1
|
Monreal-Escalante E, Angulo M, Ramos-Vega A, Trujillo E, Angulo C. Plant-made trained immunity-based vaccines: Beyond one approach. Int J Pharm 2025; 675:125572. [PMID: 40204041 DOI: 10.1016/j.ijpharm.2025.125572] [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: 02/06/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Plant-made vaccines and trained immunity-based vaccines (TIbV or TRAIMbV) represent two strategies for enhancing immunity against diseases. Plants provide an effective and cost-efficient vaccine production platform, while TIbV induces innate immune memory that can protect against both homologous and heterologous diseases. Both strategies are generally compatible; however, they have not been explored in a transdisciplinary manner. Despite their strengths in vaccinology, each faces limitations that hinder widespread adoption and health benefits. This review revisits both strategies, discussing their fundamental knowledge alongside practical and experimental examples, ultimately highlighting their limitations and perspectives to pave the way for a unified approach to combat diseases. Future scenarios are envisioned and presented if research on plant-made trained immunity-based vaccines is adopted.
Collapse
Affiliation(s)
- Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico; SECIHTI-Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Miriam Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Abel Ramos-Vega
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada (CICATA) Unidad Morelos del Instituto Politécnico Nacional (IPN), Dirección: Boulevard de la Tecnología No.1036, Código Postal 62790 Xochitepec, Morelos, Mexico
| | - Edgar Trujillo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group and Laboratorio Nacional CONAHCYT (SECIHTI) de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C. Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.
| |
Collapse
|
2
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
3
|
Göritzer K, Melnik S, Schwestka J, Arcalis E, Drapal M, Fraser P, Ma JK, Stoger E. Enhancing quality and yield of recombinant secretory IgA antibodies in Nicotiana benthamiana by endoplasmic reticulum engineering. PLANT BIOTECHNOLOGY JOURNAL 2025; 23:1178-1189. [PMID: 39822055 PMCID: PMC11933863 DOI: 10.1111/pbi.14576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
The production of complex multimeric secretory immunoglobulins (SIgA) in Nicotiana benthamiana leaves is challenging, with significant reductions in complete protein assembly and consequently yield, being the most important difficulties. Expanding the physical dimensions of the ER to mimic professional antibody-secreting cells can help to increase yields and promote protein folding and assembly. Here, we expanded the ER in N. benthamiana leaves by targeting the enzyme CTP:phosphocholine cytidylyltransferase (CCT), which catalyses the rate-limiting step in the synthesis of the key membrane component phosphatidylcholine (PC). We used CRISPR/Cas to perform site-directed mutagenesis of each of the three endogenous CCT genes in N. benthamiana by introducing frame-shifting indels to remove the auto-inhibitory C-terminal domains. We generated stable homozygous lines of N. benthamiana containing different combinations of the edited genes, including plants where all three isofunctional CCT homologues were modified. Changes in ER morphology in the mutant plants were confirmed by in vivo confocal imaging and substantially increased the yields of two fully assembled SIgAs by prolonging the ER residence time and boosting chaperone accumulation. Through a combination of ER engineering with chaperone overexpression, we increased the yields of fully assembled SIgA by an order of magnitude, reaching almost 1 g/kg fresh leaf weight. This strategy removes a major roadblock to producing SIgA and will likely facilitate the production of other complex multimeric biopharmaceutical proteins in plants.
Collapse
Affiliation(s)
- Kathrin Göritzer
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Stanislav Melnik
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Jennifer Schwestka
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Elsa Arcalis
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | | | - Julian K‐C. Ma
- Institute for Infection and ImmunitySt George's University of LondonLondonUK
| | - Eva Stoger
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
4
|
Jha K, Jaishwal P, Yadav TP, Singh SP. Self-assembling of coiled-coil peptides into virus-like particles: Basic principles, properties, design, and applications with special focus on vaccine design and delivery. Biophys Chem 2025; 318:107375. [PMID: 39674128 DOI: 10.1016/j.bpc.2024.107375] [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: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Self-assembling peptide nanoparticles (SAPN) based delivery systems, including virus-like particles (VLP), have shown great potential for becoming prominent in next-generation vaccine and drug development. The VLP can mimic properties of natural viral capsid in terms of size (20-200 nm), geometry (i.e., icosahedral structures), and the ability to generate a robust immune response (with multivalent epitopes) through activation of innate and/or adaptive immune signals. In this regard, coiled-coil (CC) domains are suitable building blocks for designing VLP because of their programmable interaction specificity, affinity, and well-established sequence-to-structure relationships. Generally, two CC domains with different oligomeric states (trimer and pentamer) are fused to form a monomeric protein through a short, flexible spacer sequence. By using combinations of symmetry axes (2-, 3- and 5- folds) that are unique to the geometry of the desired protein cage, it is possible, in principle, to assemble well-defined protein cages like VLP. In this review, we have discussed the crystallographic rules and the basic principles involved in the design of CC-based VLP. It also explored the functions of numerous noncovalent interactions in generating stable VLP structures, which play a crucial role in improving the properties of vaccine immunogenicity, drug delivery, and 3D cell culturing.
Collapse
Affiliation(s)
- Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari 845401, India
| | - Thakur Prasad Yadav
- Department of Physics, Faculty of Science, University of Allahabad, Prayagraj 211002, India.
| | | |
Collapse
|
5
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
6
|
Khudainazarova NS, Granovskiy DL, Kondakova OA, Ryabchevskaya EM, Kovalenko AO, Evtushenko EA, Arkhipenko MV, Nikitin NA, Karpova OV. Prokaryote- and Eukaryote-Based Expression Systems: Advances in Post-Pandemic Viral Antigen Production for Vaccines. Int J Mol Sci 2024; 25:11979. [PMID: 39596049 PMCID: PMC11594041 DOI: 10.3390/ijms252211979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
This review addresses the ongoing global challenge posed by emerging and evolving viral diseases, underscoring the need for innovative vaccine development strategies. It focuses on the modern approaches to creating vaccines based on recombinant proteins produced in different expression systems, including bacteria, yeast, plants, insects, and mammals. This review analyses the advantages, limitations, and applications of these expression systems for producing vaccine antigens, as well as strategies for designing safer, more effective, and potentially 'universal' antigens. The review discusses the development of vaccines for a range of viral diseases, excluding SARS-CoV-2, which has already been extensively studied. The authors present these findings with the aim of contributing to ongoing research and advancing the development of antiviral vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nikolai A. Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.S.K.); (D.L.G.); (O.A.K.); (E.M.R.); (A.O.K.); (E.A.E.); (M.V.A.); (O.V.K.)
| | | |
Collapse
|
7
|
Bastami M, Hosseini R. The codon optimised gene produces an active human basic fibroblastic growth factor in rice cell suspension culture. Growth Factors 2024; 42:171-187. [PMID: 39485262 DOI: 10.1080/08977194.2024.2423747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
The coding sequence of human basic fibroblast growth factor (hbFGF) was optimised for expression in rice. An expression cassette was constructed by fusing the PCR-amplified RAmy3D promoter, along with its 5'UTR, 3'UTR, and terminator sequences, to the codon-optimised hbFGF sequence. This cassette was inserted into the pCAMBIA1304 shuttle vector, which also contained the RAmy3D signal peptide. Agrobacterium tumefaciens strain LBA 4404 was used to transform rice callus. Among the transformed lines, the callus expressing the highest level of bFGF (38.1 mg/kg fresh weight) was identified via ELISA and selected for establishing a cell suspension culture. Expression and secretion of the recombinant bFGF into the culture medium were observed three days after incubating the transgenic rice cells in sucrose-free medium. The presence of recombinant bFGF was confirmed through Western blot and SDS-PAGE analyses. Furthermore, the rice-derived bFGF effectively stimulated the proliferation of NIH/3T3 cells, demonstrating a comparable biological activity to that of commercial bFGF.
Collapse
Affiliation(s)
- Meysam Bastami
- Department of Biotechnology, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran
| | - Ramin Hosseini
- Department of Biotechnology, Faculty of Agriculture and Natural Resources, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
8
|
Bharathi JK, Suresh P, Prakash MAS, Muneer S. Exploring recent progress of molecular farming for therapeutic and recombinant molecules in plant systems. Heliyon 2024; 10:e37634. [PMID: 39309966 PMCID: PMC11416299 DOI: 10.1016/j.heliyon.2024.e37634] [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: 04/16/2024] [Revised: 08/10/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
An excellent technique for producing pharmaceuticals called "molecular farming" enables the industrial mass production of useful recombinant proteins in genetically modified organisms. Protein-based pharmaceuticals are rising in significance because of a variety of factors, including their bioreactivity, precision, safety, and efficacy rate. Heterologous expression methods for the manufacturing of pharmaceutical products have been previously employed using yeast, bacteria, and animal cells. However, the high cost of mammalian cell system, and production, the chance for product complexity, and contamination, and the hurdles of scaling up to commercial production are the limitations of these traditional expression methods. Plants have been raised as a hopeful replacement system for the expression of biopharmaceutical products due to their potential benefits, which include low production costs, simplicity in scaling up to commercial manufacturing levels, and a lower threat of mammalian toxin contaminations and virus infections. Since plants are widely utilized as a source of therapeutic chemicals, molecular farming offers a unique way to produce molecular medicines such as recombinant antibodies, enzymes, growth factors, plasma proteins, and vaccines whose molecular basis for use in therapy is well established. Biopharming provides more economical and extensive pharmaceutical drug supplies, including vaccines for contagious diseases and pharmaceutical proteins for the treatment of conditions like heart disease and cancer. To assess its technical viability and the efficacy resulting from the adoption of molecular farming products, the following review explores the various methods and methodologies that are currently employed to create commercially valuable molecules in plant systems.
Collapse
Affiliation(s)
- Jothi Kanmani Bharathi
- Department of Genetics and Plant Breeding, Faculty of Agriculture, Annamalai University, Annamalai Nagar, 608002, Tamil Nadu, India
| | - Preethika Suresh
- School of Bioscience and Biotechnology, Vellore Institute of Technology, Vellore, Tamil-Nadu, India
- Department of Horticulture and Food Science, School of Agricultural Innovations and Advanced Learning, Vellore Institute of Technology, Vellore, Tamil-Nadu, India
| | - Muthu Arjuna Samy Prakash
- Department of Genetics and Plant Breeding, Faculty of Agriculture, Annamalai University, Annamalai Nagar, 608002, Tamil Nadu, India
| | - Sowbiya Muneer
- Department of Horticulture and Food Science, School of Agricultural Innovations and Advanced Learning, Vellore Institute of Technology, Vellore, Tamil-Nadu, India
| |
Collapse
|
9
|
Henríquez R, Muñoz-Barroso I. Viral vector- and virus-like particle-based vaccines against infectious diseases: A minireview. Heliyon 2024; 10:e34927. [PMID: 39144987 PMCID: PMC11320483 DOI: 10.1016/j.heliyon.2024.e34927] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
To overcome the limitations of conventional vaccines, new platforms for vaccine design have emerged such as those based on viral vectors and virus-like particles (VLPs). Viral vector vaccines are highly efficient and the onset of protection is quick. Many recombinant vaccine candidates for humans are based on viruses belonging to different families such as Adenoviridae, Retroviridae, Paramyxoviridae, Rhabdoviridae, and Parvoviridae. Also, the first viral vector vaccine licensed for human vaccination was the Japanese encephalitis virus vaccine. Since then, several viral vectors have been approved for vaccination against the viruses of Lassa fever, Ebola, hepatitis B, hepatitis E, SARS-CoV-2, and malaria. VLPs are nanoparticles that mimic viral particles formed from the self-assembly of structural proteins and VLP-based vaccines against hepatitis B and E viruses, human papillomavirus, and malaria have been commercialized. As evidenced by the accelerated production of vaccines against COVID-19, these new approaches are important tools for vaccinology and for generating rapid responses against pathogens and emerging pandemic threats.
Collapse
Affiliation(s)
- Ruth Henríquez
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| | - Isabel Muñoz-Barroso
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Edificio Departamental Lab.106. Plaza Doctores de la Reina S/n, 37007, Salamanca, Spain
| |
Collapse
|
10
|
Badruzzaman ATM, Cheng YC, Sung WC, Lee MS. Insect Cell-Based Quadrivalent Seasonal Influenza Virus-like Particles Vaccine Elicits Potent Immune Responses in Mice. Vaccines (Basel) 2024; 12:667. [PMID: 38932396 PMCID: PMC11209530 DOI: 10.3390/vaccines12060667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Influenza viruses can cause highly infectious respiratory diseases, posing noteworthy epidemic and pandemic threats. Vaccination is the most cost-effective intervention to prevent influenza and its complications. However, reliance on embryonic chicken eggs for commercial influenza vaccine production presents potential risks, including reductions in efficacy due to HA gene mutations and supply delays due to scalability challenges. Thus, alternative platforms are needed urgently to replace egg-based methods and efficiently meet the increasing demand for vaccines. In this study, we employed a baculovirus expression vector system to engineer HA, NA, and M1 genes from seasonal influenza strains A/H1N1, A/H3N2, B/Yamagata, and B/Victoria, generating virus-like particle (VLP) vaccine antigens, H1N1-VLP, H3N2-VLP, Yamagata-VLP, and Victoria-VLP. We then assessed their functional and antigenic characteristics, including hemagglutination assay, protein composition, morphology, stability, and immunogenicity. We found that recombinant VLPs displayed functional activity, resembling influenza virions in morphology and size while maintaining structural integrity. Comparative immunogenicity assessments in mice showed that our quadrivalent VLPs were consistent in inducing hemagglutination inhibition and neutralizing antibody titers against homologous viruses compared to both commercial recombinant HA and egg-based vaccines (Vaxigrip). The findings highlight insect cell-based VLP vaccines as promising candidates for quadrivalent seasonal influenza vaccines. Further studies are worth conducting.
Collapse
Affiliation(s)
- A. T. M. Badruzzaman
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan 350, Taiwan; (A.T.M.B.); (Y.-C.C.); (W.-C.S.)
- Department of Life Sciences, National Central University, Zhongli District, Taoyuan 320, Taiwan
| | - Yu-Chieh Cheng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan 350, Taiwan; (A.T.M.B.); (Y.-C.C.); (W.-C.S.)
| | - Wang-Chou Sung
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan 350, Taiwan; (A.T.M.B.); (Y.-C.C.); (W.-C.S.)
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan 350, Taiwan; (A.T.M.B.); (Y.-C.C.); (W.-C.S.)
| |
Collapse
|
11
|
Rutkowska DA, Du Plessis LH, Suleman E, O’Kennedy MM, Thimiri Govinda Raj DB, Lemmer Y. Development of a Plant-Expressed Subunit Vaccine against Brucellosis. Microorganisms 2024; 12:1047. [PMID: 38930429 PMCID: PMC11205566 DOI: 10.3390/microorganisms12061047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Brucellosis is an important bacterial disease of livestock and the most common zoonotic disease. The current vaccines are effective but unsafe, as they result in animal abortions and are pathogenic to humans. Virus-like particles are being investigated as molecular scaffolds for foreign antigen presentation to the immune system. Here, we sought to develop a new-generation vaccine by presenting selected Brucella melitensis T cell epitopes on the surface of Orbivirus core-like particles (CLPs) and transiently expressing these chimeric particles in Nicotiana benthamiana plants. We successfully demonstrated the assembly of five chimeric CLPs in N. benthamiana plants, with each CLP presenting a different T cell epitope. The safety and protective efficacy of three of the highest-yielding CLPs was investigated in a mouse model of brucellosis. All three plant-expressed chimeric CLPs were safe when inoculated into BALB/c mice at specific antigen doses. However, only one chimeric CLP induced protection against the virulent Brucella strain challenge equivalent to the protection induced by the commercial Rev1 vaccine. Here, we have successfully shown the assembly, safety and protective efficacy of plant-expressed chimeric CLPs presenting B. melitensis T cell epitopes. This is the first step in the development of a safe and efficacious subunit vaccine against brucellosis.
Collapse
Affiliation(s)
- Daria A. Rutkowska
- Advanced Agriculture and Food Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa;
| | - Lissinda H. Du Plessis
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom 2520, South Africa;
| | - Essa Suleman
- Advanced Agriculture and Food Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa;
| | - Martha M. O’Kennedy
- Future Production and Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (M.M.O.); (Y.L.)
| | - Deepak B. Thimiri Govinda Raj
- Synthetic Biology and Precision Medicine Centre, Future Production and Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa;
| | - Yolandy Lemmer
- Future Production and Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (M.M.O.); (Y.L.)
| |
Collapse
|
12
|
Elbohy OA, Iqbal M, Daly JM, Dunham SP. Development of Virus-like Particle Plant-Based Vaccines against Avian H5 and H9 Influenza A Viruses. Vet Sci 2024; 11:93. [PMID: 38393111 PMCID: PMC10891754 DOI: 10.3390/vetsci11020093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Avian influenza A virus (AIV) is a significant cause of mortality in poultry, causing substantial economic loss, particularly in developing countries, and has zoonotic potential. For example, highly pathogenic avian influenza (HPAI) viruses of the H5 subtype have been circulating in Egypt for around two decades. In the last decade, H5N1 viruses of clade 2.2.1 have been succeeded by the antigenically distinct H5N8 clade 2.3.4.4b viruses. Furthermore, H9N2 viruses co-circulate with the H5N8 viruses in Egyptian poultry. It is widely recognised that effective vaccination against IAV requires a close antigenic match between the vaccine and viruses circulating in the field. Therefore, approaches to develop cost-effective vaccines that can be rapidly adapted to local virus strains are required for developing countries such as Egypt. In this project, the haemagglutinin (HA) proteins of Egyptian H5 and H9 viruses were expressed by transient transfection of plants (Nicotiana benthamiana). The formation of virus-like particles (VLPs) was confirmed by transmission electron microscopy. Mice were immunised with four doses of either H5 or H9 VLPs with adjuvant. Antibody and cellular immune responses were measured against the corresponding recombinant protein using ELISA and enzyme-linked immunosorbent assay (ELISpot), respectively. Chickens were immunised with one dose of H5 VLPs, eliciting HA-specific antibodies measured by ELISA and a pseudotyped virus neutralisation test using a heterologous H5 HA. In conclusion, plant-based VLP vaccines have potential for producing an effective vaccine candidate within a short time at a relatively low cost.
Collapse
Affiliation(s)
- Ola A Elbohy
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Munir Iqbal
- Avian Influenza Group and Newcastle Disease, The Pirbright Institute, Woking GU24 0NF, UK
| | - Janet M Daly
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| | - Stephen P Dunham
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
| |
Collapse
|
13
|
Leong SL, Gras S, Grant EJ. Fighting flu: novel CD8 + T-cell targets are required for future influenza vaccines. Clin Transl Immunology 2024; 13:e1491. [PMID: 38362528 PMCID: PMC10867544 DOI: 10.1002/cti2.1491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Seasonal influenza viruses continue to cause severe medical and financial complications annually. Although there are many licenced influenza vaccines, there are billions of cases of influenza infection every year, resulting in the death of over half a million individuals. Furthermore, these figures can rise in the event of a pandemic, as seen throughout history, like the 1918 Spanish influenza pandemic (50 million deaths) and the 1968 Hong Kong influenza pandemic (~4 million deaths). In this review, we have summarised many of the currently licenced influenza vaccines available across the world and current vaccines in clinical trials. We then briefly discuss the important role of CD8+ T cells during influenza infection and why future influenza vaccines should consider targeting CD8+ T cells. Finally, we assess the current landscape of known immunogenic CD8+ T-cell epitopes and highlight the knowledge gaps required to be filled for the design of rational future influenza vaccines that incorporate CD8+ T cells.
Collapse
Affiliation(s)
- Samuel Liwei Leong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
| | - Stephanie Gras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| | - Emma J Grant
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular ScienceLa Trobe UniversityBundooraVICAustralia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery InstituteMonash UniversityClaytonVICAustralia
| |
Collapse
|
14
|
Mugunthan SP, Venkatesan D, Govindasamy C, Selvaraj D, Mani Chandra H. A preliminary study of the immunogenic response of plant-derived multi-epitopic peptide vaccine candidate of Mycoplasma gallisepticum in chickens. FRONTIERS IN PLANT SCIENCE 2024; 14:1298880. [PMID: 38322423 PMCID: PMC10846684 DOI: 10.3389/fpls.2023.1298880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Mycoplasma gallisepticum (MG) is responsible for chronic respiratory disease in avian species, characterized by symptoms like respiratory rales and coughing. Existing vaccines for MG have limited efficacy and require multiple doses. Certain MG cytoadherence proteins (GapA, CrmA, PlpA, and Hlp3) play a crucial role in the pathogen's respiratory tract colonization and infection. Plant-based proteins and therapeutics have gained attention due to their safety and efficiency. In this study, we designed a 21.4-kDa multi-epitope peptide vaccine (MEPV) using immunogenic segments from cytoadherence proteins. The MEPV's effectiveness was verified through computational simulations. We then cloned the MEPV, introduced it into the plant expression vector pSiM24-eGFP, and expressed it in Nicotiana benthamiana leaves. The plant-produced MEPV proved to be immunogenic when administered intramuscularly to chickens. It significantly boosted the production of immunoglobulin Y (IgY)-neutralizing antibodies against cytoadherence protein epitopes in immunized chickens compared to that in the control group. This preliminary investigation demonstrates that the plant-derived MEPV is effective in triggering an immune response in chickens. To establish an efficient poultry health management system and ensure the sustainability of the poultry industry, further research is needed to develop avian vaccines using plant biotechnology.
Collapse
Affiliation(s)
| | | | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Dhivya Selvaraj
- Artificial Intelligence Laboratory, School of Computer Information and Communication Engineering, Kunsan National University, Gunsan, Republic of Korea
| | - Harish Mani Chandra
- Department of Biotechnology, Thiruvalluvar University, Vellore, Tamil Nadu, India
| |
Collapse
|
15
|
Lim S, Kwon HJ, Jeong DG, Nie H, Lee S, Ko SR, Lee KS, Ryu YB, Mason HS, Kim HS, Shin AY, Kwon SY. Enhanced binding and inhibition of SARS-CoV-2 by a plant-derived ACE2 protein containing a fused mu tailpiece. Biotechnol J 2024; 19:e2300319. [PMID: 37853601 DOI: 10.1002/biot.202300319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/20/2023]
Abstract
Infectious diseases such as Coronavirus disease 2019 (COVID-19) and Middle East respiratory syndrome (MERS) present an increasingly persistent crisis in many parts of the world. COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The angiotensin-converting enzyme 2 (ACE2) is a crucial cellular receptor for SARS-CoV-2 infection. Inhibition of the interaction between SARS-CoV-2 and ACE2 has been proposed as a target for the prevention and treatment of COVID-19. We produced four recombinant plant-derived ACE2 isoforms with or without the mu tailpiece (μ-tp) of immunoglobulin M (IgM) and the KDEL endoplasmic reticulum retention motif in a plant expression system. The plant-derived ACE2 isoforms bound whole SARS-CoV-2 virus and the isolated receptor binding domains of SARS-CoV-2 Alpha, Beta, Gamma, Delta, and Omicron variants. Fusion of μ-tp and KDEL to the ACE2 protein (ACE2 μK) had enhanced binding activity with SARS-CoV-2 in comparison with unmodified ACE2 protein derived from CHO cells. Furthermore, the plant-derived ACE2 μK protein exhibited no cytotoxic effects on Vero E6 cells and effectively inhibited SARS-CoV-2 infection. The efficient and rapid scalability of plant-derived ACE2 μK protein offers potential for the development of preventive and therapeutic agents in the early response to future viral outbreaks.
Collapse
Affiliation(s)
- Sohee Lim
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hyung-Jun Kwon
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Bio-Analytical Science Division, Korea Research Institute of Bioscience and Biotechnology School of Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Hualin Nie
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sanghee Lee
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Biosystems and Bioengineering Program, Korea Research Institute of Bioscience and Biotechnology School of Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Seo-Rin Ko
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Bioinformatics, Korea Research Institute of Bioscience and Biotechnology School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea
| | - Kyu-Sun Lee
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Bio-Analytical Science Division, Korea Research Institute of Bioscience and Biotechnology School of Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Young Bae Ryu
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Hugh S Mason
- Center for Immunotherapy, Vaccines, and Virotherapy (CIVV), The Biodesign Institute at ASU, Tempe, Arizona, USA
| | - Hyun-Soon Kim
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Biosystems and Bioengineering Program, Korea Research Institute of Bioscience and Biotechnology School of Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| | - Ah-Young Shin
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Bioinformatics, Korea Research Institute of Bioscience and Biotechnology School of Bioscience, University of Science and Technology, Daejeon, Republic of Korea
| | - Suk-Yoon Kwon
- Plant Systems Engineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Biosystems and Bioengineering Program, Korea Research Institute of Bioscience and Biotechnology School of Biotechnology, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
16
|
Gashti AB, Agbayani G, Hrapovic S, Nassoury N, Coulombe N, Dudani R, Harrison BA, Akache B, Gilbert R, Chahal PS. Production, purification and immunogenicity of Gag virus-like particles carrying SARS-CoV-2 components. Vaccine 2024; 42:40-52. [PMID: 38042697 DOI: 10.1016/j.vaccine.2023.11.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/28/2023] [Accepted: 11/23/2023] [Indexed: 12/04/2023]
Abstract
The virus-like particle (VLP) platform is a robust inducer of humoral and cellular immune responses; hence, it has been used in vaccine development for several infectious diseases. In the current work, VLPs carrying SARS-CoV-2 Spike (S) protein (Wuhan strain) with an HIV-1 Gag core were produced using suspension HEK 293SF-3F6 cells by transient transfection. The Gag was fused with green fluorescent protein (GFP) for rapid quantification of the VLPs. Five different versions of Gag-Spike VLPs (Gag-S-VLPs) consisting of Gag-S alone or combined with other SARS-CoV-2 components, namely Gag-S-Nucleocapsid (N), Gag-S-Matrix (M), Gag-S-Envelope (E), Gag-S-MEN, along with Gag alone were produced and processed by clarification, nuclease treatment, concentration by tangential flow filtration (TFF) and diafiltration. A pilot mouse study was performed to evaluate the immunogenicity of the Gag-S-VLPs through the measurement of the humoral and/or cellular responses against all the mentioned SARS-CoV-2 components. Antibody response to Spike was observed in all variants. The highest number of Spike-specific IFN-γ + T cells was detected with Gag-S-VLPs. No induction of antigen-specific cellular responses to M, N or E proteins were detected with any of the Gag-S, M, E/or N VLPs tested. Therefore, the Gag-S-VLP, by reason of consistently eliciting strong antigen-specific cellular and antibody responses, was selected for further evaluation. The purification process was improved by replacing the conventional centrifugation by serial microfiltration in the clarification step, followed by Spike-affinity chromatography to get concentrated VLPs with higher purity. Three different doses of Gag-S-VLP in conjunction with two adjuvants (Quil-A or AddaVax) were used to assess the dose-dependent antigen-specific cellular and antibody responses in mice. The Gag-S-VLP adjuvanted with Quil-A resulted in a stronger Spike-specific cellular response compared to that adjuvanted with AddaVax. A strong spike neutralisation activity was observed for all doses, independent of the adjuvant combination.
Collapse
Affiliation(s)
| | - Gerard Agbayani
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Sabahudin Hrapovic
- Aquatic and Crop Resource Development Research Centre, National Research Council Canada, Montréal, Canada
| | - Nasha Nassoury
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Nathalie Coulombe
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada
| | - Renu Dudani
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Blair A Harrison
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Bassel Akache
- Human Health Therapeutics, National Research Council Canada Ottawa, ON, Canada
| | - Rénald Gilbert
- Human Health Therapeutics, National Research Council Canada, Montreal, QC, Canada; Department of Bioengineering, McGill University, Montreal, QC, Canada.
| | | |
Collapse
|
17
|
Zahmanova G, Aljabali AAA, Takova K, Minkov G, Tambuwala MM, Minkov I, Lomonossoff GP. Green Biologics: Harnessing the Power of Plants to Produce Pharmaceuticals. Int J Mol Sci 2023; 24:17575. [PMID: 38139405 PMCID: PMC10743837 DOI: 10.3390/ijms242417575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Plants are increasingly used for the production of high-quality biological molecules for use as pharmaceuticals and biomaterials in industry. Plants have proved that they can produce life-saving therapeutic proteins (Elelyso™-Gaucher's disease treatment, ZMapp™-anti-Ebola monoclonal antibodies, seasonal flu vaccine, Covifenz™-SARS-CoV-2 virus-like particle vaccine); however, some of these therapeutic proteins are difficult to bring to market, which leads to serious difficulties for the manufacturing companies. The closure of one of the leading companies in the sector (the Canadian biotech company Medicago Inc., producer of Covifenz) as a result of the withdrawal of investments from the parent company has led to the serious question: What is hindering the exploitation of plant-made biologics to improve health outcomes? Exploring the vast potential of plants as biological factories, this review provides an updated perspective on plant-derived biologics (PDB). A key focus is placed on the advancements in plant-based expression systems and highlighting cutting-edge technologies that streamline the production of complex protein-based biologics. The versatility of plant-derived biologics across diverse fields, such as human and animal health, industry, and agriculture, is emphasized. This review also meticulously examines regulatory considerations specific to plant-derived biologics, shedding light on the disparities faced compared to biologics produced in other systems.
Collapse
Affiliation(s)
- Gergana Zahmanova
- Department of Plant Physiology and Molecular Biology, University of Plovdiv, 4000 Plovdiv, Bulgaria; (K.T.)
- Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Katerina Takova
- Department of Plant Physiology and Molecular Biology, University of Plovdiv, 4000 Plovdiv, Bulgaria; (K.T.)
| | - George Minkov
- Department of Plant Physiology and Molecular Biology, University of Plovdiv, 4000 Plovdiv, Bulgaria; (K.T.)
| | - Murtaza M. Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK;
| | - Ivan Minkov
- Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
- Institute of Molecular Biology and Biotechnologies, 4108 Markovo, Bulgaria
| | | |
Collapse
|
18
|
Jung JW, Park PG, Lee WK, Shin JH, Jang MH, Seo EH, An T, Kim YB, Moon MH, Choi SK, Yun JS, Hong KJ, Kim SR. Production of Plant-Derived Japanese Encephalitis Virus Multi-Epitope Peptide in Nicotiana benthamiana and Immunological Response in Mice. Int J Mol Sci 2023; 24:11643. [PMID: 37511402 PMCID: PMC10380836 DOI: 10.3390/ijms241411643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/04/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The current production of the Japanese encephalitis virus (JEV) vaccine is based on animal cells, where various risk factors for human health should be resolved. This study used a transient expression system to express the chimeric protein composed of antigenic epitopes from the JEV envelope (E) protein in Nicotiana benthamiana. JEV multi-epitope peptide (MEP) sequences fused with FLAG-tag or 6× His-tag at the C- or N-terminus for the purification were introduced into plant expression vectors and used for transient expression. Among the constructs, vector pSK480, which expresses MEP fused with a FLAG-tag at the C-terminus, showed the highest level of expression and yield in purification. Optimization of transient expression procedures further improved the target protein yield. The purified MEP protein was applied to an ICR mouse and successfully induced an antibody against JEV, which demonstrates the potential of the plant-produced JEV MEP as an alternative vaccine candidate.
Collapse
Affiliation(s)
- Jae-Wan Jung
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea
- PhytoMab Co., Seoul 04107, Republic of Korea
| | - Pil-Gu Park
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| | - Won-Kyung Lee
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea
| | - Jun-Hye Shin
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea
- PhytoMab Co., Seoul 04107, Republic of Korea
| | - Mi-Hwa Jang
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea
- PhytoMab Co., Seoul 04107, Republic of Korea
| | - Eun-Hye Seo
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
- BK21 Plus, Department of Cellular and Molecular Medicine, Konkuk University School of Medicine, Seoul 05029, Republic of Korea
| | - Timothy An
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| | - Young Beom Kim
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Myeong Hee Moon
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | | | - Jee Sun Yun
- Eubiologics Co., Seoul 06026, Republic of Korea
| | - Kee-Jong Hong
- Department of Microbiology, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| | - Seong-Ryong Kim
- Department of Life Science, Sogang University, Seoul 04107, Republic of Korea
- PhytoMab Co., Seoul 04107, Republic of Korea
| |
Collapse
|
19
|
Knödler M, Opdensteinen P, Sankaranarayanan RA, Morgenroth A, Buhl EM, Mottaghy FM, Buyel JF. Simple plant-based production and purification of the assembled human ferritin heavy chain as a nanocarrier for tumor-targeted drug delivery and bioimaging in cancer therapy. Biotechnol Bioeng 2023; 120:1038-1054. [PMID: 36539373 DOI: 10.1002/bit.28312] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/06/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Nanoparticles are used as carriers for the delivery of drugs and imaging agents. Proteins are safer than synthetic nanocarriers due to their greater biocompatibility and the absence of toxic degradation products. In this context, ferritin has the additional benefit of inherently targeting the membrane receptor transferrin 1, which is overexpressed by most cancer cells. Furthermore, this self-assembling multimeric protein can be loaded with more than 2000 iron atoms, as well as drugs, contrast agents, and other cargos. However, recombinant ferritin currently costs ~3.5 million € g-1 , presumably because the limited number of producers cannot meet demand, making it generally unaffordable as a nanocarrier. Because plants can produce proteins at very-large-scale, we developed a simple, proof-of-concept process for the production of the human ferritin heavy chain by transient expression in Nicotiana benthamiana. We optimized the protein yields by screening different compartments and 5'-untranslated regions in PCPs, and selected the best-performing construct for production in differentiated plants. We then established a rapid and scalable purification protocol by combining pH and heat treatment before extraction, followed by an ultrafiltration/diafiltration size-based separation process. The optimized process achieved ferritin levels of ~40 mg kg-1 fresh biomass although depth filtration limited product recovery to ~7%. The purity of the recombinant product was >90% at costs ~3% of the current sales price. Our method therefore allows the production of affordable ferritin heavy chain as a carrier for therapeutic and diagnostic agents, which is suitable for further stability and functionality testing in vitro and in vivo.
Collapse
Affiliation(s)
- Matthias Knödler
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e. V., Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | - Patrick Opdensteinen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e. V., Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | | | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute for Pathology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Johannes Felix Buyel
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
- Department of Biotechnology (DBT), Institute of Bioprocess Science and Engineering (IBSE), University of Natural Resources and Life Sciences, Vienna (BOKU), Vienna, Austria
| |
Collapse
|
20
|
Wang X, Prokhnevsky AI, Skarjinskaia M, Razzak MA, Streatfield SJ, Lee J. Facilitating viral vector movement enhances heterologous protein production in an established plant system. PLANT BIOTECHNOLOGY JOURNAL 2023; 21:635-645. [PMID: 36511837 PMCID: PMC9946140 DOI: 10.1111/pbi.13977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/23/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
Molecular farming technology using transiently transformed Nicotiana plants offers an economical approach to the pharmaceutical industry to produce an array of protein targets including vaccine antigens and therapeutics. It can serve as a desirable alternative approach for those proteins that are challenging or too costly to produce in large quantities using other heterologous protein expression systems. However, since cost metrics are such a critical factor in selecting a production host, any system-wide modifications that can increase recombinant protein yields are key to further improving the platform and making it applicable for a wider range of target molecules. Here, we report on the development of a new approach to improve target accumulation in an established plant-based expression system that utilizes viral-based vectors to mediate transient expression in Nicotiana benthamiana. We show that by engineering the host plant to support viral vectors to spread more effectively between host cells through plasmodesmata, protein target accumulation can be increased by up to approximately 60%.
Collapse
Affiliation(s)
- Xu Wang
- Department of Plant and Soil Sciences, College of Agriculture and Natural ResourcesUniversity of DelawareDelawareNewarkUSA
- Present address:
Department of Plant Physiology and BiochemistryUniversity of HohenheimBaden‐WürttembergStuttgartGermany
| | | | - Marina Skarjinskaia
- Fraunhofer USA Inc.Center Mid‐Atlantic, Biotechnology DivisionDelawareNewarkUSA
| | - Md Abdur Razzak
- Department of Plant and Soil Sciences, College of Agriculture and Natural ResourcesUniversity of DelawareDelawareNewarkUSA
| | | | - Jung‐Youn Lee
- Department of Plant and Soil Sciences, College of Agriculture and Natural ResourcesUniversity of DelawareDelawareNewarkUSA
- Department of Biological Sciences, College of Arts and SciencesUniversity of DelawareDelawareNewarkUSA
- Delaware Biotechnology InstituteUniversity of DelawareDelawareNewarkUSA
| |
Collapse
|
21
|
Grandits M, Grünwald-Gruber C, Gastine S, Standing JF, Reljic R, Teh AYH, Ma JKC. Improving the efficacy of plant-made anti-HIV monoclonal antibodies for clinical use. FRONTIERS IN PLANT SCIENCE 2023; 14:1126470. [PMID: 36923134 PMCID: PMC10009187 DOI: 10.3389/fpls.2023.1126470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
Introduction Broadly neutralising antibodies are promising candidates for preventing and treating Human Immunodeficiency Virus/Acquired Immunodeficiency Syndrome (HIV/AIDS), as an alternative to or in combination with antiretroviral therapy (ART). These mAbs bind to sites on the virus essential for virus attachment and entry, thereby inhibiting entry into the host cell. However, the cost and availability of monoclonal antibodies, especially combinations of antibodies, hampers implementation of anti-HIV bNAb therapies in low- to middle- income countries (LMICs) where HIV-1 prevalence is highest. Methods We have produced three HIV broadly neutralizing antibodies (bNAbs), 10-1074, VRC01 and 3BNC117 in the Nicotiana benthamiana transient expression system. The impact of specific modifications to enhance potency and efficacy were assessed. To prolong half-life and increase bioavailability, a M252Y/S254T/T256E (YTE) or M428L/N434S (LS) mutation was introduced. To increase antibody dependent cellular cytotoxicity (ADCC), we expressed an afucosylated version of each antibody using a glycoengineered plant line. Results The majority of bNAbs and their variants could be expressed at yields of up to 47 mg/kg. Neither the expression system nor the modifications impacted the neutralization potential of the bNAbs. Afucosylated bNAbs exhibit enhanced ability to bind to FcγRIIIa and trigger ADCC, regardless of the presence of Fc amino acid mutations. Lastly, we demonstrated that Fc-modified variants expressed in plants show enhanced binding to FcRn, which results in a favourable in vivo pharmacokinetic profile compared to their unmodified counterparts. Conclusion Tobacco plants are suitable expression hosts for anti-HIV bNAbs with increased efficacy and an improved pharmacokinetic profile.
Collapse
Affiliation(s)
- Melanie Grandits
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Clemens Grünwald-Gruber
- Core Facility Mass Spectrometry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Silke Gastine
- Infection, Immunity and Inflammation Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Joseph F. Standing
- Infection, Immunity and Inflammation Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Rajko Reljic
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Audrey Y-H. Teh
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| | - Julian K-C. Ma
- Molecular Immunology Unit, Institute for Infection and Immunity, St. George's University of London, London, United Kingdom
| |
Collapse
|
22
|
Bravo-Vázquez LA, Mora-Hernández EO, Rodríguez AL, Sahare P, Bandyopadhyay A, Duttaroy AK, Paul S. Current Advances of Plant-Based Vaccines for Neurodegenerative Diseases. Pharmaceutics 2023; 15:711. [PMID: 36840033 PMCID: PMC9963606 DOI: 10.3390/pharmaceutics15020711] [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: 01/11/2023] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by the progressive degeneration and/or loss of neurons belonging to the central nervous system, and represent one of the major global health issues. Therefore, a number of immunotherapeutic approaches targeting the non-functional or toxic proteins that induce neurodegeneration in NDDs have been designed in the last decades. In this context, due to unprecedented advances in genetic engineering techniques and molecular farming technology, pioneering plant-based immunogenic antigen expression systems have been developed aiming to offer reliable alternatives to deal with important NDDs, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Diverse reports have evidenced that plant-made vaccines trigger significant immune responses in model animals, supported by the production of antibodies against the aberrant proteins expressed in the aforementioned NDDs. Moreover, these immunogenic tools have various advantages that make them a viable alternative for preventing and treating NDDs, such as high scalability, no risk of contamination with human pathogens, cold chain free production, and lower production costs. Hence, this article presents an overview of the current progress on plant-manufactured vaccines for NDDs and discusses its future prospects.
Collapse
Affiliation(s)
- Luis Alberto Bravo-Vázquez
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| | - Erick Octavio Mora-Hernández
- School of Engineering and Sciences, Campus Mexico City, Tecnologico de Monterrey, Calle del Puente, No. 222 Col. Ejidos de Huipulco, Tlalpan, Mexico City 14380, Mexico
| | - Alma L. Rodríguez
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| | - Padmavati Sahare
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM 3001, Juriquilla, Querétaro 76230, Mexico
| | - Anindya Bandyopadhyay
- International Rice Research Institute, Manila 4031, Philippines
- Reliance Industries Ltd., Navi Mumbai 400701, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317 Oslo, Norway
| | - Sujay Paul
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| |
Collapse
|
23
|
Gupta R, Arora K, Roy SS, Joseph A, Rastogi R, Arora NM, Kundu PK. Platforms, advances, and technical challenges in virus-like particles-based vaccines. Front Immunol 2023; 14:1123805. [PMID: 36845125 PMCID: PMC9947793 DOI: 10.3389/fimmu.2023.1123805] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Viral infectious diseases threaten human health and global stability. Several vaccine platforms, such as DNA, mRNA, recombinant viral vectors, and virus-like particle-based vaccines have been developed to counter these viral infectious diseases. Virus-like particles (VLP) are considered real, present, licensed and successful vaccines against prevalent and emergent diseases due to their non-infectious nature, structural similarity with viruses, and high immunogenicity. However, only a few VLP-based vaccines have been commercialized, and the others are either in the clinical or preclinical phases. Notably, despite success in the preclinical phase, many vaccines are still struggling with small-scale fundamental research owing to technical difficulties. Successful production of VLP-based vaccines on a commercial scale requires a suitable platform and culture mode for large-scale production, optimization of transduction-related parameters, upstream and downstream processing, and monitoring of product quality at each step. In this review article, we focus on the advantages and disadvantages of various VLP-producing platforms, recent advances and technical challenges in VLP production, and the current status of VLP-based vaccine candidates at commercial, preclinical, and clinical levels.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Prabuddha K. Kundu
- Department of Research and Development, Premas Biotech Pvt Ltd., Sector IV, Industrial Model Township (IMT), Manesar, Gurgaon, India
| |
Collapse
|
24
|
Hu L, Lao G, Liu R, Feng J, Long F, Peng T. The race toward a universal influenza vaccine: Front runners and the future directions. Antiviral Res 2023; 210:105505. [PMID: 36574905 DOI: 10.1016/j.antiviral.2022.105505] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Influenza virus is the pathogen of influenza (flu) and millions of people suffer from the infection worldwide, posing a significant health risk. The current influenza vaccines induce neutralizing antibodies against hemagglutinin (HA) to achieve strain-specific neutralization. The effectiveness of seasonal vaccines is usually low and unpredictable because of the antigenic variation and genetic plasticity of viruses, as well as the interference of preexisting immunity. A universal influenza vaccine is urgently needed to prevent a wide variety of influenza viruses. Nevertheless, reaching this difficult optimal goal requires a step-by-step approach. Innovative strategies and vaccine platforms are being developed in order to generate robust cross-protective immunity. In this review, we summarize candidate influenza vaccines that meet two criteria: first, they are designed to provide protection against multiple influenza viruses; second, they had passed regulatory evaluations and have entered various stages of clinical trials. We discuss these vaccine candidates based on the different vaccine-production platforms, with the focus on antigen selection, design, adjuvants, immunomodulators, and routes of vaccine delivery in the development of universal influenza vaccines.
Collapse
Affiliation(s)
- Longbo Hu
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Geqi Lao
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Rui Liu
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jin Feng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fei Long
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong South China Vaccine, Guangzhou, China; Greater Bay Area Innovative Vaccine Technology Development Center, Guangzhou International Bio-island Laboratory, Guangzhou, China.
| |
Collapse
|
25
|
Chu KB, Quan FS. Respiratory Viruses and Virus-like Particle Vaccine Development: How Far Have We Advanced? Viruses 2023; 15:v15020392. [PMID: 36851606 PMCID: PMC9965150 DOI: 10.3390/v15020392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 01/31/2023] Open
Abstract
With technological advancements enabling globalization, the intercontinental transmission of pathogens has become much easier. Respiratory viruses are one such group of pathogens that require constant monitoring since their outbreak leads to massive public health crises, as exemplified by the influenza virus, respiratory syncytial virus (RSV), and the recent coronavirus disease 2019 (COVID-19) outbreak caused by the SARS-CoV-2. To prevent the transmission of these highly contagious viruses, developing prophylactic tools, such as vaccines, is of considerable interest to the scientific community. Virus-like particles (VLPs) are highly sought after as vaccine platforms for their safety and immunogenicity profiles. Although several VLP-based vaccines against hepatitis B and human papillomavirus have been approved for clinical use by the United States Food and Drug Administration, VLP vaccines against the three aforementioned respiratory viruses are lacking. Here, we summarize the most recent progress in pre-clinical and clinical VLP vaccine development. We also outline various strategies that contributed to improving the efficacy of vaccines against each virus and briefly discuss the stability aspect of VLPs that makes it a highly desired vaccine platform.
Collapse
Affiliation(s)
- Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence:
| |
Collapse
|
26
|
Mathew M, Thomas J. Tobacco-Based Vaccines, Hopes, and Concerns: A Systematic Review. Mol Biotechnol 2022:10.1007/s12033-022-00627-5. [PMID: 36528727 PMCID: PMC9759281 DOI: 10.1007/s12033-022-00627-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022]
Abstract
Emerging infectious diseases have vigorously devastated the global economy and health sector; cost-effective plant-based vaccines (PBV) can be the potential solution to withstand the current health economic crisis. The prominent role of tobacco as an efficient expression system for PBV has been well-established for decades, through this review we highlight the importance of tobacco-based vaccines (TBV) against evolving infectious diseases in humans. Studies focusing on the use of TBV for human infectious diseases were searched in PubMed, Google Scholar, and science direct from 1995 to 2021 using the keywords Tobacco-based vaccines OR transgenic tobacco OR Nicotiana benthamiana vaccines AND Infectious diseases or communicable diseases. We carried out a critical review of the articles and studies that fulfilled the eligibility criteria and were included in this review. Of 976 studies identified, only 63 studies fulfilling the eligibility criteria were included, which focused on either the in vitro, in vivo, or clinical studies on TBV for human infectious diseases. Around 43 in vitro studies of 23 different infectious pathogens expressed in tobacco-based systems were identified and 23 in vivo analysis studies were recognized to check the immunogenicity of vaccine candidates while only 10 of these were subjected to clinical trials. Viral infectious pathogens were studied more than bacterial pathogens. From our review, it was evident that TBV can be an effective health strategy to combat the emerging viral infectious diseases which are very difficult to manage with the current health facilities. The timely administration of cost-effective TBV can prevent the outburst of viral infections, thereby can protect the global healthcare system to a greater extent.
Collapse
Affiliation(s)
- Mintu Mathew
- Department of Pharmacology, Amrita School of Pharmacy, Kochi, Kerala India
| | - Jaya Thomas
- Department of Pharmacology, Amrita School of Pharmacy, Kochi, Kerala India
| |
Collapse
|
27
|
Graña C, Ghosn L, Evrenoglou T, Jarde A, Minozzi S, Bergman H, Buckley BS, Probyn K, Villanueva G, Henschke N, Bonnet H, Assi R, Menon S, Marti M, Devane D, Mallon P, Lelievre JD, Askie LM, Kredo T, Ferrand G, Davidson M, Riveros C, Tovey D, Meerpohl JJ, Grasselli G, Rada G, Hróbjartsson A, Ravaud P, Chaimani A, Boutron I. Efficacy and safety of COVID-19 vaccines. Cochrane Database Syst Rev 2022; 12:CD015477. [PMID: 36473651 PMCID: PMC9726273 DOI: 10.1002/14651858.cd015477] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Different forms of vaccines have been developed to prevent the SARS-CoV-2 virus and subsequent COVID-19 disease. Several are in widespread use globally. OBJECTIVES: To assess the efficacy and safety of COVID-19 vaccines (as a full primary vaccination series or a booster dose) against SARS-CoV-2. SEARCH METHODS We searched the Cochrane COVID-19 Study Register and the COVID-19 L·OVE platform (last search date 5 November 2021). We also searched the WHO International Clinical Trials Registry Platform, regulatory agency websites, and Retraction Watch. SELECTION CRITERIA We included randomized controlled trials (RCTs) comparing COVID-19 vaccines to placebo, no vaccine, other active vaccines, or other vaccine schedules. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. We used GRADE to assess the certainty of evidence for all except immunogenicity outcomes. We synthesized data for each vaccine separately and presented summary effect estimates with 95% confidence intervals (CIs). MAIN RESULTS: We included and analyzed 41 RCTs assessing 12 different vaccines, including homologous and heterologous vaccine schedules and the effect of booster doses. Thirty-two RCTs were multicentre and five were multinational. The sample sizes of RCTs were 60 to 44,325 participants. Participants were aged: 18 years or older in 36 RCTs; 12 years or older in one RCT; 12 to 17 years in two RCTs; and three to 17 years in two RCTs. Twenty-nine RCTs provided results for individuals aged over 60 years, and three RCTs included immunocompromized patients. No trials included pregnant women. Sixteen RCTs had two-month follow-up or less, 20 RCTs had two to six months, and five RCTs had greater than six to 12 months or less. Eighteen reports were based on preplanned interim analyses. Overall risk of bias was low for all outcomes in eight RCTs, while 33 had concerns for at least one outcome. We identified 343 registered RCTs with results not yet available. This abstract reports results for the critical outcomes of confirmed symptomatic COVID-19, severe and critical COVID-19, and serious adverse events only for the 10 WHO-approved vaccines. For remaining outcomes and vaccines, see main text. The evidence for mortality was generally sparse and of low or very low certainty for all WHO-approved vaccines, except AD26.COV2.S (Janssen), which probably reduces the risk of all-cause mortality (risk ratio (RR) 0.25, 95% CI 0.09 to 0.67; 1 RCT, 43,783 participants; high-certainty evidence). Confirmed symptomatic COVID-19 High-certainty evidence found that BNT162b2 (BioNtech/Fosun Pharma/Pfizer), mRNA-1273 (ModernaTx), ChAdOx1 (Oxford/AstraZeneca), Ad26.COV2.S, BBIBP-CorV (Sinopharm-Beijing), and BBV152 (Bharat Biotect) reduce the incidence of symptomatic COVID-19 compared to placebo (vaccine efficacy (VE): BNT162b2: 97.84%, 95% CI 44.25% to 99.92%; 2 RCTs, 44,077 participants; mRNA-1273: 93.20%, 95% CI 91.06% to 94.83%; 2 RCTs, 31,632 participants; ChAdOx1: 70.23%, 95% CI 62.10% to 76.62%; 2 RCTs, 43,390 participants; Ad26.COV2.S: 66.90%, 95% CI 59.10% to 73.40%; 1 RCT, 39,058 participants; BBIBP-CorV: 78.10%, 95% CI 64.80% to 86.30%; 1 RCT, 25,463 participants; BBV152: 77.80%, 95% CI 65.20% to 86.40%; 1 RCT, 16,973 participants). Moderate-certainty evidence found that NVX-CoV2373 (Novavax) probably reduces the incidence of symptomatic COVID-19 compared to placebo (VE 82.91%, 95% CI 50.49% to 94.10%; 3 RCTs, 42,175 participants). There is low-certainty evidence for CoronaVac (Sinovac) for this outcome (VE 69.81%, 95% CI 12.27% to 89.61%; 2 RCTs, 19,852 participants). Severe or critical COVID-19 High-certainty evidence found that BNT162b2, mRNA-1273, Ad26.COV2.S, and BBV152 result in a large reduction in incidence of severe or critical disease due to COVID-19 compared to placebo (VE: BNT162b2: 95.70%, 95% CI 73.90% to 99.90%; 1 RCT, 46,077 participants; mRNA-1273: 98.20%, 95% CI 92.80% to 99.60%; 1 RCT, 28,451 participants; AD26.COV2.S: 76.30%, 95% CI 57.90% to 87.50%; 1 RCT, 39,058 participants; BBV152: 93.40%, 95% CI 57.10% to 99.80%; 1 RCT, 16,976 participants). Moderate-certainty evidence found that NVX-CoV2373 probably reduces the incidence of severe or critical COVID-19 (VE 100.00%, 95% CI 86.99% to 100.00%; 1 RCT, 25,452 participants). Two trials reported high efficacy of CoronaVac for severe or critical disease with wide CIs, but these results could not be pooled. Serious adverse events (SAEs) mRNA-1273, ChAdOx1 (Oxford-AstraZeneca)/SII-ChAdOx1 (Serum Institute of India), Ad26.COV2.S, and BBV152 probably result in little or no difference in SAEs compared to placebo (RR: mRNA-1273: 0.92, 95% CI 0.78 to 1.08; 2 RCTs, 34,072 participants; ChAdOx1/SII-ChAdOx1: 0.88, 95% CI 0.72 to 1.07; 7 RCTs, 58,182 participants; Ad26.COV2.S: 0.92, 95% CI 0.69 to 1.22; 1 RCT, 43,783 participants); BBV152: 0.65, 95% CI 0.43 to 0.97; 1 RCT, 25,928 participants). In each of these, the likely absolute difference in effects was fewer than 5/1000 participants. Evidence for SAEs is uncertain for BNT162b2, CoronaVac, BBIBP-CorV, and NVX-CoV2373 compared to placebo (RR: BNT162b2: 1.30, 95% CI 0.55 to 3.07; 2 RCTs, 46,107 participants; CoronaVac: 0.97, 95% CI 0.62 to 1.51; 4 RCTs, 23,139 participants; BBIBP-CorV: 0.76, 95% CI 0.54 to 1.06; 1 RCT, 26,924 participants; NVX-CoV2373: 0.92, 95% CI 0.74 to 1.14; 4 RCTs, 38,802 participants). For the evaluation of heterologous schedules, booster doses, and efficacy against variants of concern, see main text of review. AUTHORS' CONCLUSIONS Compared to placebo, most vaccines reduce, or likely reduce, the proportion of participants with confirmed symptomatic COVID-19, and for some, there is high-certainty evidence that they reduce severe or critical disease. There is probably little or no difference between most vaccines and placebo for serious adverse events. Over 300 registered RCTs are evaluating the efficacy of COVID-19 vaccines, and this review is updated regularly on the COVID-NMA platform (covid-nma.com). Implications for practice Due to the trial exclusions, these results cannot be generalized to pregnant women, individuals with a history of SARS-CoV-2 infection, or immunocompromized people. Most trials had a short follow-up and were conducted before the emergence of variants of concern. Implications for research Future research should evaluate the long-term effect of vaccines, compare different vaccines and vaccine schedules, assess vaccine efficacy and safety in specific populations, and include outcomes such as preventing long COVID-19. Ongoing evaluation of vaccine efficacy and effectiveness against emerging variants of concern is also vital.
Collapse
Affiliation(s)
- Carolina Graña
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Lina Ghosn
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Theodoros Evrenoglou
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Alexander Jarde
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | | | | | | | | | | | - Hillary Bonnet
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Rouba Assi
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | - Melanie Marti
- Department of Immunization, Vaccines and Biologicals, World Health Organization, Geneva, Switzerland
| | - Declan Devane
- Evidence Synthesis Ireland, Cochrane Ireland and HRB-Trials Methodology Research Network, National University of Ireland, Galway, Ireland
| | - Patrick Mallon
- UCD Centre for Experimental Pathogen Host Research and UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Jean-Daniel Lelievre
- Department of Clinical Immunology and Infectious Diseases, Henri Mondor Hospital, Vaccine Research Institute, Université Paris Est Créteil, Paris, France
| | - Lisa M Askie
- Quality Assurance Norms and Standards Department, World Health Organization, Geneva, Switzerland
| | - Tamara Kredo
- Cochrane South Africa, South African Medical Research Council, Cape Town, South Africa
| | | | - Mauricia Davidson
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Carolina Riveros
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | | | - Joerg J Meerpohl
- Institute for Evidence in Medicine, Medical Center & Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
| | - Giacomo Grasselli
- Department of Anesthesia, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Gabriel Rada
- Epistemonikos Foundation, Santiago, Chile
- UC Evidence Center, Cochrane Chile Associated Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asbjørn Hróbjartsson
- Centre for Evidence Based Medicine Odense (CEBMO) and Cochrane Denmark, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Philippe Ravaud
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Anna Chaimani
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| | - Isabelle Boutron
- Cochrane France, Paris, France
- Centre of Research in Epidemiology and Statistics (CRESS), INSERM, INRAE, Université de Paris, Paris, France
| |
Collapse
|
28
|
Harnessing the Potential of Plant Expression System towards the Production of Vaccines for the Prevention of Human Papillomavirus and Cervical Cancer. Vaccines (Basel) 2022; 10:vaccines10122064. [PMID: 36560473 PMCID: PMC9782824 DOI: 10.3390/vaccines10122064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Cervical cancer is the most common gynecological malignant tumor worldwide, and it remains a major health problem among women, especially in developing countries. Despite the significant research efforts employed for tumor prevention, cervical cancer ranks as the leading cause of cancer death. Human papillomavirus (HPV) is the most important risk factor for cervical cancer. Cervical cancer is a preventable disease, for which early detection could increase survival rates. Immunotherapies represent a promising approach in the treatment of cancer, and several potential candidates are in clinical trials, while some are available in the market. However, equal access to available HPV vaccines is limited due to their high cost, which remains a global challenge for cervical cancer prevention. The implementation of screening programs, disease control systems, and medical advancement in developed countries reduce the serious complications associated with the disease somewhat; however, the incidence and prevalence of cervical cancer in low-income and middle-income countries continues to gradually increase, making it the leading cause of mortality, largely due to the unaffordable and inaccessible anti-cancer therapeutic options. In recent years, plants have been considered as a cost-effective production system for the development of vaccines, therapeutics, and other biopharmaceuticals. Several proof-of-concept studies showed the possibility of producing recombinant biopharmaceuticals for cancer immunotherapy in a plant platform. This review summarizes the current knowledge and therapeutic options for the prevention of cervical cancer and discusses the potential of the plant expression platform to produce affordable HPV vaccines.
Collapse
|
29
|
Jackson MA, Chan LY, Harding MD, Craik DJ, Gilding EK. Rational domestication of a plant-based recombinant expression system expands its biosynthetic range. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:6103-6114. [PMID: 35724659 PMCID: PMC9578353 DOI: 10.1093/jxb/erac273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/20/2022] [Indexed: 05/22/2023]
Abstract
Plant molecular farming aims to provide a green, flexible, and rapid alternative to conventional recombinant expression systems, capable of producing complex biologics such as enzymes, vaccines, and antibodies. Historically, the recombinant expression of therapeutic peptides in plants has proven difficult, largely due to their small size and instability. However, some plant species harbour the capacity for peptide backbone cyclization, a feature inherent in stable therapeutic peptides. One obstacle to realizing the potential of plant-based therapeutic peptide production is the proteolysis of the precursor before it is matured into its final stabilized form. Here we demonstrate the rational domestication of Nicotiana benthamiana within two generations to endow this plant molecular farming host with an expanded repertoire of peptide sequence space. The in planta production of molecules including an insecticidal peptide, a prostate cancer therapeutic lead, and an orally active analgesic is demonstrated.
Collapse
Affiliation(s)
- Mark A Jackson
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Lai Yue Chan
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Maxim D Harding
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| | - Edward K Gilding
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
30
|
Transient Expression of Flavivirus Structural Proteins in Nicotiana benthamiana. Vaccines (Basel) 2022; 10:vaccines10101667. [PMID: 36298532 PMCID: PMC9610170 DOI: 10.3390/vaccines10101667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022] Open
Abstract
Flaviviruses are a threat to public health and can cause major disease outbreaks. Tick-borne encephalitis (TBE) is caused by a flavivirus, and it is one of the most important causes of viral encephalitis in Europe and is on the rise in Sweden. As there is no antiviral treatment available, vaccination remains the best protective measure against TBE. Currently available TBE vaccines are based on formalin-inactivated virus produced in cell culture. These vaccines must be delivered by intramuscular injection, have a burdensome immunization schedule, and may exhibit vaccine failure in certain populations. This project aimed to develop an edible TBE vaccine to trigger a stronger immune response through oral delivery of viral antigens to mucosal surfaces. We demonstrated successful expression and post-translational processing of flavivirus structural proteins which then self-assembled to form virus-like particles in Nicotiana benthamiana. We performed oral toxicity tests in mice using various plant species as potential bioreactors and evaluated the immunogenicity of the resulting edible vaccine candidate. Mice immunized with the edible vaccine candidate did not survive challenge with TBE virus. Interestingly, immunization of female mice with a commercial TBE vaccine can protect their offspring against TBE virus infection.
Collapse
|
31
|
Morales-Hernández S, Ugidos-Damboriena N, López-Sagaseta J. Self-Assembling Protein Nanoparticles in the Design of Vaccines: 2022 Update. Vaccines (Basel) 2022; 10:1447. [PMID: 36146525 PMCID: PMC9505534 DOI: 10.3390/vaccines10091447] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccines constitute a pillar in the prevention of infectious diseases. The unprecedented emergence of novel immunization strategies due to the COVID-19 pandemic has again positioned vaccination as a pivotal measure to protect humankind and reduce the clinical impact and socioeconomic burden worldwide. Vaccination pursues the ultimate goal of eliciting a protective response in immunized individuals. To achieve this, immunogens must be efficiently delivered to prime the immune system and produce robust protection. Given their safety, immunogenicity, and flexibility to display varied and native epitopes, self-assembling protein nanoparticles represent one of the most promising immunogen delivery platforms. Currently marketed vaccines against the human papillomavirus, for instance, illustrate the potential of these nanoassemblies. This review is intended to provide novelties, since 2015, on the ground of vaccine design and self-assembling protein nanoparticles, as well as a comparison with the current emergence of mRNA-based vaccines.
Collapse
Affiliation(s)
- Sergio Morales-Hernández
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| | - Nerea Ugidos-Damboriena
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| | - Jacinto López-Sagaseta
- Unit of Protein Crystallography and Structural Immunology, Navarrabiomed-Public University of Navarra (UPNA), 31008 Pamplona, Spain
- Navarra University Hospital, 31008 Pamplona, Spain
| |
Collapse
|
32
|
Abstract
The idea of producing vaccines in plants originated in the late 1980s. Initially, it was contemplated that this notion could facilitate the concept of edible vaccines, making them more cost effective and easily accessible. Initial studies on edible vaccines focussed on the use of a variety of different transgenic plant host species for the production of vaccine antigens. However, adequate expression levels of antigens, the difficulties predicted with administration of consistent doses, and regulatory rules required for growth of transgenic plants gave way to the development of vaccine candidates that could be purified and administered parenterally. The field has subsequently advanced with improved expression techniques including a shift from using transgenic to transient expression of antigens, refinement of purification protocols, a deeper understanding of the biological processes and a wealth of evidence of immunogenicity and efficacy of plant-produced vaccine candidates, all contributing to the successful practice of what is now known as biopharming or plant molecular farming. The establishment of this technology has resulted in the development of many different types of vaccine candidates including subunit vaccines and various different types of nanoparticle vaccines targeting a wide variety of bacterial and viral diseases. This has brought further acceptance of plants as a suitable platform for vaccine production and in this review, we discuss the most recent advances in the production of vaccines in plants for human use.
Collapse
Affiliation(s)
- Jennifer Stander
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Sandiswa Mbewana
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Ann E Meyers
- Biopharming Research Unit, Department of Molecular and Cell Biology, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa.
| |
Collapse
|
33
|
Carascal MB, Pavon RDN, Rivera WL. Recent Progress in Recombinant Influenza Vaccine Development Toward Heterosubtypic Immune Response. Front Immunol 2022; 13:878943. [PMID: 35663997 PMCID: PMC9162156 DOI: 10.3389/fimmu.2022.878943] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Flu, a viral infection caused by the influenza virus, is still a global public health concern with potential to cause seasonal epidemics and pandemics. Vaccination is considered the most effective protective strategy against the infection. However, given the high plasticity of the virus and the suboptimal immunogenicity of existing influenza vaccines, scientists are moving toward the development of universal vaccines. An important property of universal vaccines is their ability to induce heterosubtypic immunity, i.e., a wide immune response coverage toward different influenza subtypes. With the increasing number of studies and mounting evidence on the safety and efficacy of recombinant influenza vaccines (RIVs), they have been proposed as promising platforms for the development of universal vaccines. This review highlights the current progress and advances in the development of RIVs in the context of heterosubtypic immunity induction toward universal vaccine production. In particular, this review discussed existing knowledge on influenza and vaccine development, current hemagglutinin-based RIVs in the market and in the pipeline, other potential vaccine targets for RIVs (neuraminidase, matrix 1 and 2, nucleoprotein, polymerase acidic, and basic 1 and 2 antigens), and deantigenization process. This review also provided discussion points and future perspectives in looking at RIVs as potential universal vaccine candidates for influenza.
Collapse
Affiliation(s)
- Mark B Carascal
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines.,Clinical and Translational Research Institute, The Medical City, Pasig City, Philippines
| | - Rance Derrick N Pavon
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| | - Windell L Rivera
- Pathogen-Host-Environment Interactions Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman, Quezon City, Philippines
| |
Collapse
|
34
|
Liu T, Li L, Cheng C, He B, Jiang T. Emerging prospects of protein/peptide-based nanoassemblies for drug delivery and vaccine development. NANO RESEARCH 2022; 15:7267-7285. [PMID: 35692441 PMCID: PMC9166156 DOI: 10.1007/s12274-022-4385-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 05/09/2023]
Abstract
Proteins have been widely used in the biomedical field because of their well-defined architecture, accurate molecular weight, excellent biocompatibility and biodegradability, and easy-to-functionalization. Inspired by the wisdom of nature, increasing proteins/peptides that possess self-assembling capabilities have been explored and designed to generate nanoassemblies with unique structure and function, including spatially organized conformation, passive and active targeting, stimuli-responsiveness, and high stability. These characteristics make protein/peptide-based nanoassembly an ideal platform for drug delivery and vaccine development. In this review, we focus on recent advances in subsistent protein/peptide-based nanoassemblies, including protein nanocages, virus-like particles, self-assemblable natural proteins, and self-assemblable artificial peptides. The origin and characteristics of various protein/peptide-based assemblies and their applications in drug delivery and vaccine development are summarized. In the end, the prospects and challenges are discussed for the further development of protein/peptide-based nanoassemblies.
Collapse
Affiliation(s)
- Taiyu Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Lu Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| |
Collapse
|
35
|
Ma Y, Wang Y, Dong C, Gonzalez GX, Zhu W, Kim J, Wei L, Kang S, Wang B. SARS-CoV-2 Spike Stem Protein Nanoparticles Elicited Broad ADCC and Robust Neutralization against Variants in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200836. [PMID: 35607768 PMCID: PMC9233155 DOI: 10.1002/smll.202200836] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/01/2022] [Indexed: 05/03/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused the global pandemic. The virus is rapidly evolving, characterized by the emergence of several major variants. Stable prefusion spike protein (Pre) is the immunogen in current vaccines but is limited in protecting against different variants. Here, the immune responses induced by the relatively conserved stem subunit (S2) of spike protein versus Pre are investigated. Pre generates the most robust neutralization responses against SARS-CoV-2 variants in vesicular stomatitis virus pseudovirus-based assessment but elicits less antibody-dependent cellular cytotoxicity (ADCC) activity than S2. By contrast, S2 induces the most balanced immunoglobulin G (IgG) antibodies with potent and broad ADCC activity although produces weaker neutralization. The immunogenicity of S2 and Pre improves by incorporating the two proteins into double-layered protein nanoparticles. The resulting protein nanoparticles Pre/S2 elicit higher neutralizing antibodies than Pre alone, and stronger ADCC than S2 alone. Moreover, nanoparticles produce more potent and balanced serum IgG antibodies than the corresponding soluble protein mixture, and the immune responses are sustained for at least four months after the immunization. Thus, the double-layered protein nanoparticles have the potential to be developed into broader SARS-CoV-2 vaccines with excellent safety profiles.
Collapse
Affiliation(s)
- Yao Ma
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Ye Wang
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Chunhong Dong
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Gilbert X. Gonzalez
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Wandi Zhu
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Joo Kim
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Lai Wei
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Sang‐Moo Kang
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| | - Bao‐Zhong Wang
- Center for InflammationImmunity & InfectionInstitute for Biomedical SciencesGeorgia State UniversityAtlantaGA30302USA
| |
Collapse
|
36
|
Volokhov DV, Fry AM, Furtak V, Jones RM, Musiychuk K, Norikane J, Green BJ, Srinivas GB, Streatfield SJ, Yusibov V. An ELISA-based antigenicity test of rabies recombinant glycoprotein cannot predict its protective potency in vivo. Mol Cell Probes 2022; 63:101815. [DOI: 10.1016/j.mcp.2022.101815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 03/18/2022] [Indexed: 11/15/2022]
|
37
|
Monreal-Escalante E, Ramos-Vega A, Angulo C, Bañuelos-Hernández B. Plant-Based Vaccines: Antigen Design, Diversity, and Strategies for High Level Production. Vaccines (Basel) 2022; 10:100. [PMID: 35062761 PMCID: PMC8782010 DOI: 10.3390/vaccines10010100] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/25/2021] [Accepted: 01/01/2022] [Indexed: 12/18/2022] Open
Abstract
Vaccines for human use have conventionally been developed by the production of (1) microbial pathogens in eggs or mammalian cells that are then inactivated, or (2) by the production of pathogen proteins in mammalian and insect cells that are purified for vaccine formulation, as well as, more recently, (3) by using RNA or DNA fragments from pathogens. Another approach for recombinant antigen production in the last three decades has been the use of plants as biofactories. Only have few plant-produced vaccines been evaluated in clinical trials to fight against diseases, of which COVID-19 vaccines are the most recent to be FDA approved. In silico tools have accelerated vaccine design, which, combined with transitory antigen expression in plants, has led to the testing of promising prototypes in pre-clinical and clinical trials. Therefore, this review deals with a description of immunoinformatic tools and plant genetic engineering technologies used for antigen design (virus-like particles (VLP), subunit vaccines, VLP chimeras) and the main strategies for high antigen production levels. These key topics for plant-made vaccine development are discussed and perspectives are provided.
Collapse
Affiliation(s)
- Elizabeth Monreal-Escalante
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, Instituto PoliItécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, BCS, Mexico; (A.R.-V.); (C.A.)
- CONACYT—Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, BCS, Mexico
| | - Abel Ramos-Vega
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, Instituto PoliItécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, BCS, Mexico; (A.R.-V.); (C.A.)
| | - Carlos Angulo
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, Instituto PoliItécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz 23096, BCS, Mexico; (A.R.-V.); (C.A.)
| | - Bernardo Bañuelos-Hernández
- Escuela de Veterinaria, Universidad De La Salle Bajío, Avenida Universidad 602, Lomas del Campestre, Leon 37150, GTO, Mexico
| |
Collapse
|
38
|
Abstract
The regulation of molecular farming is a complex topic because plants and plant-based systems are relative newcomers among the many production platforms available for recombinant proteins. The regulations specific for different types of product (human/veterinary pharmaceuticals and medical devices, cosmetics, diagnostics, and research reagents) must therefore be overlaid with the regulations governing hitherto unfamiliar production platforms, and this must be achieved in different jurisdictions that handle genetically modified organisms (and genetically modified plants in particular) in very different ways. This chapter uses examples of different product types and production methods in three different jurisdictions (the USA, the EU, and Canada) to demonstrate some of the challenges facing the regulatory authorities.
Collapse
|
39
|
Garces KN, Cocores AN, Goadsby PJ, Monteith TS. Headache After Vaccination: An Update on Recent Clinical Trials and Real-World Reporting. Curr Pain Headache Rep 2022; 26:895-918. [PMID: 36418848 PMCID: PMC9685066 DOI: 10.1007/s11916-022-01094-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The aim of this review is to characterize headache as a vaccine adverse event (VAE) in clinical trials. RECENT FINDINGS Of the recent phase III vaccine RCTs (non-COVID-19), 53 studies reported on headache (13 infectious agents). The median rate (interquartile range) of headache was 15.6% (IQR: 9.6-37.6%). Of these, 24.5% of the RCTs reported headache greater in the vaccine group compared to the placebo/control group. In the herpes zoster vaccination trials, headache was more common in all active groups: median rate 33.9% (IQR: 29.7-40.5%) as compared to placebo: median rate 17.7% (IQR: 15.4-23.8%). Influenza and HPV vaccination trials were the 2nd and 3rd most common to have headache as a VAE. Of the 6 widely distributed COVID-19 vaccinations, median rate of post-vaccination headache was 39% (IQR: 28-50%). Headache is a common VAE in vaccine trials. Standardized grading methods, predictors of persistence, and treatment regimens are warranted.
Collapse
Affiliation(s)
- Kimberly N. Garces
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| | - Alexandra N. Cocores
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| | - Peter J. Goadsby
- NIHR King’s Clinical Research Facility & Headache Group-Wolfson CARD, King’s College London, London, UK ,Department of Neurology, University of California, Los Angeles, Los Angeles, CA USA
| | - Teshamae S. Monteith
- Division of Headache, Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL USA
| |
Collapse
|
40
|
Lobato Gómez M, Huang X, Alvarez D, He W, Baysal C, Zhu C, Armario‐Najera V, Blanco Perera A, Cerda Bennasser P, Saba‐Mayoral A, Sobrino‐Mengual G, Vargheese A, Abranches R, Abreu IA, Balamurugan S, Bock R, Buyel J, da Cunha NB, Daniell H, Faller R, Folgado A, Gowtham I, Häkkinen ST, Kumar S, Ramalingam SK, Lacorte C, Lomonossoff GP, Luís IM, Ma JK, McDonald KA, Murad A, Nandi S, O’Keefe B, Oksman‐Caldentey K, Parthiban S, Paul MJ, Ponndorf D, Rech E, Rodrigues JCM, Ruf S, Schillberg S, Schwestka J, Shah PS, Singh R, Stoger E, Twyman RM, Varghese IP, Vianna GR, Webster G, Wilbers RHP, Capell T, Christou P. Contributions of the international plant science community to the fight against human infectious diseases - part 1: epidemic and pandemic diseases. PLANT BIOTECHNOLOGY JOURNAL 2021; 19:1901-1920. [PMID: 34182608 PMCID: PMC8486245 DOI: 10.1111/pbi.13657] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 05/03/2023]
Abstract
Infectious diseases, also known as transmissible or communicable diseases, are caused by pathogens or parasites that spread in communities by direct contact with infected individuals or contaminated materials, through droplets and aerosols, or via vectors such as insects. Such diseases cause ˜17% of all human deaths and their management and control places an immense burden on healthcare systems worldwide. Traditional approaches for the prevention and control of infectious diseases include vaccination programmes, hygiene measures and drugs that suppress the pathogen, treat the disease symptoms or attenuate aggressive reactions of the host immune system. The provision of vaccines and biologic drugs such as antibodies is hampered by the high cost and limited scalability of traditional manufacturing platforms based on microbial and animal cells, particularly in developing countries where infectious diseases are prevalent and poorly controlled. Molecular farming, which uses plants for protein expression, is a promising strategy to address the drawbacks of current manufacturing platforms. In this review article, we consider the potential of molecular farming to address healthcare demands for the most prevalent and important epidemic and pandemic diseases, focussing on recent outbreaks of high-mortality coronavirus infections and diseases that disproportionately affect the developing world.
Collapse
Affiliation(s)
- Maria Lobato Gómez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Xin Huang
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Derry Alvarez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Wenshu He
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Can Baysal
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Changfu Zhu
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Victoria Armario‐Najera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Amaya Blanco Perera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Pedro Cerda Bennasser
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Andera Saba‐Mayoral
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | | | - Ashwin Vargheese
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Rita Abranches
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Isabel Alexandra Abreu
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Shanmugaraj Balamurugan
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreIndia
| | - Ralph Bock
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Johannes.F. Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Nicolau B. da Cunha
- Centro de Análise Proteômicas e Bioquímicas de BrasíliaUniversidade Católica de BrasíliaBrasíliaBrazil
| | - Henry Daniell
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Roland Faller
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
| | - André Folgado
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Iyappan Gowtham
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreIndia
| | - Suvi T. Häkkinen
- Industrial Biotechnology and Food SolutionsVTT Technical Research Centre of Finland LtdEspooFinland
| | - Shashi Kumar
- International Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Sathish Kumar Ramalingam
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreIndia
| | - Cristiano Lacorte
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in BiologyParque Estação BiológicaBrasiliaBrazil
| | | | - Ines M. Luís
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Julian K.‐C. Ma
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Karen. A. McDonald
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Andre Murad
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in BiologyParque Estação BiológicaBrasiliaBrazil
| | - Somen Nandi
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Barry O’Keefe
- Molecular Targets ProgramCenter for Cancer Research, National Cancer Institute, and Natural Products BranchDevelopmental Therapeutics ProgramDivision of Cancer Treatment and DiagnosisNational Cancer Institute, NIHFrederickMDUSA
| | | | - Subramanian Parthiban
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreIndia
| | - Mathew J. Paul
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Daniel Ponndorf
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
- Department of Biological ChemistryJohn Innes CentreNorwichUK
| | - Elibio Rech
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in BiologyParque Estação BiológicaBrasiliaBrazil
| | - Julio C. M. Rodrigues
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in BiologyParque Estação BiológicaBrasiliaBrazil
| | - Stephanie Ruf
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for PhytopathologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Jennifer Schwestka
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Priya S. Shah
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Department of Microbiology and Molecular GeneticsUniversity of California, DavisDavisCAUSA
| | - Rahul Singh
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eva Stoger
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Inchakalody P. Varghese
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityCoimbatoreIndia
| | - Giovanni R. Vianna
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in BiologyParque Estação BiológicaBrasiliaBrazil
| | - Gina Webster
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Ruud H. P. Wilbers
- Laboratory of NematologyPlant Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Teresa Capell
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Paul Christou
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
- ICREACatalan Institute for Research and Advanced StudiesBarcelonaSpain
| |
Collapse
|
41
|
Nosaki S, Hoshikawa K, Ezura H, Miura K. Transient protein expression systems in plants and their applications. PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2021; 38:297-304. [PMID: 34782815 PMCID: PMC8562577 DOI: 10.5511/plantbiotechnology.21.0610a] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/10/2021] [Indexed: 06/01/2023]
Abstract
The production of recombinant proteins is important in academic research to identify protein functions. Moreover, recombinant enzymes are used in the food and chemical industries, and high-quality proteins are required for diagnostic, therapeutic, and pharmaceutical applications. Though many recombinant proteins are produced by microbial or mammalian cell-based expression systems, plants have been promoted as alternative, cost-effective, scalable, safe, and sustainable expression systems. The development and improvement of transient expression systems have significantly reduced the period of protein production and increased the yield of recombinant proteins in plants. In this review, we consider the importance of plant-based expression systems for recombinant protein production and as genetic engineering tools.
Collapse
Affiliation(s)
- Shohei Nosaki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Ken Hoshikawa
- Japan International Research Center for Agricultural Sciences, Tsukuba, Ibaraki 305-8686, Japan
| | - Hiroshi Ezura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Kenji Miura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
- Tsukuba-Plant Innovation Research Center, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| |
Collapse
|
42
|
Maharjan PM, Choe S. Plant-Based COVID-19 Vaccines: Current Status, Design, and Development Strategies of Candidate Vaccines. Vaccines (Basel) 2021; 9:992. [PMID: 34579229 PMCID: PMC8473425 DOI: 10.3390/vaccines9090992] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/01/2023] Open
Abstract
The prevalence of the coronavirus disease 2019 (COVID-19) pandemic in its second year has led to massive global human and economic losses. The high transmission rate and the emergence of diverse SARS-CoV-2 variants demand rapid and effective approaches to preventing the spread, diagnosing on time, and treating affected people. Several COVID-19 vaccines are being developed using different production systems, including plants, which promises the production of cheap, safe, stable, and effective vaccines. The potential of a plant-based system for rapid production at a commercial scale and for a quick response to an infectious disease outbreak has been demonstrated by the marketing of carrot-cell-produced taliglucerase alfa (Elelyso) for Gaucher disease and tobacco-produced monoclonal antibodies (ZMapp) for the 2014 Ebola outbreak. Currently, two plant-based COVID-19 vaccine candidates, coronavirus virus-like particle (CoVLP) and Kentucky Bioprocessing (KBP)-201, are in clinical trials, and many more are in the preclinical stage. Interim phase 2 clinical trial results have revealed the high safety and efficacy of the CoVLP vaccine, with 10 times more neutralizing antibody responses compared to those present in a convalescent patient's plasma. The clinical trial of the CoVLP vaccine could be concluded by the end of 2021, and the vaccine could be available for public immunization thereafter. This review encapsulates the efforts made in plant-based COVID-19 vaccine development, the strategies and technologies implemented, and the progress accomplished in clinical trials and preclinical studies so far.
Collapse
Affiliation(s)
- Puna Maya Maharjan
- G+FLAS Life Sciences, 123 Uiryodanji-gil, Osong-eup, Heungdeok-gu, Cheongju-si 28161, Korea;
| | - Sunghwa Choe
- G+FLAS Life Sciences, 38 Nakseongdae-ro, Gwanak-gu, Seoul 08790, Korea
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Gwanak-gu, Seoul 08826, Korea
| |
Collapse
|
43
|
Citiulo F, Crosatti C, Cattivelli L, Biselli C. Frontiers in the Standardization of the Plant Platform for High Scale Production of Vaccines. PLANTS (BASEL, SWITZERLAND) 2021; 10:1828. [PMID: 34579360 PMCID: PMC8467261 DOI: 10.3390/plants10091828] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
The recent COVID-19 pandemic has highlighted the value of technologies that allow a fast setup and production of biopharmaceuticals in emergency situations. The plant factory system can provide a fast response to epidemics/pandemics. Thanks to their scalability and genome plasticity, plants represent advantageous platforms to produce vaccines. Plant systems imply less complicated production processes and quality controls with respect to mammalian and bacterial cells. The expression of vaccines in plants is based on transient or stable transformation systems and the recent progresses in genome editing techniques, based on the CRISPR/Cas method, allow the manipulation of DNA in an efficient, fast, and easy way by introducing specific modifications in specific sites of a genome. Nonetheless, CRISPR/Cas is far away from being fully exploited for vaccine expression in plants. In this review, an overview of the potential conjugation of the renewed vaccine technologies (i.e., virus-like particles-VLPs, and industrialization of the production process) with genome editing to produce vaccines in plants is reported, illustrating the potential advantages in the standardization of the plant platforms, with the overtaking of constancy of large-scale production challenges, facilitating regulatory requirements and expediting the release and commercialization of the vaccine products of genome edited plants.
Collapse
Affiliation(s)
- Francesco Citiulo
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100 Siena, Italy;
| | - Cristina Crosatti
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Luigi Cattivelli
- Council for Agricultural Research and Economics, Research Centre for Genomics and Bioinformatics, Via San Protaso 302, 29017 Fiorenzuola d’Arda, Italy; (C.C.); (L.C.)
| | - Chiara Biselli
- Council for Agricultural Research and Economics, Research Centre for Viticulture and Enology, Viale Santa Margherita 80, 52100 Arezzo, Italy
| |
Collapse
|
44
|
Siriwattananon K, Manopwisedjaroen S, Shanmugaraj B, Rattanapisit K, Phumiamorn S, Sapsutthipas S, Trisiriwanich S, Prompetchara E, Ketloy C, Buranapraditkun S, Wijagkanalan W, Tharakhet K, Kaewpang P, Leetanasaksakul K, Kemthong T, Suttisan N, Malaivijitnond S, Ruxrungtham K, Thitithanyanont A, Phoolcharoen W. Plant-Produced Receptor-Binding Domain of SARS-CoV-2 Elicits Potent Neutralizing Responses in Mice and Non-human Primates. FRONTIERS IN PLANT SCIENCE 2021; 12:682953. [PMID: 34054909 PMCID: PMC8158422 DOI: 10.3389/fpls.2021.682953] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/15/2021] [Indexed: 05/11/2023]
Abstract
The emergence of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has affected global public health and economy. Despite the substantial efforts, only few vaccines are currently approved and some are in the different stages of clinical trials. As the disease rapidly spreads, an affordable and effective vaccine is urgently needed. In this study, we investigated the immunogenicity of plant-produced receptor-binding domain (RBD) of SARS-CoV-2 in order to use as a subunit vaccine. In this regard, RBD of SARS-CoV-2 was fused with Fc fragment of human IgG1 and transiently expressed in Nicotiana benthamiana by agroinfiltration. The plant-produced RBD-Fc fusion protein was purified from the crude extract by using protein A affinity column chromatography. Two intramuscular administration of plant-produced RBD-Fc protein formulated with alum as an adjuvant have elicited high neutralization titers in immunized mice and cynomolgus monkeys. Further it has induced a mixed Th1/Th2 immune responses and vaccine-specific T-lymphocyte responses which was confirmed by interferon-gamma (IFN-γ) enzyme-linked immunospot assay. Altogether, our results demonstrated that the plant-produced SARS-CoV-2 RBD has the potential to be used as an effective vaccine candidate against SARS-CoV-2. To our knowledge, this is the first report demonstrating the immunogenicity of plant-produced SARS-CoV-2 RBD protein in mice and non-human primates.
Collapse
Affiliation(s)
- Konlavat Siriwattananon
- Research Unit for Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | | | | | | | - Supaporn Phumiamorn
- Department of Medical Sciences, Ministry of Public Health, Institute of Biological Products, Nonthaburi, Thailand
| | - Sompong Sapsutthipas
- Department of Medical Sciences, Ministry of Public Health, Institute of Biological Products, Nonthaburi, Thailand
| | - Sakalin Trisiriwanich
- Department of Medical Sciences, Ministry of Public Health, Institute of Biological Products, Nonthaburi, Thailand
| | - Eakachai Prompetchara
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chutitorn Ketloy
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supranee Buranapraditkun
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Kittipan Tharakhet
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
| | - Papatsara Kaewpang
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
| | - Kantinan Leetanasaksakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Taratorn Kemthong
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | - Nutchanat Suttisan
- National Primate Research Center of Thailand-Chulalongkorn University, Saraburi, Thailand
| | | | - Kiat Ruxrungtham
- Faculty of Medicine, Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center, Chula VRC), Chulalongkorn University, Bangkok, Thailand
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Waranyoo Phoolcharoen
- Research Unit for Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
45
|
Ghosn L, Chaimani A, Evrenoglou T, Davidson M, Graña C, Schmucker C, Bollig C, Henschke N, Sguassero Y, Nejstgaard CH, Menon S, Nguyen TV, Ferrand G, Kapp P, Riveros C, Ávila C, Devane D, Meerpohl JJ, Rada G, Hróbjartsson A, Grasselli G, Tovey D, Ravaud P, Boutron I. Interleukin-6 blocking agents for treating COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 3:CD013881. [PMID: 33734435 PMCID: PMC8406988 DOI: 10.1002/14651858.cd013881] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Interleukin 6 (IL-6) blocking agents have been used for treating severe coronavirus disease 2019 (COVID-19). Their immunosuppressive effect might be valuable in patients with COVID-19 characterised by substantial immune system dysfunction by controlling inflammation and promoting disease tolerance. OBJECTIVES To assess the effect of IL-6 blocking agents compared to standard care alone or with placebo on efficacy and safety outcomes in COVID-19. We will update this assessment regularly. SEARCH METHODS We searched the World Health Organization (WHO) International Clinical Trials Registry Platform (up to 11 February 2021) and the L-OVE platform, and Cochrane COVID-19 Study Register to identify trials up to 26 February 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating IL-6 blocking agents compared with standard care alone or with placebo for people with COVID-19, regardless of disease severity. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. The protocol was amended to reduce the number of outcomes considered. Two review authors independently collected data and assessed the risk of bias with the Cochrane Risk of Bias 2 tool. We rated the certainty of evidence with the GRADE approach for the critical outcomes such as clinical improvement (defined as hospital discharge or improvement on the scale used by trialists to evaluate clinical progression or recovery) (day (D) 28 / ≥ D60); WHO Clinical Progression Score of level 7 or above (i.e. the proportion of participants with mechanical ventilation +/- additional organ support OR death) (D28 / ≥ D60); all-cause mortality (D28 / ≥ D60); incidence of any adverse events; and incidence of serious adverse events. MAIN RESULTS We identified 10 RCTs with available data including one platform trial comparing tocilizumab and sarilumab with standard of care. These trials evaluated tocilizumab (nine RCTs including two platform trials; seven were reported as peer-reviewed articles, two as preprints; 6428 randomised participants); and two sarilumab (one platform trial reported as peer reviewed article, one reported as preprint, 880 randomised participants). All trials included were multicentre trials. They were conducted in Brazil, China, France, Italy, UK, USA, and four were multi-country trials. The mean age range of participants ranged from 56 to 65 years; 4572 (66.3%) of trial participants were male. Disease severity ranged from mild to critical disease. The reported proportion of participants on oxygen at baseline but not intubated varied from 56% to 100% where reported. Five trials reported the inclusion of intubated patients at baseline. We identified a further 20 registered RCTs of tocilizumab compared to placebo/standard care (five completed without available results, five terminated without available results, eight ongoing, two not recruiting); 11 RCTs of sarilumab (two completed without results, three terminated without available results, six ongoing); six RCTs of clazakisumab (five ongoing, one not recruiting); two RCTs of olokizumab (one completed, one not recruiting); one of siltuximab (ongoing) and one RCT of levilimab (completed without available results). Of note, three were cancelled (2 tocilizumab, 1 clazakisumab). One multiple-arm RCT evaluated both tocilizumab and sarilumab compared to standard of care, one three-arm RCT evaluated tocilizumab and siltuximab compared to standard of care and consequently they appear in each respective comparison. Tocilizumab versus standard care alone or with placebo a. Effectiveness of tocilizumab for patients with COVID-19 Tocilizumab probably results in little or no increase in the outcome of clinical improvement at D28 (RR 1.06, 95% CI 1.00 to 1.13; I2 = 40.9%; 7 RCTs, 5585 participants; absolute effect: 31 more with clinical improvement per 1000 (from 0 fewer to 67 more); moderate-certainty evidence). However, we cannot exclude that some subgroups of patients could benefit from the treatment. We did not obtain data for longer-term follow-up (≥ D60). The effect of tocilizumab on the proportion of participants with a WHO Clinical Progression Score of level of 7 or above is uncertain at D28 (RR 0.99, 95% CI 0.56 to 1.74; I2 = 64.4%; 3 RCTs, 712 participants; low-certainty evidence). We did not obtain data for longer-term follow-up (≥ D60). Tocilizumab reduces all-cause mortality at D28 compared to standard care alone or placebo (RR 0.89, 95% CI 0.82 to 0.97; I2 = 0.0%; 8 RCTs, 6363 participants; absolute effect: 32 fewer deaths per 1000 (from 52 fewer to 9 fewer); high-certainty evidence). The evidence suggests uncertainty around the effect on mortality at ≥ D60 (RR 0.86, 95% CI 0.53 to 1.40; I2 = 0.0%; 2 RCTs, 519 participants; low-certainty evidence). b. Safety of tocilizumab for patients with COVID-19 The evidence is very uncertain about the effect of tocilizumab on adverse events (RR 1.23, 95% CI 0.87 to 1.72; I2 = 86.4%; 7 RCTs, 1534 participants; very low-certainty evidence). Nevertheless, tocilizumab probably results in slightly fewer serious adverse events than standard care alone or placebo (RR 0.89, 95% CI 0.75 to 1.06; I2 = 0.0%; 8 RCTs, 2312 participants; moderate-certainty evidence). Sarilumab versus standard care alone or with placebo The evidence is uncertain about the effect of sarilumab on all-cause mortality at D28 (RR 0.77, 95% CI 0.43 to 1.36; 2 RCTs, 880 participants; low certainty), on all-cause mortality at ≥ D60 (RR 1.00, 95% CI 0.50 to 2.0; 1 RCT, 420 participants; low certainty), and serious adverse events (RR 1.17, 95% CI 0.77 to 1.77; 2 RCTs, 880 participants; low certainty). It is unlikely that sarilumab results in an important increase of adverse events (RR 1.05, 95% CI 0.88 to 1.25; 1 RCT, 420 participants; moderate certainty). However, an increase cannot be excluded No data were available for other critical outcomes. AUTHORS' CONCLUSIONS On average, tocilizumab reduces all-cause mortality at D28 compared to standard care alone or placebo and probably results in slightly fewer serious adverse events than standard care alone or placebo. Nevertheless, tocilizumab probably results in little or no increase in the outcome clinical improvement (defined as hospital discharge or improvement measured by trialist-defined scales) at D28. The impact of tocilizumab on other outcomes is uncertain or very uncertain. With the data available, we were not able to explore heterogeneity. Individual patient data meta-analyses are needed to be able to identify which patients are more likely to benefit from this treatment. Evidence for an effect of sarilumab is uncertain and evidence for other anti-IL6 agents is unavailable. Thirty-nine RCTs of IL-6 blocking agents with no results are currently registered, of which nine are completed and seven trials were terminated with no results available. The findings of this review will be updated as new data are made available on the COVID-NMA platform (covid-nma.com).
Collapse
Affiliation(s)
- Lina Ghosn
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Anna Chaimani
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Theodoros Evrenoglou
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Mauricia Davidson
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Carolina Graña
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Christine Schmucker
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Bollig
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | - Camilla Hansen Nejstgaard
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Sonia Menon
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Thu Van Nguyen
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Gabriel Ferrand
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Philipp Kapp
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Carolina Riveros
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | | | - Declan Devane
- HRB-Trials Methodology Research Network, National University of Ireland Galway, Galway, Ireland
- Evidence Synthesis Ireland and Cochrane Ireland, Galway, Ireland
| | - Joerg J Meerpohl
- Cochrane Germany, Cochrane Germany Foundation, Freiburg, Germany
- Institute for Evidence in Medicine (for Cochrane Germany Foundation), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gabriel Rada
- Epistemonikos Foundation, Santiago, Chile
- UC Evidence Center, Cochrane Chile Associated Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Asbjørn Hróbjartsson
- Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Open Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Giacomo Grasselli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Anesthesia, Intensive Care and Emergency, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Department of Pathophysiology and Transplantation, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Philippe Ravaud
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| | - Isabelle Boutron
- Cochrane France, Paris, France
- Centre d'Épidémiologie Clinique, AP-HP (Assistance Publique des Hôpitaux de Paris), Hôpital Hôtel Dieu, Paris, France
- Université de Paris, Centre of Research in Epidemiology and Statistics (CRESS), INSERM, F-75004, Paris, France
| |
Collapse
|