Bharath Kumar Chagaleti
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Pavithra Lakshmi Narayanan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Vijay Babu Anandan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Haritha Manoharan
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - G V Anjana
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Ramalingam Peraman
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER) Hajipur, Hajipur, India
| | - S Karthik Raja Namasivayam
- Department of Research & Innovation, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - M Kavisri
- Department of Civil Engineering, Saveetha School of Engineering, SIMATS Deemed University, Chennai, India
| | - Jesu Arockiaraj
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Kathiravan Muthu Kumaradoss
- Dr. APJ Abdul Kalam Research Lab, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, India
| | - Meivelu Moovendhan
- Centre for Ocean Research, Col. Dr. Jeppiar Research Park, Sathyabama Institute of Science and Technology, Chennai, India
| |
Collapse
|
2
|
Lee H, Hong B, Kim S, Kim JH, Choi NK, Jung SY, Shin JY. Post-marketing surveillance study on influenza vaccine in South Korea using a nationwide spontaneous reporting database with multiple data mining methods. Sci Rep 2022; 12:20256. [PMID: 36424402 PMCID: PMC9691710 DOI: 10.1038/s41598-022-21986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/07/2022] [Indexed: 11/26/2022] Open
Abstract
Safety profiles of the influenza vaccine and its subtypes are still limited. We aimed to address this knowledge gap using multiple data mining methods and calculated performance measurements to evaluate the precision of different detection methods. We conducted a post-marketing surveillance study between 2005 and 2019 using the Korea Adverse Event Reporting System database. Three data mining methods were applied: (a) proportional reporting ratio, (b) information component, and (c) tree-based scan statistics. We evaluated the performance of each method in comparison with the known adverse events (AEs) described in the labeling information. Compared to other vaccines, we identified 36 safety signals for the influenza vaccine, and 7 safety signals were unlabeled. In subtype-stratified analyses, application site disorders were reported more frequently with quadrivalent and cell-based vaccines, while a wide range of AEs were noted for trivalent and egg-based vaccines. Tree-based scan statistics showed well-balanced performance. Among the detected signals of influenza vaccines, narcolepsy requires special attention. A wider range of AEs were detected as signals for trivalent and egg-based vaccines. Although tree-based scan statistics showed balanced performance, complementary use of other techniques would be beneficial when large noise due to false positives is expected.
Collapse
Affiliation(s)
- Hyesung Lee
- grid.264381.a0000 0001 2181 989XSchool of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419 South Korea ,grid.264381.a0000 0001 2181 989XDepartment of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Bin Hong
- grid.264381.a0000 0001 2181 989XSchool of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419 South Korea
| | - SangHee Kim
- grid.264381.a0000 0001 2181 989XSchool of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419 South Korea
| | - Ju Hwan Kim
- grid.264381.a0000 0001 2181 989XSchool of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419 South Korea ,grid.264381.a0000 0001 2181 989XPresent Address: Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Nam-Kyong Choi
- grid.255649.90000 0001 2171 7754Department of Health Convergence, Ewha Womans University, Seoul, South Korea ,grid.255649.90000 0001 2171 7754Graduate School of Industrial Pharmaceutical Science, Ewha Womans University, Seoul, Korea
| | - Sun-Young Jung
- grid.254224.70000 0001 0789 9563College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea ,grid.254224.70000 0001 0789 9563Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 South Korea
| | - Ju-Young Shin
- grid.264381.a0000 0001 2181 989XSchool of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419 South Korea ,grid.264381.a0000 0001 2181 989XDepartment of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea ,grid.264381.a0000 0001 2181 989XDepartment of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
3
|
Novel Strategies of Immunization against COVID-19. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.1.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
COVID-19 manifested itself as a global pandemic in 2019 but even in 2021, it is still not successfully contained. This virus has claimed millions of lives worldwide and rendered many more jobless. Apart from causing mild to severe pneumonia, the virus has also caused a loss of livelihood for thousands globally, along with widespread trauma and depression. Since the transmission rate of the virus is so high, temporary prophylaxis relied on sanitization, wearing masks and physical distancing. However, a long-term solution for stopping viral spread is vaccination. Apart from being the fastest way to induce immunity against the virus, vaccination is also the cheapest and most practical way. However, a vaccine can only be commercially available after it has passed through various clinical trial phases. So far, more than two hundred potential vaccine candidates underwent different phases of the clinical trial, and some of the front-runners have shown more than 90% efficacy. This review has compiled all such vaccine candidates, their types, their modes of action, and the associated pros and cons. The current advances in clinical trials of vaccines have also been discussed, such as plant-based and cocktail vaccines that have recently emerged. Nowadays, novel strains like Delta plus are also emerging and posing a threat. Thus, it is mandatory to get vaccinated and choose a vaccine that provides long-term protection against multiple strains.
Collapse
|
4
|
Pathania S, Pathania D, Chauhan P, Sharma M. Himalayan poisonous plants for traditional healings and protection from viral attack: a comprehensive review. TOXIN REV 2022. [DOI: 10.1080/15569543.2021.2023575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Shriya Pathania
- School of Biological and Environmental Sciences, Shoolini University, Solan, HP, India
| | - Diksha Pathania
- School of Biological and Environmental Sciences, Shoolini University, Solan, HP, India
| | - Priyanka Chauhan
- School of Biological and Environmental Sciences, Shoolini University, Solan, HP, India
| | - Mamta Sharma
- School of Biological and Environmental Sciences, Shoolini University, Solan, HP, India
| |
Collapse
|
5
|
McMillan CLD, Cheung STM, Modhiran N, Barnes J, Amarilla AA, Bielefeldt-Ohmann H, Lee LYY, Guilfoyle K, van Amerongen G, Stittelaar K, Jakon V, Lebas C, Reading P, Short KR, Young PR, Watterson D, Chappell KJ. Development of molecular clamp stabilized hemagglutinin vaccines for Influenza A viruses. NPJ Vaccines 2021; 6:135. [PMID: 34750396 PMCID: PMC8575991 DOI: 10.1038/s41541-021-00395-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 10/01/2021] [Indexed: 11/25/2022] Open
Abstract
Influenza viruses cause a significant number of infections and deaths annually. In addition to seasonal infections, the risk of an influenza virus pandemic emerging is extremely high owing to the large reservoir of diverse influenza viruses found in animals and the co-circulation of many influenza subtypes which can reassort into novel strains. Development of a universal influenza vaccine has proven extremely challenging. In the absence of such a vaccine, rapid response technologies provide the best potential to counter a novel influenza outbreak. Here, we demonstrate that a modular trimerization domain known as the molecular clamp allows the efficient production and purification of conformationally stabilised prefusion hemagglutinin (HA) from a diverse range of influenza A subtypes. These clamp-stabilised HA proteins provided robust protection from homologous virus challenge in mouse and ferret models and some cross protection against heterologous virus challenge. This work provides a proof-of-concept for clamp-stabilised HA vaccines as a tool for rapid response vaccine development against future influenza A virus pandemics.
Collapse
Affiliation(s)
- Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Stacey T M Cheung
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - James Barnes
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.,Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, 4072 and 4029, Australia.,School of Veterinary Science, The University of Queensland Gatton Campus, Gatton, QLD, 4343, Australia
| | - Leo Yi Yang Lee
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Kate Guilfoyle
- Viroclinics Xplore, Landerd Campus, Nistelrooise Baan 3, 5374 RE, Schaijk, The Netherlands
| | - Geert van Amerongen
- Viroclinics Xplore, Landerd Campus, Nistelrooise Baan 3, 5374 RE, Schaijk, The Netherlands
| | - Koert Stittelaar
- Viroclinics Xplore, Landerd Campus, Nistelrooise Baan 3, 5374 RE, Schaijk, The Netherlands
| | - Virginie Jakon
- Vaccine Formulation Institute, Chemin des Aulx 14, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - Celia Lebas
- Vaccine Formulation Institute, Chemin des Aulx 14, 1228 Plan-Les-Ouates, Geneva, Switzerland
| | - Patrick Reading
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC, 3000, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia.,Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, 4072 and 4029, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, 4072 and 4029, Australia. .,The Australian Institute of Biotechnology and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, 4072 and 4029, Australia. .,The Australian Institute of Biotechnology and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.
| | - Keith J Chappell
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, 4072, Australia. .,Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, QLD, 4072 and 4029, Australia. .,The Australian Institute of Biotechnology and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia.
| |
Collapse
|
6
|
Laurent A, Abdel-Sayed P, Hirt-Burri N, Scaletta C, Michetti M, de Buys Roessingh A, Raffoul W, Applegate LA. Evolution of Diploid Progenitor Lung Cell Applications: From Optimized Biotechnological Substrates to Potential Active Pharmaceutical Ingredients in Respiratory Tract Regenerative Medicine. Cells 2021; 10:2526. [PMID: 34685505 PMCID: PMC8533713 DOI: 10.3390/cells10102526] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
The objective of this review is to describe the evolution of lung tissue-derived diploid progenitor cell applications, ranging from historical biotechnological substrate functions for vaccine production and testing to current investigations around potential therapeutic use in respiratory tract regenerative medicine. Such cell types (e.g., MRC-5 or WI-38 sources) were extensively studied since the 1960s and have been continuously used over five decades as safe and sustainable industrial vaccine substrates. Recent research and development efforts around diploid progenitor lung cells (e.g., FE002-Lu or Walvax-2 sources) consist in qualification for potential use as optimal and renewed vaccine production substrates and, alternatively, for potential therapeutic applications in respiratory tract regenerative medicine. Potentially effective, safe, and sustainable cell therapy approaches for the management of inflammatory lung diseases or affections and related symptoms (e.g., COVID-19 patients and burn patient severe inhalation syndrome) using local homologous allogeneic cell-based or cell-derived product administrations are considered. Overall, lung tissue-derived progenitor cells isolated and produced under good manufacturing practices (GMP) may be used with high versatility. They can either act as key industrial platforms optimally conforming to specific pharmacopoeial requirements or as active pharmaceutical ingredients (API) for potentially effective promotion of lung tissue repair or regeneration.
Collapse
Affiliation(s)
- Alexis Laurent
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- TEC-PHARMA SA, Manufacturing Department, CH-1038 Bercher, Switzerland
- LAM Biotechnologies SA, Manufacturing Department, CH-1066 Épalinges, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Corinne Scaletta
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
| | - Anthony de Buys Roessingh
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Wassim Raffoul
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Plastic, Reconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, CH-1066 Épalinges, Switzerland; (A.L.); (P.A.-S.); (N.H.-B.); (C.S.); (M.M.)
- Romand Burn Center, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| |
Collapse
|
7
|
Development, Biological Characterization, and Immunological Evaluation of Virosome Vaccine against Newcastle Disease in Pakistan. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8879277. [PMID: 33575353 PMCID: PMC7864732 DOI: 10.1155/2021/8879277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 01/13/2021] [Accepted: 01/22/2021] [Indexed: 11/24/2022]
Abstract
Newcastle disease (ND) is a highly fatal, infectious, viral disease, and despite immunization with live and inactivated vaccines, the disease is still endemic, causing heavy morbidity and mortality leading to huge economic losses to the poultry industry in Pakistan. Therefore, the present study was aimed for the first time in the country at using novel virosomal technology to develop the ND vaccine using an indigenous highly virulent strain of the virus. ND virosome was prepared using Triton X-100, and SM2 Bio-Beads were used to remove the detergent and reconstitute the viral membrane into virosome. Confirmation was done by transmission electron microscopy and protein analysis by SDS-PAGE. In vitro cell adhesion property was observed by incorporating green fluorescent protein (GFP), producing plasmid into virosome and in vitro cell culture assay. Sterility, safety, and stability of the vaccine were tested before in vivo evaluation of immunogenicity and challenge protection study in commercial broiler. The virosome vaccine was administered (30 μg/bird) at days 7 and 14 through the intranasal route in comparison with commercially available live and inactivated ND vaccines. Results revealed significantly high (p < 0.05) and clinically protective hemagglutination inhibition (HI) antibody titers at 7, 14, 21, and 28 days postimmunization with the virosome vaccine in comparison to the negative control. The GMTs were comparable to live and inactivated vaccines with nonsignificant (p > 0.05) differences throughout the experiment. Antibody levels increased in all vaccinated groups gradually from the 7th day and were maximum at 28th-day postvaccination. In the virosome-administered group, GMT was 83.18 and 77.62 at 21st and 28th-days postvaccination, respectively. Challenge revealed 100%, 90%, and 80% protection in virosome, live, and inactivated vaccinated groups, respectively. Under given experimental conditions, we can conclude that ND virosome vaccine prepared from the indigenous virus was found to be safe and immunogenic.
Collapse
|
8
|
Dhama K, Natesan S, Iqbal Yatoo M, Patel SK, Tiwari R, Saxena SK, Harapan H. Plant-based vaccines and antibodies to combat COVID-19: current status and prospects. Hum Vaccin Immunother 2020; 16:2913-2920. [PMID: 33270484 PMCID: PMC7754927 DOI: 10.1080/21645515.2020.1842034] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/19/2022] Open
Abstract
Globally, researchers are undertaking significant efforts to design and develop effective vaccines, therapeutics, and antiviral drugs to curb the spread of coronavirus disease 2019 (COVID-19). Plants have been used for the production of vaccines, monoclonal antibodies, immunomodulatory proteins, drugs, and pharmaceuticals via molecular farming/transient expression system and are considered as bioreactors or factories for their bulk production. These biological products are stable, safe, effective, easily available, and affordable. Plant molecular farming could facilitate rapid production of biologics on an industrial scale, and has the potential to fulfill emergency demands, such as in the present situation of the COVID-19 pandemic. This article aims to describe the methodology and basics of plant biopharming, in addition to its prospective applications for developing effective vaccines and antibodies to counter COVID-19.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Senthilkumar Natesan
- Division of Biological & Life Sciences, Indian Institute of Public Health Gandhinagar, Ganghinagar, India
| | - Mohd. Iqbal Yatoo
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, Alusteng Srinagar, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, India
| | - Shailesh Kumar Patel
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, India
| | - Shailendra K Saxena
- Centre for Advanced Research (CFAR), Faculty of Medicine, King George’s Medical University (KGMU), Lucknow, India
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| |
Collapse
|
9
|
Pérez-Rubio A, Ancochea J, Eiros Bouza JM. Quadrivalent cell culture influenza virus vaccine. Comparison to egg-derived vaccine. Hum Vaccin Immunother 2020; 16:1746-1752. [PMID: 32255723 DOI: 10.1080/21645515.2019.1701912] [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] [Indexed: 10/24/2022] Open
Abstract
Influenza virus infections pose a serious public health problem and vaccination is the most effective public health intervention against them. The current manufacture of influenza vaccines in embryonated chicken eggs entails significant limitations. These limitations have been overcome by producing vaccines in cell culture, which allow a faster and more flexible response to potential pandemic threats. Given the impact of influenza B virus on disease burden, the availability of quadrivalent vaccines is useful for increasing the rate of protection from disease. This paper analyzes the limitations of the current production of influenza vaccine in eggs and the advantages of vaccines developed in cell culture, as well as their safety, tolerability, efficacy and effectiveness. Additionally, we reflect on the contribution of new quadrivalent vaccines from cell culture as an alternative in seasonal vaccination campaigns against influenza.
Collapse
Affiliation(s)
- Alberto Pérez-Rubio
- Dirección Médica, Hospital Clínico Universitario de Valladolid , Valladolid, Castilla y León, Spain
| | - Julio Ancochea
- Neumology, Hospital Universitario de la Princesa , Madrid, Spain
| | | |
Collapse
|
10
|
Capell T, Twyman RM, Armario-Najera V, Ma JKC, Schillberg S, Christou P. Potential Applications of Plant Biotechnology against SARS-CoV-2. TRENDS IN PLANT SCIENCE 2020; 25:635-643. [PMID: 32371057 PMCID: PMC7181989 DOI: 10.1016/j.tplants.2020.04.009] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 05/17/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus responsible for an ongoing human pandemic (COVID-19). There is a massive international effort underway to develop diagnostic reagents, vaccines, and antiviral drugs in a bid to slow down the spread of the disease and save lives. One part of that international effort involves the research community working with plants, bringing researchers from all over the world together with commercial enterprises to achieve the rapid supply of protein antigens and antibodies for diagnostic kits, and scalable production systems for the emergency manufacturing of vaccines and antiviral drugs. Here, we look at some of the ways in which plants can and are being used in the fight against COVID-19.
Collapse
Affiliation(s)
- Teresa Capell
- Department of Crop and Forest Sciences, University of Lleida-Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain
| | | | - Victoria Armario-Najera
- Department of Crop and Forest Sciences, University of Lleida-Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain
| | - Julian K-C Ma
- Institute for Infection and Immunity, St George's University of London, London, UK.
| | | | - Paul Christou
- Department of Crop and Forest Sciences, University of Lleida-Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain; ICREA, Catalan Institute for Research and Advanced Studies, Passeig Lluıís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|
11
|
Sharma J, Shepardson K, Johns LL, Wellham J, Avera J, Schwarz B, Rynda-Apple A, Douglas T. A Self-Adjuvanted, Modular, Antigenic VLP for Rapid Response to Influenza Virus Variability. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18211-18224. [PMID: 32233444 DOI: 10.1021/acsami.9b21776] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The continuous evolution of influenza A virus (IAV) requires the influenza vaccine formulations to be updated annually to provide adequate protection. Recombinant protein-based vaccines provide safer, faster, and a more scalable alternative to the conventional embryonated egg approach for developing vaccines. However, these vaccines are typically poorer in immunogenicity than the vaccines containing inactivated or attenuated influenza viruses and require administration of a large antigen dosage together with potent adjuvants. The presentation of protein antigens on the surface of virus-like particles (VLP) provides an attractive strategy to rapidly induce stronger antigen-specific immune responses. Here we have examined the immunogenic potential and protective efficacy of P22 VLPs conjugated with multiple copies of the globular head domain of the hemagglutinin (HA) protein from the PR8 strain of IAV in a murine model of influenza pathogenesis. Using a covalent attachment strategy (SpyTag/SpyCatcher), we conjugated the HA globular head, which was recombinantly expressed in a genetically modified E. coli strain and found to refold as a monomer, to preassembled P22 VLPs. Immunization of mice with this P22-HAhead conjugate provided full protection from morbidity and mortality following infection with a homologous IAV strain. Moreover, the P22-HAhead conjugate also elicited an accelerated and enhanced HA head specific IgG response, which was significantly higher than the soluble HA head, or the admixture of P22 and HA head without the need for adjuvants. Thus, our results show that the HA head can be easily prepared by in vitro refolding in a modified E. coli strain, maintaining its intact structure and enabling the induction of a strong immune response when conjugated to P22 VLPs, even when presented as a monomer. These results also demonstrate that the P22 VLPs can be rapidly modified in a modular fashion, resulting in an effective vaccine construct that can generate protective immunity without the need for additional adjuvants.
Collapse
Affiliation(s)
- Jhanvi Sharma
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Kelly Shepardson
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Laura L Johns
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Julia Wellham
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - John Avera
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
- Matrivax Research and Development Corporation, Boston, Massachusetts 02118, United Sates
| | - Benjamin Schwarz
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
- Immunity to Pulmonary Pathogens section, Laboratory of Bacteriology, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana 59840, United States
| | - Agnieszka Rynda-Apple
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Trevor Douglas
- Department of Chemistry, Indiana University, 800 E. Kirkwood Avenue, Bloomington, Indiana 47405, United States
| |
Collapse
|
12
|
Jawinski K, Hartmann M, Singh C, Kinnear E, Busse DC, Ciabattini A, Fiorino F, Medaglini D, Trombetta CM, Montomoli E, Contreras V, Le Grand R, Coiffier C, Primard C, Verrier B, Tregoning JS. Recombinant Haemagglutinin Derived From the Ciliated Protozoan Tetrahymena thermophila Is Protective Against Influenza Infection. Front Immunol 2019; 10:2661. [PMID: 31798589 PMCID: PMC6863932 DOI: 10.3389/fimmu.2019.02661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Current influenza vaccines manufactured using eggs have considerable limitations, both in terms of scale up production and the potential impact passaging through eggs can have on the antigenicity of the vaccine virus strains. Alternative methods of manufacture are required, particularly in the context of an emerging pandemic strain. Here we explore the production of recombinant influenza haemagglutinin using the ciliated protozoan Tetrahymena thermophila. For the first time we were able to produce haemagglutinin from both seasonal influenza A and B strains. This ciliate derived material was immunogenic, inducing an antibody response in both mice and non-human primates. Mice immunized with ciliate derived haemagglutinin were protected against challenge with homologous influenza A or B viruses. The antigen could also be combined with submicron particles containing a Nod2 ligand, significantly boosting the immune response and reducing the dose of antigen required. Thus, we show that Tetrahymena can be used as a manufacturing platform for viral vaccine antigens.
Collapse
Affiliation(s)
| | | | - Charanjit Singh
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Ekaterina Kinnear
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - David C Busse
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy.,VisMederi s.r.l., Siena, Italy
| | - Vanessa Contreras
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Roger Le Grand
- CEA-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Celine Coiffier
- Laboratoire de Biologie Tissulaire et d'Ingénierie Thérapeutique, UMR 5305, Université Lyon 1, CNRS, IBCP, Lyon, France
| | | | | | - John S Tregoning
- Department of Infectious Disease, St Mary's Campus, Imperial College London, London, United Kingdom
| |
Collapse
|
13
|
Trombetta CM, Marchi S, Manini I, Lazzeri G, Montomoli E. Challenges in the development of egg-independent vaccines for influenza. Expert Rev Vaccines 2019; 18:737-750. [DOI: 10.1080/14760584.2019.1639503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Giacomo Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| |
Collapse
|
14
|
Kingstad-Bakke BA, Chandrasekar SS, Phanse Y, Ross KA, Hatta M, Suresh M, Kawaoka Y, Osorio JE, Narasimhan B, Talaat AM. Effective mosaic-based nanovaccines against avian influenza in poultry. Vaccine 2019; 37:5051-5058. [PMID: 31300285 DOI: 10.1016/j.vaccine.2019.06.077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/15/2019] [Accepted: 06/24/2019] [Indexed: 01/09/2023]
Abstract
Avian influenza virus (AIV) is an extraordinarily diverse pathogen that causes significant morbidity in domesticated poultry populations and threatens human life with looming pandemic potential. Controlling avian influenza in susceptible populations requires highly effective, economical and broadly reactive vaccines. Several AIV vaccines have proven insufficient despite their wide use, and better technologies are needed to improve their immunogenicity and broaden effectiveness. Previously, we developed a "mosaic" H5 subtype hemagglutinin (HA) AIV vaccine and demonstrated its broad protection against diverse highly pathogenic H5N1 and seasonal H1N1 virus strains in mouse and non-human primate models. There is a significant interest in developing effective and safe vaccines against AIV that cannot contribute to the emergence of new strains of the virus once circulating in poultry. Here, we report on the development of an H5 mosaic (H5M) vaccine antigen formulated with polyanhydride nanoparticles (PAN) that provide sustained release of encapsulated antigens. H5M vaccine constructs were immunogenic whether delivered by the modified virus Ankara (MVA) strain or encapsulated within PAN. Both humoral and cellular immune responses were generated in both specific-pathogen free (SPF) and commercial chicks. Importantly, chicks vaccinated by H5M constructs were protected in terms of viral shedding from divergent challenge with a low pathogenicity avian influenza (LPAI) strain at 8 weeks post-vaccination. In addition, protective levels of humoral immunity were generated against highly pathogenic avian influenza (HPAI) of the similar H5N1 and genetically dissimilar H5N2 viruses. Overall, the developed platform technologies (MVA vector and PAN encapsulation) were safe and provided high levels of sustained protection against AIV in chickens. Such approaches could be used to design more efficacious vaccines against other important poultry infections.
Collapse
Affiliation(s)
- Brock A Kingstad-Bakke
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA; Pan Genome Systems, Madison, WI, USA
| | - Shaswath S Chandrasekar
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | | | - Kathleen A Ross
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Masato Hatta
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - M Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Jorge E Osorio
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Adel M Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, USA; Pan Genome Systems, Madison, WI, USA.
| |
Collapse
|
15
|
Cheng A, Hsieh SM, Pan SC, Li YH, Hsieh EF, Lee HC, Lin TW, Lai KL, Chen C, Shi-Chung Chang S, Chang SC. The safety and immunogenicity of a cell-derived adjuvanted H5N1 vaccine - A phase I randomized clinical trial. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:685-692. [PMID: 31255574 DOI: 10.1016/j.jmii.2019.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 02/09/2019] [Accepted: 03/25/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Development of an efficacious egg-free mock-up H5N1 vaccine is key to our preparedness against pandemic avian flu. METHODS This is a single-center, randomized, observer-blinded phase I clinical trial evaluating the safety and immunogenicity of an alum-adjuvanted Madin-Darby canine kidney (MDCK)-derived inactivated whole-virion H5N1 influenza vaccine in healthy adults. Hemagglutination inhibition (HAI) and neutralizing antibody titers were measured using horse and turkey red blood cells (RBCs). RESULTS Thirty-six adult subjects were randomized to receive two doses of 0.5 mL of the MDCK-derived H5N1 alum-adjuvanted vaccine containing 7.5, 15, or 30 μg of hemagglutinin (HA) 21 days apart. The candidate vaccine was well tolerated and safe across the three dosing groups. The most frequent adverse event was injection site pain (46.5%). Both HAI and neutralizing antibody titers increased after each vaccination in all three dosing groups. The best HAI responses, namely a seroconversion rate of 91.7% and a geometric mean ratio of 9.51 were achieved with the HA dose of 30 μg assayed using horse RBCs at day 42. HAI titers against H5N1 avian influenza virus was significantly higher when measured using horse RBCs compared with turkey RBCs. CONCLUSIONS This Phase I trial showed the MDCK-derived H5N1 candidate vaccine is safe and immunogenic. The source of RBCs has a significant impact on the measurement of HAI titers (ClinicalTrials.gov number: NCT01675284.).
Collapse
Affiliation(s)
- Aristine Cheng
- Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Szu-Min Hsieh
- Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Ching Pan
- Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Han Li
- Medigen Vaccine Biologics Corporation, Hsinchu, Taiwan
| | | | | | - Ting-Wan Lin
- Medigen Vaccine Biologics Corporation, Hsinchu, Taiwan
| | | | - Charles Chen
- Medigen Vaccine Biologics Corporation, Hsinchu, Taiwan
| | | | - Shan-Chwen Chang
- Department of Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
16
|
Xu X, Ding Z, Yuan Q, Ding J, Li J, Wang W, Cong Y, Ouyang W, Wang Y, Qian J, Yin R. A genotype VII Newcastle disease virus-like particles confer full protection with reduced virus load and decreased virus shedding. Vaccine 2018; 37:444-451. [PMID: 30545716 DOI: 10.1016/j.vaccine.2018.11.068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/17/2018] [Accepted: 11/25/2018] [Indexed: 11/24/2022]
Abstract
Newcastle disease (ND) is one of the most severe avian infectious disease inflicting a great loss on poultry industry worldwide. The control of ND relies on proper vaccination strategies. The vaccine strains of Newcastle disease virus (NDV) mainly belong to genotype I, II or III, which cannot fully prohibit virus shedding against the prevalent genotype VII virulent strain attack. To develop a safe, genotype matched vaccine candidate, we employed a bac-to-bac expression system and constructed a genotype VII NDV strain based virus-like particles (NDV VLPs). It was constructed with NDV M protein as the skeleton, and protective antigen F and HN proteins displayed on the surface. The NDV VLPs exhibited a similar appearance to the live NDV particles, but with denser F and HN proteins displayed on the surface. The immunization assay indicated that NDV VLPs stimulated a longer protection period, less tissue virus loading and shorter virus shedding period than the commercialized LaSota-formulated vaccine when challenged with genotype VII NDV strain. These results proposed the potential role of NDV VLPs as an alternative to current live genotype unmatched vaccine for the control and eliminate NDV in the avian flocks.
Collapse
Affiliation(s)
- Xiaohong Xu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Zhuang Ding
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Qianliang Yuan
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Jiaxin Ding
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Jindou Li
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Weiqi Wang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Yanlong Cong
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China
| | - Wei Ouyang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences/Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture, Nanjing 210014, China
| | - Yongshan Wang
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences/Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture, Nanjing 210014, China
| | - Jing Qian
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China; Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences/Key Laboratory for Veterinary Bio-Product Engineering, Ministry of Agriculture, Nanjing 210014, China
| | - Renfu Yin
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun 130062, China.
| |
Collapse
|
17
|
Keshavarz M, Mirzaei H, Salemi M, Momeni F, Mousavi MJ, Sadeghalvad M, Arjeini Y, Solaymani-Mohammadi F, Sadri Nahand J, Namdari H, Mokhtari-Azad T, Rezaei F. Influenza vaccine: Where are we and where do we go? Rev Med Virol 2018; 29:e2014. [PMID: 30408280 DOI: 10.1002/rmv.2014] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/22/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022]
Abstract
The alarming rise of morbidity and mortality caused by influenza pandemics and epidemics has drawn attention worldwide since the last few decades. This life-threatening problem necessitates the development of a safe and effective vaccine to protect against incoming pandemics. The currently available flu vaccines rely on inactivated viral particles, M2e-based vaccine, live attenuated influenza vaccine (LAIV) and virus like particle (VLP). While inactivated vaccines can only induce systemic humoral responses, LAIV and VLP vaccines stimulate both humoral and cellular immune responses. Yet, these vaccines have limited protection against newly emerging viral strains. These strains, however, can be targeted by universal vaccines consisting of conserved viral proteins such as M2e and capable of inducing cross-reactive immune response. The lack of viral genome in VLP and M2e-based vaccines addresses safety concern associated with existing attenuated vaccines. With the emergence of new recombinant viral strains each year, additional effort towards developing improved universal vaccine is warranted. Besides various types of vaccines, microRNA and exosome-based vaccines have been emerged as new types of influenza vaccines which are associated with new and effective properties. Hence, development of a new generation of vaccines could contribute to better treatment of influenza.
Collapse
Affiliation(s)
- Mohsen Keshavarz
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Salemi
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Fatemeh Momeni
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Mousavi
- Department of Immunology and Allergy, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.,Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Sadeghalvad
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yaser Arjeini
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Solaymani-Mohammadi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Javid Sadri Nahand
- Department of Medical Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haideh Namdari
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Wang Y, Deng L, Kang SM, Wang BZ. Universal influenza vaccines: from viruses to nanoparticles. Expert Rev Vaccines 2018; 17:967-976. [PMID: 30365905 DOI: 10.1080/14760584.2018.1541408] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The current seasonal influenza vaccine confers only limited protection due to waning antibodies or the antigenic shift and drift of major influenza surface antigens. A universal influenza vaccine which induces broad cross-protection against divergent influenza viruses with a comparable or better efficacy to seasonal influenza vaccines against matched strains will negate the need for an annual update of vaccine strains and protect against possible influenza pandemics. AREAS COVERED In this review, we summarize the recent progress in nanoparticle-based universal influenza vaccine development. We compared the most potent nanoparticle categories, focusing on how they encapsulate conserved influenza epitopes, stimulate the innate and adaptive immune systems, exhibit antigen depot effect, extend the period for antigen-processing and presentation, and exert an intrinsic adjuvant effect on inducing robust immune responses. EXPERT COMMENTARY The development of an effective universal influenza vaccine is an urgent task. Traditional influenza vaccine approaches are not sufficient for preventing recurrent epidemics or occasional pandemics. Nanoparticles are compatible with different immunogens and immune stimulators and can overcome the intrinsically low immunogenicity of conserved influenza virus antigens. We foresee that an affordable universal influenza vaccine will be available within ten years by integrating nanoparticles with other targeted delivery and controlled release technology.
Collapse
Affiliation(s)
- Ye Wang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Lei Deng
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Sang-Moo Kang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| | - Bao-Zhong Wang
- a Center for Inflammation, Immunity & Infection , Georgia State University Institute for Biomedical Sciences , Atlanta , GA , USA
| |
Collapse
|
19
|
Pérez Rubio A, Eiros JM. Cell culture-derived flu vaccine: Present and future. Hum Vaccin Immunother 2018; 14:1874-1882. [PMID: 29672213 PMCID: PMC6149758 DOI: 10.1080/21645515.2018.1460297] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 03/12/2018] [Accepted: 03/29/2018] [Indexed: 12/14/2022] Open
Abstract
The benefit of influenza vaccines is difficult to estimate due to the complexity of accurately assessing the burden of influenza. To improve the efficacy of influenza vaccines, vaccine manufacturers have developed quadrivalent influenza vaccine (QIV) formulations for seasonal vaccination by including both influenza B lineages. Three parallel approaches for producing influenza vaccines are attracting the interest of many vaccine manufacturing companies. The first and oldest is the conventional egg-derived influenza vaccine, which is used by the current licensed influenza vaccines. The second approach is a cell culture-derived influenza vaccine, and the third and most recent is synthetic vaccines. Here, we analyze the difficulties with vaccines production in eggs and compare this to cell culture-derived influenza vaccines and discuss the future of cell culture-derived QIVs.
Collapse
Affiliation(s)
| | - Jose María Eiros
- Servicio Microbiología, Hospital Universitario Rio Hortega, Valladolid, Spain
| |
Collapse
|
20
|
Huang X, Karabudak A, Comber JD, Philip M, Morcol T, Philip R. A novel immunization approach for dengue infection based on conserved T cell epitopes formulated in calcium phosphate nanoparticles. Hum Vaccin Immunother 2017; 13:2612-2625. [PMID: 28933657 DOI: 10.1080/21645515.2017.1369639] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Dengue virus (DV) is the etiologic agent of dengue fever, the most significant mosquito-borne viral disease in humans. Most DV vaccine approaches are focused on generating antibody mediated responses; one such DV vaccine is approved for use in humans but its efficacy is limited. While it is clear that T cell responses play important role in DV infection and subsequent disease manifestations, fewer studies are aimed at developing vaccines that induce robust T cells responses. Potent T cell based vaccines require 2 critical components: the identification of specific T cell stimulating MHC associated peptides, and an optimized vaccine delivery vehicle capable of simultaneously delivering the antigens and any required adjuvants. We have previously identified and characterized DV specific HLA-A2 and -A24 binding DV serotypes conserved epitopes, and the feasibility of an epitope based vaccine for DV infection. In this study, we build on those previous studies and describe an investigational DV vaccine using T cell epitopes incorporated into a calcium phosphate nanoparticle (CaPNP) delivery system. This study presents a comprehensive analysis of functional immunogenicity of DV CaPNP/multipeptide formulations in vitro and in vivo and demonstrates the CaPNP/multipeptide vaccine is capable of inducing T cell responses against all 4 serotypes of DV. This synthetic vaccine is also cost effective, straightforward to manufacture, and stable at room temperature in a lyophilized form. This formulation may serve as an effective candidate DV vaccine that protects against all 4 serotypes as either a prophylactic or therapeutic vaccine.
Collapse
Affiliation(s)
| | | | | | | | - Tulin Morcol
- b Captivate Pharmaceuticals , Doylestown , PA , USA
| | | |
Collapse
|
21
|
Manini I, Trombetta CM, Lazzeri G, Pozzi T, Rossi S, Montomoli E. Egg-Independent Influenza Vaccines and Vaccine Candidates. Vaccines (Basel) 2017; 5:E18. [PMID: 28718786 PMCID: PMC5620549 DOI: 10.3390/vaccines5030018] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 11/16/2022] Open
Abstract
Vaccination remains the principal way to control seasonal infections and is the most effective method of reducing influenza-associated morbidity and mortality. Since the 1940s, the main method of producing influenza vaccines has been an egg-based production process. However, in the event of a pandemic, this method has a significant limitation, as the time lag from strain isolation to final dose formulation and validation is six months. Indeed, production in eggs is a relatively slow process and production yields are both unpredictable and highly variable from strain to strain. In particular, if the next influenza pandemic were to arise from an avian influenza virus, and thus reduce the egg-laying hen population, there would be a shortage of embryonated eggs available for vaccine manufacturing. Although the production of egg-derived vaccines will continue, new technological developments have generated a cell-culture-based influenza vaccine and other more recent platforms, such as synthetic influenza vaccines.
Collapse
Affiliation(s)
- Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Claudia Maria Trombetta
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Giacomo Lazzeri
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Teresa Pozzi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Stefania Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
- VisMederi S.r.l., Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy.
| |
Collapse
|
22
|
Edgue G, Twyman RM, Beiss V, Fischer R, Sack M. Antibodies from plants for bionanomaterials. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9. [DOI: 10.1002/wnan.1462] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/05/2017] [Accepted: 01/16/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Gueven Edgue
- Department of Molecular Biotechnology; RWTH Aachen University; Aachen Germany
| | | | - Veronique Beiss
- Department of Molecular Biotechnology; RWTH Aachen University; Aachen Germany
| | - Rainer Fischer
- Department of Molecular Biotechnology; RWTH Aachen University; Aachen Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME; Aachen Germany
| | - Markus Sack
- Department of Molecular Biotechnology; RWTH Aachen University; Aachen Germany
| |
Collapse
|
23
|
Wang L, Chang TZ, He Y, Kim JR, Wang S, Mohan T, Berman Z, Tompkins SM, Tripp RA, Compans RW, Champion JA, Wang BZ. Coated protein nanoclusters from influenza H7N9 HA are highly immunogenic and induce robust protective immunity. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2017; 13:253-262. [PMID: 27622321 PMCID: PMC5237404 DOI: 10.1016/j.nano.2016.09.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 08/05/2016] [Accepted: 09/01/2016] [Indexed: 01/08/2023]
Abstract
Recurring influenza viruses pose an annual threat to public health. A time-saving, cost-effective and egg-independent influenza vaccine approach is important particularly when responding to an emerging pandemic. We fabricated coated, two-layer protein nanoclusters from recombinant trimeric hemagglutinin from an avian-origin H7N9 influenza A virus as an approach for vaccine development in response to an emerging pandemic. Assessment of the virus-specific immune responses and protective efficacy in mice immunized with the nanoclusters demonstrated that the vaccine candidates were highly immunogenic, able to induce protective immunity and long-lasting humoral antibody responses to this virus without the use of adjuvants. Because the advantages of the highly immunogenic coated nanoclusters also include rapid productions in an egg-independent system, this approach has great potential for influenza vaccine production not only in response to an emerging pandemic, but also as a replacement for conventional seasonal influenza vaccines.
Collapse
Affiliation(s)
- Li Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Timothy Z Chang
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA.
| | - Yuan He
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Jong R Kim
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Shelly Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Teena Mohan
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - Zachary Berman
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Julie A Champion
- Georgia Institute of Technology, School of Chemical & Biomolecular Engineering, Atlanta, GA, USA.
| | - Bao-Zhong Wang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA; Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, Atlanta, GA, USA.
| |
Collapse
|
24
|
Vector-based genetically modified vaccines: Exploiting Jenner's legacy. Vaccine 2016; 34:6436-6448. [PMID: 28029542 PMCID: PMC7115478 DOI: 10.1016/j.vaccine.2016.06.059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/02/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022]
Abstract
The global vaccine market is diverse while facing a plethora of novel developments. Genetic modification (GM) techniques facilitate the design of ’smarter’ vaccines. For many of the major infectious diseases of humans, like AIDS and malaria, but also for most human neoplastic disorders, still no vaccines are available. It may be speculated that novel GM technologies will significantly contribute to their development. While a promising number of studies is conducted on GM vaccines and GM vaccine technologies, the contribution of GM technology to newly introduced vaccines on the market is disappointingly limited. In this study, the field of vector-based GM vaccines is explored. Data on currently available, actually applied, and newly developed vectors is retrieved from various sources, synthesised and analysed, in order to provide an overview on the use of vector-based technology in the field of GM vaccine development. While still there are only two vector-based vaccines on the human vaccine market, there is ample activity in the fields of patenting, preclinical research, and different stages of clinical research. Results of this study revealed that vector-based vaccines comprise a significant part of all GM vaccines in the pipeline. This study further highlights that poxviruses and adenoviruses are among the most prominent vectors in GM vaccine development. After the approval of the first vectored human vaccine, based on a flavivirus vector, vaccine vector technology, especially based on poxviruses and adenoviruses, holds great promise for future vaccine development. It may lead to cheaper methods for the production of safe vaccines against diseases for which no or less perfect vaccines exist today, thus catering for an unmet medical need. After the introduction of Jenner’s vaccinia virus as the first vaccine more than two centuries ago, which eventually led to the recent eradication of smallpox, this and other viruses may now be the basis for constructing vectors that may help us control other major scourges of mankind.
Collapse
|
25
|
Pillet S, Racine T, Nfon C, Di Lenardo TZ, Babiuk S, Ward BJ, Kobinger GP, Landry N. Plant-derived H7 VLP vaccine elicits protective immune response against H7N9 influenza virus in mice and ferrets. Vaccine 2015; 33:6282-9. [PMID: 26432915 DOI: 10.1016/j.vaccine.2015.09.065] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/18/2015] [Accepted: 09/19/2015] [Indexed: 12/23/2022]
Abstract
In March 2013, the Chinese Centre for Disease Control and Prevention confirmed the first reported case of human infection with an avian influenza A H7N9 virus. Infection with this virus often caused severe pneumonia and acute respiratory distress syndrome resulting in a case fatality rate >35%. The risk of pandemic highlighted, once again, the need for a more rapid and scalable vaccine response capability. Here, we describe the rapid (19 days) development of a plant-derived VLP vaccine based on the hemagglutinin sequence of influenza H7N9 A/Hangzhou/1/2013. The immunogenicity of the H7 VLP vaccine was assessed in mice and ferrets after one or two intramuscular dose(s) with and without adjuvant (alum or GLA-SE™). In ferrets, we also measured H7-specific cell-mediated immunity. The mice and ferrets were then challenged with H7N9 A/Anhui/1/2013 influenza virus. A single immunization with the adjuvanted vaccine elicited a strong humoral response and protected mice against an otherwise lethal challenge. Two doses of unadjuvanted vaccine significantly increased humoral response and resulted in 100% protection with significant reduction of clinical signs leading to nearly asymptomatic infections. In ferrets, a single immunization with the alum-adjuvanted H7 VLP vaccine induced strong humoral and CMI responses with antigen-specific activation of CD3(+) T cells. Compared to animals injected with placebo, ferrets vaccinated with alum-adjuvanted vaccine displayed no weight loss during the challenge. Moreover, the vaccination significantly reduced the viral load in lungs and nasal washes 3 days after the infection. This candidate plant-made H7 vaccine therefore induced protective responses after either one adjuvanted or two unadjuvanted doses. Studies are currently ongoing to better characterize the immune response elicited by the plant-derived VLP vaccines. Regardless, these data are very promising for the rapid production of an immunogenic and protective vaccine against this potentially pandemic virus.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- Body Weight
- Disease Models, Animal
- Female
- Ferrets
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunization Schedule
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Influenza Vaccines/immunology
- Influenza Vaccines/isolation & purification
- Injections, Intramuscular
- Lung/virology
- Male
- Mice, Inbred BALB C
- Nasal Cavity/virology
- Orthomyxoviridae Infections/pathology
- Orthomyxoviridae Infections/prevention & control
- Placebos/administration & dosage
- Plants, Genetically Modified
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Survival Analysis
- Nicotiana
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/isolation & purification
- Viral Load
Collapse
Affiliation(s)
- S Pillet
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada; Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - T Racine
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - C Nfon
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - T Z Di Lenardo
- Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - S Babiuk
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada
| | - B J Ward
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada
| | - G P Kobinger
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada; Department of Immunology, University of Manitoba, MB, Canada; Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - N Landry
- Medicago Inc., 1020 Route de l'Église, Bureau 600, Québec, QC, Canada.
| |
Collapse
|
26
|
Amen O, Vemula SV, Zhao J, Ibrahim R, Hussein A, Hewlett IK, Moussa S, Mittal SK. Identification and characterization of a highly pathogenic H5N1 avian influenza A virus during an outbreak in vaccinated chickens in Egypt. Virus Res 2015; 210:337-43. [PMID: 26363196 DOI: 10.1016/j.virusres.2015.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/07/2015] [Accepted: 09/04/2015] [Indexed: 11/19/2022]
Abstract
Highly pathogenic avian influenza A (HPAI) H5N1 viruses continue to be a major veterinary and public health problem in Egypt. Continued surveillance of these viruses is necessary to devise strategies to control the spread of the virus and to monitor its evolutionary patterns. This is a report of the identification of a variant strain of HPAI H5N1 virus during an outbreak in 2010 in vaccinated chicken flocks in a poultry farm in Assiut, Egypt. Vaccination of chickens with an oil-emulsified inactivated A/chicken/Mexico/232/94 (H5N2) vaccine induced high levels of hemagglutination inhibition (HI) antibody titers reaching up to 9 log2. However, all flocks irrespective of the number of vaccine doses and the resultant HI titer levels came down with severe influenza infections. The qRT-PCR and rapid antigen test confirmed the influenza virus to be from H5N1 subtype. Sequencing of the hemagglutinin (HA) gene fragment from ten independent samples demonstrated that a single H5N1 strain was involved. This strain belonged to clade 2.2.1 and had several mutations in the receptor-binding site of the HA protein, thereby producing a variant strain of HPAI H5N1 virus which was antigenically different from the parent clade 2.2.1 virus circulating in Egypt at that time. In order to define the variability in HPAI H5N1 viruses over time in Egypt, we sequenced another H5N1 virus that was causing infections in chickens in 2014. Phylogenetic analysis revealed that both viruses had further distanced from the parent virus circulating during 2010. This study highlights that the antigenic mutations in HPAI H5N1 viruses represent a definitive challenge for the development of an effective vaccine for poultry. Overall, the results emphasize the need for continued surveillance of H5N1 outbreaks and extensive characterization of virus isolates from vaccinated and non-vaccinated poultry populations to better understand genetic changes and their implications.
Collapse
Affiliation(s)
- O Amen
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Poultry Diseases Department, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - S V Vemula
- Laboratory of Molecular Virology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - J Zhao
- Laboratory of Molecular Virology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - R Ibrahim
- Poultry Diseases Department, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - A Hussein
- Department of Animal Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - I K Hewlett
- Laboratory of Molecular Virology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - S Moussa
- Poultry Diseases Department, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - S K Mittal
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
27
|
Production of influenza virus-like particles from stably transfected Trichoplusia ni BT1 TN-5B1-4 cells. BIOTECHNOL BIOPROC E 2015. [DOI: 10.1007/s12257-014-0768-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
28
|
Raven N, Rasche S, Kuehn C, Anderlei T, Klöckner W, Schuster F, Henquet M, Bosch D, Büchs J, Fischer R, Schillberg S. Scaled-up manufacturing of recombinant antibodies produced by plant cells in a 200-L orbitally-shaken disposable bioreactor. Biotechnol Bioeng 2015; 112:308-21. [PMID: 25117428 DOI: 10.1002/bit.25352] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/16/2014] [Accepted: 07/22/2014] [Indexed: 12/18/2022]
Abstract
Tobacco BY-2 cells have emerged as a promising platform for the manufacture of biopharmaceutical proteins, offering efficient protein secretion, favourable growth characteristics and cultivation in containment under a controlled environment. The cultivation of BY-2 cells in disposable bioreactors is a useful alternative to conventional stainless steel stirred-tank reactors, and orbitally-shaken bioreactors could provide further advantages such as simple bag geometry, scalability and predictable process settings. We carried out a scale-up study, using a 200-L orbitally-shaken bioreactor holding disposable bags, and BY-2 cells producing the human monoclonal antibody M12. We found that cell growth and recombinant protein accumulation were comparable to standard shake flask cultivation, despite a 200-fold difference in cultivation volume. Final cell fresh weights of 300-387 g/L and M12 yields of ∼20 mg/L were achieved with both cultivation methods. Furthermore, we established an efficient downstream process for the recovery of M12 from the culture broth. The viscous spent medium prevented clarification using filtration devices, but we used expanded bed adsorption (EBA) chromatography with SP Sepharose as an alternative for the efficient capture of the M12 antibody. EBA was introduced as an initial purification step prior to protein A affinity chromatography, resulting in an overall M12 recovery of 75-85% and a purity of >95%. Our results demonstrate the suitability of orbitally-shaken bioreactors for the scaled-up cultivation of plant cell suspension cultures and provide a strategy for the efficient purification of antibodies from the BY-2 culture medium.
Collapse
Affiliation(s)
- Nicole Raven
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Fischer R, Buyel JF, Schillberg S, Twyman RM. Molecular Farming in Plants: The Long Road to the Market. COMMERCIAL PLANT-PRODUCED RECOMBINANT PROTEIN PRODUCTS 2014. [DOI: 10.1007/978-3-662-43836-7_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Mallajosyula JK, Hiatt E, Hume S, Johnson A, Jeevan T, Chikwamba R, Pogue GP, Bratcher B, Haydon H, Webby RJ, McCormick AA. Single-dose monomeric HA subunit vaccine generates full protection from influenza challenge. Hum Vaccin Immunother 2013; 10:586-95. [PMID: 24378714 DOI: 10.4161/hv.27567] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recombinant subunit vaccines are an efficient strategy to meet the demands of a possible influenza pandemic, because of rapid and scalable production. However, vaccines made from recombinant hemagglutinin (HA) subunit protein are often of low potency, requiring high dose or boosting to generate a sustained immune response. We have improved the immunogenicity of a plant-made HA vaccine by chemical conjugation to the surface of the Tobacco mosaic virus (TMV) which is non infectious in mammals. We have previously shown that TMV is taken up by mammalian dendritic cells and is a highly effective antigen carrier. In this work, we tested several TMV-HA conjugation chemistries, and compared immunogenicity in mice as measured by anti-HA IgG titers and hemagglutination inhibition (HAI). Importantly, pre-existing immunity to TMV did not reduce initial or boosted titers. Further optimization included dosing with and without alum or oil-in water adjuvants. Surprisingly, we were able to stimulate potent immunogenicity and HAI titers with a single 15 µg dose of HA as a TMV conjugate. We then evaluated the efficacy of the TMV-HA vaccine in a lethal virus challenge in mice. Our results show that a single dose of the TMV-HA conjugate vaccine is sufficient to generate 50% survival, or 100% survival with adjuvant, compared with 10% survival after vaccination with a commercially available H1N1 vaccine. TMV-HA is an effective dose-sparing influenza vaccine, using a single-step process to rapidly generate large quantities of highly effective flu vaccine from an otherwise low potency HA subunit protein.
Collapse
Affiliation(s)
| | - Ernie Hiatt
- Kentucky BioProcessing LLC; Owensboro, KY USA
| | - Steve Hume
- Kentucky BioProcessing LLC; Owensboro, KY USA
| | | | | | - Rachel Chikwamba
- Council for Scientific and Industrial Research; Pretoria, South Africa
| | - Gregory P Pogue
- Kentucky BioProcessing LLC; Owensboro, KY USA; IC2 Institute; The University of Texas at Austin; Austin, TX USA
| | | | - Hugh Haydon
- Kentucky BioProcessing LLC; Owensboro, KY USA
| | | | | |
Collapse
|
31
|
Vemula SV, Amen O, Katz JM, Donis R, Sambhara S, Mittal SK. Beta-defensin 2 enhances immunogenicity and protection of an adenovirus-based H5N1 influenza vaccine at an early time. Virus Res 2013; 178:398-403. [PMID: 24051000 DOI: 10.1016/j.virusres.2013.09.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 08/30/2013] [Accepted: 09/06/2013] [Indexed: 11/16/2022]
Abstract
Reports of human infections with highly pathogenic H5N1 avian influenza viruses in many countries in Asia and Africa with varying case fatality rates highlight the pandemic potential of these viruses. In order to contain a rapidly spreading influenza virus in a pandemic scenario, a vaccine which can induce rapid and robust immune responses, preferably in a single dose, is necessary. Murine beta-defensin 2 (Mbd2), a small molecular weight protein expressed by epithelial cells, has been shown to enhance antigen-specific immune responses by recruiting and activating professional antigen presenting cells to the site of vaccination. This study assessed the potential of Mbd2 to enhance the immunogenicity and protective efficacy of a human adenovirus (HAd)-based vaccine expressing the hemagglutinin (HA) and nucleoprotein (NP) [HAd-HA-NP] of an H5N1 influenza virus. A single inoculation of mice with both HAd-HA-NP and a HAd vector expressing Murine β-defensin 2 (HAd-Mbd2) resulted in significantly higher levels of both humoral and cell-mediated immune responses compared to the groups vaccinated only with HAd-HA-NP. These responses were evident even at day 7 post-immunization. Furthermore, the HAd-HA-NP+HAd-Mbd2-immunized group receiving the lowest vector dose (2 × 10(7)+1 × 10(7)) was completely protected against an rgH5N1 virus challenge on day 7 post-vaccination. These results highlight the potential of Mbd2 as a genetic adjuvant in inducing rapid and robust immune responses to a HAd-based vaccine.
Collapse
Affiliation(s)
- Sai V Vemula
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, USA
| | | | | | | | | | | |
Collapse
|
32
|
Fast Single-Use VLP Vaccine Productions Based on Insect Cells and the Baculovirus Expression Vector System: Influenza as Case Study. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2013; 138:99-125. [DOI: 10.1007/10_2013_186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Mooney AJ, Tompkins SM. Experimental vaccines against potentially pandemic and highly pathogenic avian influenza viruses. Future Virol 2013; 8:25-41. [PMID: 23440999 PMCID: PMC3579652 DOI: 10.2217/fvl.12.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Influenza A viruses continue to emerge and re-emerge, causing outbreaks, epidemics and occasionally pandemics. While the influenza vaccines licensed for public use are generally effective against seasonal influenza, issues arise with production, immunogenicity, and efficacy in the case of vaccines against pandemic and emerging influenza viruses, and highly pathogenic avian influenza virus in particular. Thus, there is need of improved influenza vaccines and vaccination strategies. This review discusses advances in alternative influenza vaccines, touching briefly on licensed vaccines and vaccine antigens; then reviewing recombinant subunit vaccines, virus-like particle vaccines and DNA vaccines, with the main focus on virus-vectored vaccine approaches.
Collapse
Affiliation(s)
- Alaina J Mooney
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| | - S Mark Tompkins
- Department of Infectious Diseases, University of Georgia, 111 Carlton St, Athens, GA 30602, USA
| |
Collapse
|
34
|
Chan CYY, Tambyah PA. Preflucel®: a Vero-cell culture-derived trivalent influenza vaccine. Expert Rev Vaccines 2012; 11:759-73. [PMID: 22913252 DOI: 10.1586/erv.12.55] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Vaccination is the principal means to reduce the impact of influenza infection. Effective vaccination programs require a reliable and safe production system. Traditionally, influenza vaccines are produced in embryonated chicken eggs. Over the last two decades, new cell culture-derived vaccines have been licensed and manufactured, and other vaccines are still in various phases of development. Vero cells have been used for the development of a wide variety of vaccines including influenza vaccines. Pandemic and avian influenza vaccines derived from Vero cells have been shown to be well tolerated and immunogenic in animal and Phase I-II clinical studies. A Phase III randomized, double-blind, placebo-controlled trial of a trivalent influenza vaccine produced in Vero-cell culture was conducted in 7250 adults aged 18-49 years. Overall protective efficacy for antigenically matched influenza vaccine was 78.5%. The vaccine was well tolerated with no treatment-related serious adverse events and compared favorably with egg-derived vaccines from previous trials. Vero-cell-derived influenza vaccines have the potential to be an important parts of the influenza vaccine strategy, especially if an avian-derived strain becomes predominant or the demand outstrips the capacity of egg-based production systems.
Collapse
Affiliation(s)
- Candice Yuen-Yue Chan
- Division of Infectious Diseases, Department of Medicine, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| | | |
Collapse
|
35
|
Employing Live Microbes for Vaccine Delivery. DEVELOPMENT OF NOVEL VACCINES 2012. [PMCID: PMC7123214 DOI: 10.1007/978-3-7091-0709-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
|
36
|
Abstract
Manufacturing of cell culture-derived virus particles for vaccination and gene therapy is a rapidly growing field in the biopharmaceutical industry. The process involves a number of complex tasks and unit operations ranging from selection of host cells and virus strains for the cultivation in bioreactors to the purification and formulation of the final product. For the majority of cell culture-derived products, efforts focused on maximization of bioreactor yields, whereas design and optimization of downstream processes were often neglected. Owing to this biased focus, downstream procedures today often constitute a bottleneck in various manufacturing processes and account for the majority of the overall production costs. For efficient production methods, particularly in sight of constantly increasing economic pressure within human healthcare systems, highly productive downstream schemes have to be developed. Here, we discuss unit operations and downstream trains to purify virus particles for use as vaccines and vectors for gene therapy.
Collapse
Affiliation(s)
- Michael W Wolf
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany.
| | | |
Collapse
|
37
|
Song JM, Choi CW, Kwon SO, Compans RW, Kang SM, Kim SI. Proteomic characterization of influenza H5N1 virus-like particles and their protective immunogenicity. J Proteome Res 2011; 10:3450-9. [PMID: 21688770 DOI: 10.1021/pr200086v] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recombinant virus-like particles (VLPs) have been shown to induce protective immunity. Despite their potential significance as promising vaccine candidates, the protein composition of VLPs produced in insect cells has not been well characterized. Here we report a proteomic analysis of influenza VLPs containing hemagglutinin (HA) and matrix M1 proteins from a human isolate of avian influenza H5N1 virus (H5 VLPs) produced in insect cells using the recombinant baculovirus expression system. Comprehensive proteomic analysis of purified H5 VLPs identified viral proteins and 37 additional host-derived proteins, many of which are known to be present in other enveloped viruses. Proteins involved in different cellular structures and functions were found to be present in H5 VLPs including those from the cytoskeleton, translation, chaperone, and metabolism. Immunization with purified H5 VLPs induced protective immunity, which was comparable to the inactivated whole virus containing all viral components. Unpurified H5 VLPs containing excess amounts of noninfluenza soluble proteins also conferred 100% protection against lethal challenge although lower immune responses were induced. These results provide important implications consistent with the idea that VLP production in insect cells may involve similar cellular machinery as other RNA enveloped viruses during synthesis, assembly, trafficking, and budding processes.
Collapse
Affiliation(s)
- Jae-Min Song
- Division of Life Science, Korea Basic Science Institute, Daejeon, 305-333, South Korea
| | | | | | | | | | | |
Collapse
|
38
|
Sharma A, Bangari DS, Tandon M, Hogenesch H, Mittal SK. Evaluation of innate immunity and vector toxicity following inoculation of bovine, porcine or human adenoviral vectors in a mouse model. Virus Res 2010; 153:134-42. [PMID: 20659505 DOI: 10.1016/j.virusres.2010.07.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/15/2010] [Accepted: 07/17/2010] [Indexed: 01/16/2023]
Abstract
Nonhuman adenovirus (Ad) vectors derived from bovine Ad serotype 3 (BAd3) or porcine Ad serotype 3 (PAd3) can circumvent pre-existing immunity against human Ad (HAd). We have previously reported differential transduction of human and nonhuman cells by these Ad vectors, and their distinct receptor usage and biodistribution. To compare the induction of innate immunity, vector toxicity and vector uptake by Kupffer cells (KCs) following intravenous administration of PAd3, BAd3, or HAd5 vectors in mice, we determined mRNA expression levels of proinflammatory chemokines and cytokines, and Toll-like receptors (TLRs) in the liver and spleen. Tissue toxicity of these vectors was assessed by comparing serum levels of liver-specific enzymes, histopathology and Kupffer cell (KC) depletion. Compared to the HAd5 vector, PAd3 and BAd3 vectors were more potent stimulators of innate immune responses as indicated by enhanced mRNA expression of TLRs and proinflammatory chemokines and cytokine genes. Histopathological changes in the liver were most pronounced in HAd5-inoculated mice while BAd3- or PAd3-inoculated mice revealed mild histologic changes that were confined to early time points. Inoculation with HAd5 or PAd3 vectors resulted in a significant (P<0.05) decline of the number of KCs in the liver. Together, these results extend our previous observations regarding distinct in vivo biology of nonhuman and human Ad vectors.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Comparative Pathobiology, School of Veterinary Medicine, and Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | |
Collapse
|
39
|
Wu CY, Yeh YC, Yang YC, Chou C, Liu MT, Wu HS, Chan JT, Hsiao PW. Mammalian expression of virus-like particles for advanced mimicry of authentic influenza virus. PLoS One 2010; 5:e9784. [PMID: 20339535 PMCID: PMC2842297 DOI: 10.1371/journal.pone.0009784] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 02/28/2010] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Influenza A viruses are major human and animal pathogens with huge economic and societal impact from illness, hospitalizations, and deaths. Virus-like particles (VLPs) of influenza virus have been suggested as a vaccine candidate offering improved safety and efficacy. To develop this concept further, we established a flexible platform to efficiently generate different subtypes of mammalian-expressed influenza VLPs. Here we demonstrate that these mammalian VLPs strongly resemble the authentic viruses in structure, particle size and composition of host factors, and even glycosylation of viral antigens. METHODOLOGY/PRINCIPAL FINDINGS In this study, a mammalian VLP system was established by stable co-expression of four influenza structural proteins (HA, NA, M1, and M2) in a Vero cell line. By replacing the surface glycoproteins of HA and NA, we converted the H3N2-VLP subtype to H5N1-VLP. After centrifugation purification of conditioned media, the particle morphologies, average sizes, and hemagglutination abilities of secreted VLPs were characterized, and the VLP constituents were identified by LC/MS/MS. Protease protection assays demonstrated that specific cellular proteins that co-purified with influenza virions were integrated into mammalian VLPs. The glycosylation profiles of mammalian VLPs as revealed by deglycosylation assays were similar to that of progeny viruses produced from Vero cells. Vaccination of mice with 2.5 microg and above of H5N1-VLP elicited H5-specific IgG1 antibodies and resulted in full protection against lethal infection with homologous virus. These results provide compelling evidence that mammalian VLPs closely emulate the exterior of authentic virus particles not only in antigen presentation but also in biological properties and should provide promising vaccine candidates. CONCLUSIONS/SIGNIFICANCE This flexible mammalian influenza VLP system offers a superior alternative to the conventional reverse genetic vaccine platform without concerns over inadequate presentation of immune antigens or limitations imposed by the manipulation of real viruses.
Collapse
Affiliation(s)
- Chia-Ying Wu
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Chun Yeh
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chih Yang
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Ching Chou
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| | - Ming-Tsan Liu
- Center for Disease Control, Department of Health, Taipei, Taiwan
| | - Ho-Sheng Wu
- Center for Disease Control, Department of Health, Taipei, Taiwan
| | | | - Pei-Wen Hsiao
- Agricultural Biotech Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|