1
|
Su H, van Eerde A, Rimstad E, Bock R, Branza-Nichita N, Yakovlev IA, Clarke JL. Plant-made vaccines against viral diseases in humans and farm animals. FRONTIERS IN PLANT SCIENCE 2023; 14:1170815. [PMID: 37056490 PMCID: PMC10086147 DOI: 10.3389/fpls.2023.1170815] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/14/2023] [Indexed: 06/19/2023]
Abstract
Plants provide not only food and feed, but also herbal medicines and various raw materials for industry. Moreover, plants can be green factories producing high value bioproducts such as biopharmaceuticals and vaccines. Advantages of plant-based production platforms include easy scale-up, cost effectiveness, and high safety as plants are not hosts for human and animal pathogens. Plant cells perform many post-translational modifications that are present in humans and animals and can be essential for biological activity of produced recombinant proteins. Stimulated by progress in plant transformation technologies, substantial efforts have been made in both the public and the private sectors to develop plant-based vaccine production platforms. Recent promising examples include plant-made vaccines against COVID-19 and Ebola. The COVIFENZ® COVID-19 vaccine produced in Nicotiana benthamiana has been approved in Canada, and several plant-made influenza vaccines have undergone clinical trials. In this review, we discuss the status of vaccine production in plants and the state of the art in downstream processing according to good manufacturing practice (GMP). We discuss different production approaches, including stable transgenic plants and transient expression technologies, and review selected applications in the area of human and veterinary vaccines. We also highlight specific challenges associated with viral vaccine production for different target organisms, including lower vertebrates (e.g., farmed fish), and discuss future perspectives for the field.
Collapse
Affiliation(s)
- Hang Su
- Division of Biotechnology and Plant Health, NIBIO - Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - André van Eerde
- Division of Biotechnology and Plant Health, NIBIO - Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - Espen Rimstad
- Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - Ralph Bock
- Department III, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Norica Branza-Nichita
- Department of Viral Glycoproteins, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Igor A. Yakovlev
- Division of Biotechnology and Plant Health, NIBIO - Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - Jihong Liu Clarke
- Division of Biotechnology and Plant Health, NIBIO - Norwegian Institute of Bioeconomy Research, Ås, Norway
| |
Collapse
|
2
|
Kim SA, Lee Y, Ko Y, Kim S, Kim GB, Lee NK, Ahn W, Kim N, Nam GH, Lee EJ, Kim IS. Protein-based nanocages for vaccine development. J Control Release 2023; 353:767-791. [PMID: 36516900 DOI: 10.1016/j.jconrel.2022.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Protein nanocages have attracted considerable attention in various fields of nanomedicine due to their intrinsic properties, including biocompatibility, biodegradability, high structural stability, and ease of modification of their surfaces and inner cavities. In vaccine development, these protein nanocages are suited for efficient targeting to and retention in the lymph nodes and can enhance immunogenicity through various mechanisms, including excellent uptake by antigen-presenting cells and crosslinking with multiple B cell receptors. This review highlights the superiority of protein nanocages as antigen delivery carriers based on their physiological and immunological properties such as biodistribution, immunogenicity, stability, and multifunctionality. With a focus on design, we discuss the utilization and efficacy of protein nanocages such as virus-like particles, caged proteins, and artificial caged proteins against cancer and infectious diseases such as coronavirus disease 2019 (COVID-19). In addition, we summarize available knowledge on the protein nanocages that are currently used in clinical trials and provide a general outlook on conventional distribution techniques and hurdles faced, particularly for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Yeju Ko
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Seohyun Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Gi Beom Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Na Kyeong Lee
- Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Wonkyung Ahn
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Gi-Hoon Nam
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
3
|
Plant-Derived Recombinant Vaccines against Zoonotic Viruses. Life (Basel) 2022; 12:life12020156. [PMID: 35207444 PMCID: PMC8878793 DOI: 10.3390/life12020156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging and re-emerging zoonotic diseases cause serious illness with billions of cases, and millions of deaths. The most effective way to restrict the spread of zoonotic viruses among humans and animals and prevent disease is vaccination. Recombinant proteins produced in plants offer an alternative approach for the development of safe, effective, inexpensive candidate vaccines. Current strategies are focused on the production of highly immunogenic structural proteins, which mimic the organizations of the native virion but lack the viral genetic material. These include chimeric viral peptides, subunit virus proteins, and virus-like particles (VLPs). The latter, with their ability to self-assemble and thus resemble the form of virus particles, are gaining traction among plant-based candidate vaccines against many infectious diseases. In this review, we summarized the main zoonotic diseases and followed the progress in using plant expression systems for the production of recombinant proteins and VLPs used in the development of plant-based vaccines against zoonotic viruses.
Collapse
|
4
|
Producing Vaccines against Enveloped Viruses in Plants: Making the Impossible, Difficult. Vaccines (Basel) 2021; 9:vaccines9070780. [PMID: 34358196 PMCID: PMC8310165 DOI: 10.3390/vaccines9070780] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 12/13/2022] Open
Abstract
The past 30 years have seen the growth of plant molecular farming as an approach to the production of recombinant proteins for pharmaceutical and biotechnological uses. Much of this effort has focused on producing vaccine candidates against viral diseases, including those caused by enveloped viruses. These represent a particular challenge given the difficulties associated with expressing and purifying membrane-bound proteins and achieving correct assembly. Despite this, there have been notable successes both from a biochemical and a clinical perspective, with a number of clinical trials showing great promise. This review will explore the history and current status of plant-produced vaccine candidates against enveloped viruses to date, with a particular focus on virus-like particles (VLPs), which mimic authentic virus structures but do not contain infectious genetic material.
Collapse
|
5
|
Venkataraman S, Hefferon K, Makhzoum A, Abouhaidar M. Combating Human Viral Diseases: Will Plant-Based Vaccines Be the Answer? Vaccines (Basel) 2021; 9:vaccines9070761. [PMID: 34358177 PMCID: PMC8310141 DOI: 10.3390/vaccines9070761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 12/28/2022] Open
Abstract
Molecular pharming or the technology of application of plants and plant cell culture to manufacture high-value recombinant proteins has progressed a long way over the last three decades. Whether generated in transgenic plants by stable expression or in plant virus-based transient expression systems, biopharmaceuticals have been produced to combat several human viral diseases that have impacted the world in pandemic proportions. Plants have been variously employed in expressing a host of viral antigens as well as monoclonal antibodies. Many of these biopharmaceuticals have shown great promise in animal models and several of them have performed successfully in clinical trials. The current review elaborates the strategies and successes achieved in generating plant-derived vaccines to target several virus-induced health concerns including highly communicable infectious viral diseases. Importantly, plant-made biopharmaceuticals against hepatitis B virus (HBV), hepatitis C virus (HCV), the cancer-causing virus human papillomavirus (HPV), human immunodeficiency virus (HIV), influenza virus, zika virus, and the emerging respiratory virus, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have been discussed. The use of plant virus-derived nanoparticles (VNPs) and virus-like particles (VLPs) in generating plant-based vaccines are extensively addressed. The review closes with a critical look at the caveats of plant-based molecular pharming and future prospects towards further advancements in this technology. The use of biopharmed viral vaccines in human medicine and as part of emergency response vaccines and therapeutics in humans looks promising for the near future.
Collapse
Affiliation(s)
- Srividhya Venkataraman
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
- Correspondence:
| | - Kathleen Hefferon
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| | - Abdullah Makhzoum
- Department of Biological Sciences & Biotechnology, Botswana International University of Science & Technology, Palapye, Botswana;
| | - Mounir Abouhaidar
- Virology Laboratory, Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3B2, Canada; (K.H.); (M.A.)
| |
Collapse
|
6
|
Charlton Hume HK, Vidigal J, Carrondo MJT, Middelberg APJ, Roldão A, Lua LHL. Synthetic biology for bioengineering virus-like particle vaccines. Biotechnol Bioeng 2019; 116:919-935. [PMID: 30597533 PMCID: PMC7161758 DOI: 10.1002/bit.26890] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/08/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Vaccination is the most effective method of disease prevention and control. Many viruses and bacteria that once caused catastrophic pandemics (e.g., smallpox, poliomyelitis, measles, and diphtheria) are either eradicated or effectively controlled through routine vaccination programs. Nonetheless, vaccine manufacturing remains incredibly challenging. Viruses exhibiting high antigenic diversity and high mutation rates cannot be fairly contested using traditional vaccine production methods and complexities surrounding the manufacturing processes, which impose significant limitations. Virus-like particles (VLPs) are recombinantly produced viral structures that exhibit immunoprotective traits of native viruses but are noninfectious. Several VLPs that compositionally match a given natural virus have been developed and licensed as vaccines. Expansively, a plethora of studies now confirms that VLPs can be designed to safely present heterologous antigens from a variety of pathogens unrelated to the chosen carrier VLPs. Owing to this design versatility, VLPs offer technological opportunities to modernize vaccine supply and disease response through rational bioengineering. These opportunities are greatly enhanced with the application of synthetic biology, the redesign and construction of novel biological entities. This review outlines how synthetic biology is currently applied to engineer VLP functions and manufacturing process. Current and developing technologies for the identification of novel target-specific antigens and their usefulness for rational engineering of VLP functions (e.g., presentation of structurally diverse antigens, enhanced antigen immunogenicity, and improved vaccine stability) are described. When applied to manufacturing processes, synthetic biology approaches can also overcome specific challenges in VLP vaccine production. Finally, we address several challenges and benefits associated with the translation of VLP vaccine development into the industry.
Collapse
Affiliation(s)
- Hayley K. Charlton Hume
- The University of Queensland, Australian Institute of Bioengineering and NanotechnologySt LuciaQueenslandAustralia
| | - João Vidigal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | - Manuel J. T. Carrondo
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
| | - Anton P. J. Middelberg
- Faculty of Engineering, Computer and Mathematical Sciences, The University of AdelaideAdelaideSouth AustraliaAustralia
| | - António Roldão
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | | |
Collapse
|
7
|
Ibrahim A, Odon V, Kormelink R. Plant Viruses in Plant Molecular Pharming: Toward the Use of Enveloped Viruses. FRONTIERS IN PLANT SCIENCE 2019; 10:803. [PMID: 31275344 PMCID: PMC6594412 DOI: 10.3389/fpls.2019.00803] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/04/2019] [Indexed: 05/03/2023]
Abstract
Plant molecular pharming has emerged as a reliable platform for recombinant protein expression providing a safe and low-cost alternative to bacterial and mammalian cells-based systems. Simultaneously, plant viruses have evolved from pathogens to molecular tools for recombinant protein expression, chimaeric viral vaccine production, and lately, as nanoagents for drug delivery. This review summarizes the genesis of viral vectors and agroinfection, the development of non-enveloped viruses for various biotechnological applications, and the on-going research on enveloped plant viruses.
Collapse
|
8
|
Steele JFC, Peyret H, Saunders K, Castells‐Graells R, Marsian J, Meshcheriakova Y, Lomonossoff GP. Synthetic plant virology for nanobiotechnology and nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:e1447. [PMID: 28078770 PMCID: PMC5484280 DOI: 10.1002/wnan.1447] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/12/2016] [Accepted: 11/23/2016] [Indexed: 12/12/2022]
Abstract
Nanotechnology is a rapidly expanding field seeking to utilize nano-scale structures for a wide range of applications. Biologically derived nanostructures, such as viruses and virus-like particles (VLPs), provide excellent platforms for functionalization due to their physical and chemical properties. Plant viruses, and VLPs derived from them, have been used extensively in biotechnology. They have been characterized in detail over several decades and have desirable properties including high yields, robustness, and ease of purification. Through modifications to viral surfaces, either interior or exterior, plant-virus-derived nanoparticles have been shown to support a range of functions of potential interest to medicine and nano-technology. In this review we highlight recent and influential achievements in the use of plant virus particles as vehicles for diverse functions: from delivery of anticancer compounds, to targeted bioimaging, vaccine production to nanowire formation. WIREs Nanomed Nanobiotechnol 2017, 9:e1447. doi: 10.1002/wnan.1447 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
| | - Hadrien Peyret
- Department of Biology ChemistryJohn Innes CentreNorwichUK
| | - Keith Saunders
- Department of Biology ChemistryJohn Innes CentreNorwichUK
| | | | | | | | | |
Collapse
|
9
|
Salazar-González JA, Bañuelos-Hernández B, Rosales-Mendoza S. Current status of viral expression systems in plants and perspectives for oral vaccines development. PLANT MOLECULAR BIOLOGY 2015; 87:203-17. [PMID: 25560432 DOI: 10.1007/s11103-014-0279-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 12/22/2014] [Indexed: 05/23/2023]
Abstract
During the last 25 years, the technology to produce recombinant vaccines in plant cells has evolved from modest proofs of the concept to viable technologies adopted by some companies due to significant improvements in the field. Viral-based expression strategies have importantly contributed to this success owing to high yields, short production time (which is in most cases free of tissue culture steps), and the implementation of confined processes for production under GMPs. Herein the distinct expression systems based on viral elements are analyzed. This review also presents the outlook on how these technologies have been successfully applied to the development of plant-based vaccines, some of them being in advanced stages of development. Perspectives on how viral expression systems could allow for the development of innovative oral vaccines constituted by minimally-processed plant biomass are discussed.
Collapse
Affiliation(s)
- Jorge A Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, 78210, San Luis Potosí, SLP, Mexico
| | | | | |
Collapse
|
10
|
Aboul-Ata AAE, Vitti A, Nuzzaci M, El-Attar AK, Piazzolla G, Tortorella C, Harandi AM, Olson O, Wright SA, Piazzolla P. Plant-based vaccines: novel and low-cost possible route for Mediterranean innovative vaccination strategies. Adv Virus Res 2014; 89:1-37. [PMID: 24751193 DOI: 10.1016/b978-0-12-800172-1.00001-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A plant bioreactor has enormous capability as a system that supports many biological activities, that is, production of plant bodies, virus-like particles (VLPs), and vaccines. Foreign gene expression is an efficient mechanism for getting protein vaccines against different human viral and nonviral diseases. Plants make it easy to deal with safe, inexpensive, and provide trouble-free storage. The broad spectrum of safe gene promoters is being used to avoid risk assessments. Engineered virus-based vectors have no side effect. The process can be manipulated as follows: (a) retrieve and select gene encoding, use an antigenic protein from GenBank and/or from a viral-genome sequence, (b) design and construct hybrid-virus vectors (viral vector with a gene of interest) eventually flanked by plant-specific genetic regulatory elements for constitutive expression for obtaining chimeric virus, (c) gene transformation and/or transfection, for transient expression, into a plant-host model, that is, tobacco, to get protocols processed positively, and then moving into edible host plants, (d) confirmation of protein expression by bioassay, PCR-associated tests (RT-PCR), Northern and Western blotting analysis, and serological assay (ELISA), (e) expression for adjuvant recombinant protein seeking better antigenicity, (f) extraction and purification of expressed protein for identification and dosing, (g) antigenicity capability evaluated using parental or oral delivery in animal models (mice and/or rabbit immunization), and (h) growing of construct-treated edible crops in protective green houses. Some successful cases of heterologous gene-expressed protein, as edible vaccine, are being discussed, that is, hepatitis C virus (HCV). R9 mimotope, also named hypervariable region 1 (HVR1), was derived from the HVR1 of HCV. It was used as a potential neutralizing epitope of HCV. The mimotope was expressed using cucumber mosaic virus coat protein (CP), alfalfa mosaic virus CP P3/RNA3, and tobacco mosaic virus (TMV) CP-tobacco mild green mosaic virus (TMGMV) CP as expression vectors into tobacco plants. Expressed recombinant protein has not only been confirmed as a therapeutic but also as a diagnostic tool. Herpes simplex virus 2 (HSV-2), HSV-2 gD, and HSV-2 VP16 subunits were transfected into tobacco plants, using TMV CP-TMGMV CP expression vectors.
Collapse
Affiliation(s)
- Aboul-Ata E Aboul-Ata
- Molecular Biology Laboratory II, Plant Virus and Phytoplasma Research Department, Plant Pathology Research Institute, ARC, Giza, Egypt.
| | - Antonella Vitti
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| | - Maria Nuzzaci
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| | - Ahmad K El-Attar
- Molecular Biology Laboratory II, Plant Virus and Phytoplasma Research Department, Plant Pathology Research Institute, ARC, Giza, Egypt
| | - Giuseppina Piazzolla
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Allergology and Immunology, University of Bari, Bari, Italy
| | - Cosimo Tortorella
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Allergology and Immunology, University of Bari, Bari, Italy
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Olof Olson
- Department of Pure and Applied Biochemistry, Lund University, Lund, Sweden
| | - Sandra A Wright
- Department of Electronics, Mathematics and Natural Sciences, University of Gävle, Gävle, Sweden
| | - Pasquale Piazzolla
- School of Agricultural, Forestry, Food and Environmental Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
11
|
Cañizares MC, Lomonossoff GP, Nicholson L. Development of cowpea mosaic virus-based vectors for the production of vaccines in plants. Expert Rev Vaccines 2014; 4:687-97. [PMID: 16221070 DOI: 10.1586/14760584.4.5.687] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Plant viruses are emerging as an attractive alternative to stable genetic transformation for the expression of foreign proteins in plants. The main advantages of using this strategy are that viral genomes are small and easy to manipulate, infection of plants with modified viruses is simpler and quicker than the regeneration of stably transformed plants and the sequence inserted into a virus vector will be highly amplified. One use of these virus expression systems is for vaccine production. Among plant viruses, cowpea mosaic virus makes an ideal candidate for the production of such vaccines because it grows extremely well in host plants, is very stable, and the purification of virus particles, if required, is straightforward. In this article, the authors review the progress made in the development of cowpea mosaic virus-based vectors for vaccine production, making use of two main approaches: epitope presentation and polypeptide expression.
Collapse
Affiliation(s)
- M Carmen Cañizares
- Department of Biological Chemistry, John Innes Centre, Norwich, NR4 7UH, UK.
| | | | | |
Collapse
|
12
|
Jones RM, Chichester JA, Mett V, Jaje J, Tottey S, Manceva S, Casta LJ, Gibbs SK, Musiychuk K, Shamloul M, Norikane J, Mett V, Streatfield SJ, van de Vegte-Bolmer M, Roeffen W, Sauerwein RW, Yusibov V. A plant-produced Pfs25 VLP malaria vaccine candidate induces persistent transmission blocking antibodies against Plasmodium falciparum in immunized mice. PLoS One 2013; 8:e79538. [PMID: 24260245 PMCID: PMC3832600 DOI: 10.1371/journal.pone.0079538] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/20/2013] [Indexed: 11/18/2022] Open
Abstract
Malaria transmission blocking vaccines (TBVs) are considered an effective means to control and eventually eliminate malaria. The Pfs25 protein, expressed predominantly on the surface of the sexual and sporogonic stages of Plasmodium falciparum including gametes, zygotes and ookinetes, is one of the primary targets for TBV. It has been demonstrated that plants are an effective, highly scalable system for the production of recombinant proteins, including virus-like particles (VLPs). We engineered VLPs (Pfs25-CP VLP) comprising Pfs25 fused to the Alfalfa mosaic virus coat protein (CP) and produced these non-enveloped hybrid VLPs in Nicotiana benthamiana plants using a Tobacco mosaic virus-based ‘launch’ vector. Purified Pfs25-CP VLPs were highly consistent in size (19.3±2.4 nm in diameter) with an estimated 20–30% incorporation of Pfs25 onto the VLP surface. Immunization of mice with one or two doses of Pfs25-CP VLPs plus Alhydrogel® induced serum antibodies with complete transmission blocking activity through the 6 month study period. These results support the evaluation of Pfs25-CP VLP as a potential TBV candidate and the feasibility of the ‘launch’ vector technology for the production of VLP-based recombinant vaccines against infectious diseases.
Collapse
Affiliation(s)
- R. Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jessica A. Chichester
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Vadim Mett
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Jennifer Jaje
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Slobodanka Manceva
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Louis J. Casta
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Sandra K. Gibbs
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Konstantin Musiychuk
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Valentina Mett
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Stephen J. Streatfield
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | | | - Will Roeffen
- Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
- * E-mail:
| |
Collapse
|
13
|
Chen Q, Lai H. Plant-derived virus-like particles as vaccines. Hum Vaccin Immunother 2013; 9:26-49. [PMID: 22995837 PMCID: PMC3667944 DOI: 10.4161/hv.22218] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/06/2012] [Accepted: 09/13/2012] [Indexed: 02/07/2023] Open
Abstract
Virus-like particles (VLPs) are self-assembled structures derived from viral antigens that mimic the native architecture of viruses but lack the viral genome. VLPs have emerged as a premier vaccine platform due to their advantages in safety, immunogenicity, and manufacturing. The particulate nature and high-density presentation of viral structure proteins on their surface also render VLPs as attractive carriers for displaying foreign epitopes. Consequently, several VLP-based vaccines have been licensed for human use and achieved significant clinical and economical success. The major challenge, however, is to develop novel production platforms that can deliver VLP-based vaccines while significantly reducing production times and costs. Therefore, this review focuses on the essential role of plants as a novel, speedy and economical production platform for VLP-based vaccines. The advantages of plant expression systems are discussed in light of their distinctive posttranslational modifications, cost-effectiveness, production speed, and scalability. Recent achievements in the expression and assembly of VLPs and their chimeric derivatives in plant systems as well as their immunogenicity in animal models are presented. Results of human clinical trials demonstrating the safety and efficacy of plant-derived VLPs are also detailed. Moreover, the promising implications of the recent creation of "humanized" glycosylation plant lines as well as the very recent approval of the first plant-made biologics by the U. S. Food and Drug Administration (FDA) for plant production and commercialization of VLP-based vaccines are discussed. It is speculated that the combined potential of plant expression systems and VLP technology will lead to the emergence of successful vaccines and novel applications of VLPs in the near future.
Collapse
Affiliation(s)
- Qiang Chen
- Center for Infectious Diseases and Vaccinology, Biodesign Institute at Arizona State University, Tempe, AZ USA.
| | | |
Collapse
|
14
|
Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine 2012; 31:58-83. [PMID: 23142589 PMCID: PMC7115575 DOI: 10.1016/j.vaccine.2012.10.083] [Citation(s) in RCA: 423] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 10/13/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are a class of subunit vaccines that differentiate themselves from soluble recombinant antigens by stronger protective immunogenicity associated with the VLP structure. Like parental viruses, VLPs can be either non-enveloped or enveloped, and they can form following expression of one or several viral structural proteins in a recombinant heterologous system. Depending on the complexity of the VLP, it can be produced in either a prokaryotic or eukaryotic expression system using target-encoding recombinant vectors, or in some cases can be assembled in cell-free conditions. To date, a wide variety of VLP-based candidate vaccines targeting various viral, bacterial, parasitic and fungal pathogens, as well as non-infectious diseases, have been produced in different expression systems. Some VLPs have entered clinical development and a few have been licensed and commercialized. This article reviews VLP-based vaccines produced in different systems, their immunogenicity in animal models and their status in clinical development.
Collapse
Affiliation(s)
- Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | | | | |
Collapse
|
15
|
Evidence against extracellular exposure of a highly immunogenic region in the C-terminal domain of the simian immunodeficiency virus gp41 transmembrane protein. J Virol 2011; 86:1145-57. [PMID: 22072749 DOI: 10.1128/jvi.06463-11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The generally accepted model for human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein topology includes a single membrane-spanning domain. An alternate model has been proposed which features multiple membrane-spanning domains. Consistent with the alternate model, a high percentage of HIV-1-infected individuals produce unusually robust antibody responses to a region of envelope, the so-called "Kennedy epitope," that in the conventional model should be in the cytoplasm. Here we show analogous, robust antibody responses in simian immunodeficiency virus SIVmac239-infected rhesus macaques to a region of SIVmac239 envelope located in the C-terminal domain, which in the conventional model should be inside the cell. Sera from SIV-infected rhesus macaques consistently reacted with overlapping oligopeptides corresponding to a region located within the cytoplasmic domain of gp41 by the generally accepted model, at intensities comparable to those observed for immunodominant areas of the surface component gp120. Rabbit serum raised against this highly immunogenic region (HIR) reacted with SIV envelope in cell surface-staining experiments, as did monoclonal anti-HIR antibodies isolated from an SIVmac239-infected rhesus macaque. However, control experiments demonstrated that this surface staining could be explained in whole or in part by the release of envelope protein from expressing cells into the supernatant and the subsequent attachment to the surfaces of cells in the culture. Serum and monoclonal antibodies directed against the HIR failed to neutralize even the highly neutralization-sensitive strain SIVmac316. Furthermore, a potential N-linked glycosylation site located close to the HIR and postulated to be outside the cell in the alternate model was not glycosylated. An artificially introduced glycosylation site within the HIR was also not utilized for glycosylation. Together, these data support the conventional model of SIV envelope as a type Ia transmembrane protein with a single membrane-spanning domain and without any extracellular loops.
Collapse
|
16
|
Plummer EM, Manchester M. Viral nanoparticles and virus-like particles: platforms for contemporary vaccine design. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 3:174-196. [PMID: 20872839 PMCID: PMC7169818 DOI: 10.1002/wnan.119] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Current vaccines that provide protection against infectious diseases have primarily relied on attenuated or inactivated pathogens. Virus‐like particles (VLPs), comprised of capsid proteins that can initiate an immune response but do not include the genetic material required for replication, promote immunogenicity and have been developed and approved as vaccines in some cases. In addition, many of these VLPs can be used as molecular platforms for genetic fusion or chemical attachment of heterologous antigenic epitopes. This approach has been shown to provide protective immunity against the foreign epitopes in many cases. A variety of VLPs and virus‐based nanoparticles are being developed for use as vaccines and epitope platforms. These particles have the potential to increase efficacy of current vaccines as well as treat diseases for which no effective vaccines are available. WIREs Nanomed Nanobiotechnol 2011 3 174–196 DOI: 10.1002/wnan.119 This article is categorized under:
Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease
Collapse
Affiliation(s)
- Emily M Plummer
- Cell Biology Department, The Scripps Research Institute, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Marianne Manchester
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Sainsbury F, Cañizares MC, Lomonossoff GP. Cowpea mosaic virus: the plant virus-based biotechnology workhorse. ANNUAL REVIEW OF PHYTOPATHOLOGY 2010; 48:437-55. [PMID: 20455698 DOI: 10.1146/annurev-phyto-073009-114242] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In the 50 years since it was first described, Cowpea mosaic virus (CPMV) has become one of the most intensely studied plant viruses. Research in the past 15 to 20 years has shifted from studying the underlying genetics and structure of the virus to focusing on ways in which it can be exploited in biotechnology. This work led first to the use of virus particles to present peptides, then to the creation of a variety of replicating virus vectors and finally to the development of a highly efficient protein expression system that does not require viral replication. The circle has been completed by the use of the latter system to create empty particles for peptide presentation and other novel uses. The history of CPMV in biotechnology can be likened to an Ouroborus, an ancient symbol depicting a snake or dragon swallowing its own tail, thus forming a circle.
Collapse
Affiliation(s)
- Frank Sainsbury
- Department of Biological Chemistry, John Innes Centre, Norwich NR4 7UH,United Kingdom.
| | | | | |
Collapse
|
18
|
Gonzalez MJ, Plummer EM, Rae CS, Manchester M. Interaction of Cowpea mosaic virus (CPMV) nanoparticles with antigen presenting cells in vitro and in vivo. PLoS One 2009; 4:e7981. [PMID: 19956734 PMCID: PMC2776531 DOI: 10.1371/journal.pone.0007981] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Accepted: 10/27/2009] [Indexed: 11/19/2022] Open
Abstract
Background Plant viruses such as Cowpea mosaic virus (CPMV) are increasingly being developed for applications in nanobiotechnology including vaccine development because of their potential for producing large quantities of antigenic material in plant hosts. In order to improve efficacy of viral nanoparticles in these types of roles, an investigation of the individual cell types that interact with the particles is critical. In particular, it is important to understand the interactions of a potential vaccine with antigen presenting cells (APCs) of the immune system. CPMV was previously shown to interact with vimentin displayed on cell surfaces to mediate cell entry, but the expression of surface vimentin on APCs has not been characterized. Methodology The binding and internalization of CPMV by several populations of APCs was investigated both in vitro and in vivo by flow cytometry and fluorescence confocal microscopy. The association of the particles with mouse gastrointestinal epithelium and Peyer's patches was also examined by confocal microscopy. The expression of surface vimentin on APCs was also measured. Conclusions We found that CPMV is bound and internalized by subsets of several populations of APCs both in vitro and in vivo following intravenous, intraperitoneal, and oral administration, and also by cells isolated from the Peyer's patch following gastrointestinal delivery. Surface vimentin was also expressed on APC populations that could internalize CPMV. These experiments demonstrate that APCs capture CPMV particles in vivo, and that further tuning the interaction with surface vimentin may facilitate increased uptake by APCs and priming of antibody responses. These studies also indicate that CPMV particles likely access the systemic circulation following oral delivery via the Peyer's patch.
Collapse
Affiliation(s)
- Maria J. Gonzalez
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Emily M. Plummer
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Chris S. Rae
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Marianne Manchester
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
19
|
Meshcheryakova YA, Eldarov MA, Migunov AI, Stepanova LA, Repko IA, Kiselev CI, Lomonossoff GP, Skryabin KG. Cowpea mosaic virus chimeric particles bearing the ectodomain of matrix protein 2 (M2E) of the influenza a virus: Production and characterization. Mol Biol 2009. [DOI: 10.1134/s0026893309040219] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Abstract
It has been demonstrated that HIV-1 gp120 resembles several important properties of immunoglobulins allowing it strong influence on the human immune system, especially through induction of the deceptive imprinting and deregulation of the immune network. On the other hand there are many unanswered questions concerning properties and control of the genetically modified viruses and bacteria used as vectors in AIDS vaccines. This situation opens a serious question about the safety of vectored AIDS vaccine and the ethics of their trials in humans.
Collapse
Affiliation(s)
- Veljko Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Yugoslavia.
| | | | | |
Collapse
|
21
|
Marusic C, Vitale A, Pedrazzini E, Donini M, Frigerio L, Bock R, Dix PJ, McCabe MS, Bellucci M, Benvenuto E. Plant-based strategies aimed at expressing HIV antigens and neutralizing antibodies at high levels. Nef as a case study. Transgenic Res 2009; 18:499-512. [PMID: 19169897 PMCID: PMC2758358 DOI: 10.1007/s11248-009-9244-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 01/06/2009] [Indexed: 12/31/2022]
Abstract
The first evidence that plants represent a valid, safe and cost-effective alternative to traditional expression systems for large-scale production of antigens and antibodies was described more than 10 years ago. Since then, considerable improvements have been made to increase the yield of plant-produced proteins. These include the use of signal sequences to target proteins to different cellular compartments, plastid transformation to achieve high transgene dosage, codon usage optimization to boost gene expression, and protein fusions to improve recombinant protein stability and accumulation. Thus, several HIV/SIV antigens and neutralizing anti-HIV antibodies have recently been successfully expressed in plants by stable nuclear or plastid transformation, and by transient expression systems based on plant virus vectors or Agrobacterium-mediated infection. The current article gives an overview of plant expressed HIV antigens and antibodies and provides an account of the use of different strategies aimed at increasing the expression of the accessory multifunctional HIV-1 Nef protein in transgenic plants.
Collapse
Affiliation(s)
- Carla Marusic
- Dipartimento BAS, Sezione Genetica e Genomica Vegetale, ENEA, C.R. Casaccia, via Anguillarese 301, 00123, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Meyers A, Chakauya E, Shephard E, Tanzer FL, Maclean J, Lynch A, Williamson AL, Rybicki EP. Expression of HIV-1 antigens in plants as potential subunit vaccines. BMC Biotechnol 2008; 8:53. [PMID: 18573204 PMCID: PMC2443125 DOI: 10.1186/1472-6750-8-53] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 06/23/2008] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) has infected more than 40 million people worldwide, mainly in sub-Saharan Africa. The high prevalence of HIV-1 subtype C in southern Africa necessitates the development of cheap, effective vaccines. One means of production is the use of plants, for which a number of different techniques have been successfully developed. HIV-1 Pr55Gag is a promising HIV-1 vaccine candidate: we compared the expression of this and a truncated Gag (p17/p24) and the p24 capsid subunit in Nicotiana spp. using transgenic plants and transient expression via Agrobacterium tumefaciens and recombinant tobamovirus vectors. We also investigated the influence of subcellular localisation of recombinant protein to the chloroplast and the endoplasmic reticulum (ER) on protein yield. We partially purified a selected vaccine candidate and tested its stimulation of a humoral and cellular immune response in mice. RESULTS Both transient and transgenic expression of the HIV antigens were successful, although expression of Pr55Gag was low in all systems; however, the Agrobacterium-mediated transient expression of p24 and p17/p24 yielded best, to more than 1 mg p24/kg fresh weight. Chloroplast targeted protein levels were highest in transient and transgenic expression of p24 and p17/p24. The transiently-expressed p17/p24 was not immunogenic in mice as a homologous vaccine, but it significantly boosted a humoral and T cell immune response primed by a gag DNA vaccine, pTHGagC. CONCLUSION Transient agroinfiltration was best for expression of all of the recombinant proteins tested, and p24 and p17/p24 were expressed at much higher levels than Pr55Gag. Our results highlight the usefulness of plastid signal peptides in enhancing the production of recombinant proteins meant for use as vaccines. The p17/p24 protein effectively boosted T cell and humoral responses in mice primed by the DNA vaccine pTHGagC, showing that this plant-produced protein has potential for use as a vaccine.
Collapse
MESH Headings
- AIDS Vaccines/biosynthesis
- AIDS Vaccines/genetics
- Adjuvants, Immunologic/genetics
- Agrobacterium tumefaciens/genetics
- Animals
- Chloroplasts/genetics
- Chloroplasts/metabolism
- Endoplasmic Reticulum/genetics
- Endoplasmic Reticulum/metabolism
- Female
- Gene Expression
- Gene Expression Regulation, Plant
- Genes, gag
- Genetic Vectors
- HIV Antigens/biosynthesis
- HIV Antigens/genetics
- HIV Antigens/immunology
- HIV Infections/immunology
- HIV Seronegativity
- HIV-1/genetics
- Humans
- Mice
- Mice, Inbred BALB C
- Plants, Genetically Modified
- Nicotiana/genetics
- Tobamovirus/genetics
- Transformation, Genetic
- Vaccines, Subunit/biosynthesis
- Vaccines, Subunit/genetics
- gag Gene Products, Human Immunodeficiency Virus/biosynthesis
- gag Gene Products, Human Immunodeficiency Virus/genetics
Collapse
Affiliation(s)
- Ann Meyers
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Ereck Chakauya
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
- CSIR Biosciences, Pretoria 0001, South Africa
| | - Enid Shephard
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- MRC/UCT Liver Research Centre, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Fiona L Tanzer
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - James Maclean
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Alisson Lynch
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| | - Anna-Lise Williamson
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Observatory 7925, South Africa
| | - Edward P Rybicki
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Department of Molecular and Cell Biology, Faculty of Science, University of Cape Town, P. Bag X3 Rondebosch 7701, South Africa
| |
Collapse
|
23
|
Ramírez YJP, Tasciotti E, Gutierrez-Ortega A, Donayre Torres AJ, Olivera Flores MT, Giacca M, Gómez Lim MA. Fruit-specific expression of the human immunodeficiency virus type 1 tat gene in tomato plants and its immunogenic potential in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:685-92. [PMID: 17460112 PMCID: PMC1951073 DOI: 10.1128/cvi.00028-07] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human immunodeficiency virus type 1 (HIV-1) Tat protein is considered a potential candidate vaccine antigen. In an effort to design a strategy for noninvasive vaccination against HIV-1, we developed transgenic tomatoes expressing the Tat protein. Two independent plants testing positive in transgene detection analysis were selected and grown to maturity. Monoclonal antibodies against Tat recognized a protein of the expected size. Interestingly, expression of Tat seemed to be toxic to the plant, as in all cases the fruit exhibited underdeveloped reproductive structures and no seeds. Nine groups of 10 pathogen-free BALB/c male mice were primed either orally, intraperitoneally, or intramuscularly with 10 mg of tomato fruit extract derived from transgenic or wild-type plants and with 10 microg of Tat86 recombinant protein. Mice were immunized at days 0, 14, and 28, and given boosters after 15 weeks; sera were drawn 7 days after each booster, and the antibody titer was determined by enzyme-linked immunosorbent assay. All three immunization approaches induced the development of a strong anti-Tat immunological response, which increased over time. Isotype subclass determination showed the presence of mucosal (immunoglobulin A) immunity soon after the beginning of the oral immunization protocol, and the data were confirmed by the presence of anti-Tat antibodies in fecal pellets and in vaginal washes. We also demonstrated that sera from immunized mice inhibited with high efficiency recombinant Tat-dependent transactivation of the HIV-1 long terminal repeat promoter. This neutralization activity might be relevant for the suppression of extracellular Tat activities, which play an important role in HIV disease development.
Collapse
Affiliation(s)
- Yuri Jorge Peña Ramírez
- Departamento de Ingeniería Genética, Cinvestav Campus Guanajuato, Irapuato, Km 9.6 Libramiento norte, Apartado Postal 629, Irapuato, Gto., México 365002
| | | | | | | | | | | | | |
Collapse
|
24
|
Santi L, Huang Z, Mason H. Virus-like particles production in green plants. Methods 2007; 40:66-76. [PMID: 16997715 PMCID: PMC2677071 DOI: 10.1016/j.ymeth.2006.05.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2005] [Accepted: 05/03/2006] [Indexed: 12/13/2022] Open
Abstract
Viruses-like particles (VLPs), assembled from capsid structural subunits of several different viruses, have found a number of biomedical applications such as vaccines and novel delivery systems for nucleic acids and small molecules. Production of recombinant proteins in different plant systems has been intensely investigated and improved upon in the last two decades. Plant-derived antibodies, vaccines, and microbicides have received great attention and shown immense promise. In the case of mucosal vaccines, orally delivered plant-produced VLPs require minimal processing of the plant tissue, thus offering an inexpensive and safe alternative to more conventional live attenuated and killed virus vaccines. For other applications which require higher level of purification, recent progress in expression levels using plant viral vectors have shown that plants can compete with traditional fermentation systems. In this review, the different methods used in the production of VLPs in green plants are described. Specific examples of expression, assembly, and immunogenicity of several plant-derived VLPs are presented.
Collapse
Affiliation(s)
- Luca Santi
- Biodesign Institute at Arizona State University, Tempe, 852878-5401, USA
| | | | | |
Collapse
|
25
|
Yusibov V, Rabindran S, Commandeur U, Twyman RM, Fischer R. The Potential of Plant Virus Vectors for Vaccine Production. Drugs R D 2006; 7:203-17. [PMID: 16784246 DOI: 10.2165/00126839-200607040-00001] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Plants viruses are versatile vectors that allow the rapid and convenient production of recombinant proteins in plants. Compared with production systems based on transgenic plants, viral vectors are easier to manipulate and recombinant proteins can be produced more quickly and in greater yields. Over the last few years, there has been much interest in the development of plant viruses as vectors for the production of vaccines, either as whole polypeptides or epitopes displayed on the surface of chimeric viral particles. Several viruses have been extensively developed for vaccine production, including tobacco mosaic virus, potato virus X and cowpea mosaic virus. Vaccine candidates have been produced against a range of human and animal diseases, and in many cases have shown immunogenic activity and protection in the face of disease challenge. In this review, we discuss the advantages of plant virus vectors, the development of different viruses as vector systems, and the immunological experiments that have demonstrated the principle of plant virus-derived vaccines.
Collapse
Affiliation(s)
- Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware 19711, USA.
| | | | | | | | | |
Collapse
|
26
|
Dimmock NJ. The complex antigenicity of a small external region of the C-terminal tail of the HIV-1 gp41 envelope protein: a lesson in epitope analysis. Rev Med Virol 2005; 15:365-81. [PMID: 16106492 DOI: 10.1002/rmv.476] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The newly discovered external tail loop within the C-terminal tail of the gp41 transmembrane subunit of the HIV-1 envelope protein comprises approximately 40 residues, and within this are 18-residues ((734)PDRPEGIEEEGGERDRDR(751)) that include three antibody-reactive regions. The antigenicity is complex, and changes according to the biological context of the gp41. It is thus of interest both to the HIV specialist and protein immunologists. The antibody-reactive region, centred on the sequence ERDRD, encompasses three distinct epitopes which are expressed in different combinations on infected cells, wt virions, prefusion virion-cell complexes, and a neutralising antibody escape mutant virion. In addition ERDRD-specific antibodies have one or more antiviral activities, and variously neutralise the infectivity of free virions, neutralise virions already attached to the target cell, reduce the production of infectious progeny, and inhibit the ability of infected cells to fuse with non-infected cells. Antibodies to PDRPEG and IEEE have no apparent antiviral activity even though the footprints of the IEEE- and ERDRD-specific antibodies overlap. This review marshals the available experimental data with the aim of understanding the significance of the gp41 tail loop to the HIV-1 life cycle, and its relevance to potential anti-viral measures. There are lessons here, too, that are relevant to the comprehension of the antigenicity of short protein segments in general.
Collapse
Affiliation(s)
- Nigel J Dimmock
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK.
| |
Collapse
|
27
|
Hollier MJ, Dimmock NJ. The C-terminal tail of the gp41 transmembrane envelope glycoprotein of HIV-1 clades A, B, C, and D may exist in two conformations: an analysis of sequence, structure, and function. Virology 2005; 337:284-96. [PMID: 15913700 PMCID: PMC7111842 DOI: 10.1016/j.virol.2005.04.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Revised: 03/02/2005] [Accepted: 04/11/2005] [Indexed: 11/17/2022]
Abstract
In addition to the major ectodomain, the gp41 transmembrane glycoprotein of HIV-1 is now known to have a minor ectodomain that is part of the long C-terminal tail. Both ectodomains are highly antigenic, carry neutralizing and non-neutralizing epitopes, and are involved in virus-mediated fusion activity. However, data have so far been biologically based, and derived solely from T cell line-adapted (TCLA), B clade viruses. Here we have carried out sequence and theoretically based structural analyses of 357 gp41 C-terminal sequences of mainly primary isolates of HIV-1 clades A, B, C, and D. Data show that all these viruses have the potential to form a tail loop structure (the minor ectodomain) supported by three, β-sheet, membrane-spanning domains (MSDs). This means that the first (N-terminal) tyrosine-based sorting signal of the gp41 tail is situated outside the cell membrane and is non-functional, and that gp41 that reaches the cell surface may be recycled back into the cytoplasm through the activity of the second tyrosine-sorting signal. However, we suggest that only a minority of cell-associated gp41 molecules – those destined for incorporation into virions – has 3 MSDs and the minor ectodomain. Most intracellular gp41 has the conventional single MSD, no minor ectodomain, a functional first tyrosine-based sorting signal, and in line with current thinking is degraded intracellularly. The gp41 structural diversity suggested here can be viewed as an evolutionary strategy to minimize HIV-1 envelope glycoprotein expression on the cell surface, and hence possible cytotoxicity and immune attack on the infected cell.
Collapse
|
28
|
Webster DE, Thomas MC, Pickering R, Whyte A, Dry IB, Gorry PR, Wesselingh SL. Is there a role for plant‐made vaccines in the prevention of HIV/AIDS? Immunol Cell Biol 2005; 83:239-47. [PMID: 15877601 DOI: 10.1111/j.1440-1711.2005.01341.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although educational programs have had some impact, immunization against HIV will be necessary to control the AIDS pandemic. To be effective, vaccination will need to be accessible and affordable, directed against multiple antigens, and delivered in multiple doses. Plant-based vaccines that are heat-stable and easy to produce and administer are suited to this type of strategy. Pilot studies by a number of groups have demonstrated that plant viral expression systems can produce HIV antigens in quantities that are appropriate for use in vaccines. In addition, these plant-made HIV antigens have been shown to be immunogenic. However, given the need for potent cross-clade humoral and T-cell immunity for protection against HIV, and the uncertainty surrounding the efficacy of protein subunit vaccines, it is most likely that plant-made HIV vaccines will find their niche as booster immunizations in prime-boost vaccination schedules.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/immunology
- Acquired Immunodeficiency Syndrome/prevention & control
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- HIV Infections/immunology
- HIV Infections/prevention & control
- Humans
- Immune Tolerance/immunology
- Immunity, Cellular/immunology
- Mice
- Plants, Genetically Modified/genetics
- Plants, Genetically Modified/metabolism
- Vaccines, Edible/administration & dosage
- Vaccines, Edible/biosynthesis
- Vaccines, Edible/genetics
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/biosynthesis
- Vaccines, Synthetic/genetics
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/biosynthesis
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- Diane E Webster
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
29
|
Heap CJ, Reading SA, Dimmock NJ. An antibody specific for the C-terminal tail of the gp41 transmembrane protein of human immunodeficiency virus type 1 mediates post-attachment neutralization, probably through inhibition of virus-cell fusion. J Gen Virol 2005; 86:1499-1507. [PMID: 15831963 DOI: 10.1099/vir.0.80414-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Evidence has been presented which shows that part of the C-terminal tail of the gp41 transmembrane protein of human immunodeficiency virus type 1 (HIV-1) contains a neutralization epitope and is thus exposed on the external surface of the virion. Here, SAR1, a monoclonal antibody, which was stimulated by immunization with a plant virus expressing 60 copies of the GERDRDR sequence from the exposed gp41 tail, and has an unusual pattern of neutralization activity, giving little or no neutralization of free virions, but effecting modest post-attachment neutralization (PAN) of virus bound to target cells was investigated. Here, the properties of PAN were investigated. It was found that PAN could be mediated at 4 or 20 degrees C, but that at 20 degrees C maximum PAN required virus-cell complexes to be incubated for 3 h before addition of antibody. Further PAN appeared stable at 20 degrees C and could be mediated for at least 5 h at this temperature. In contrast, when virus-cell complexes formed at 20 degrees C but then shifted to 37 degrees C for various times before addition of SAR1, PAN was maximal after just 10 min, and was lost after 30 min incubation. Thus, PAN at 37 degrees C is transient and temperature-dependent. Since this scenario recalled the temperature requirements of virus-cell fusion, fusion of HIV-1-infected and non-infected cells was investigated, and it was found that SAR1 inhibited this process by up to 75 %, in a dose-dependent manner. However, antibodies to adjacent epitopes did not inhibit fusion. These data confirm the external location of the SAR1 epitope, implicate the gp41 C-terminal tail in the HIV-1 fusion process for the first time, and suggest that SAR1 mediates PAN by inhibiting virus-mediated fusion.
Collapse
Affiliation(s)
- Caroline J Heap
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Steven A Reading
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Nigel J Dimmock
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
30
|
Piazzolla G, Nuzzaci M, Tortorella C, Panella E, Natilla A, Boscia D, De Stradis A, Piazzolla P, Antonaci S. Immunogenic properties of a chimeric plant virus expressing a hepatitis C virus (HCV)-derived epitope: new prospects for an HCV vaccine. J Clin Immunol 2005; 25:142-52. [PMID: 15821891 DOI: 10.1007/s10875-005-2820-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2004] [Indexed: 01/09/2023]
Abstract
A vaccine against Hepatitis C virus (HCV) is urgently needed due to the unsatisfactory clinical response to current therapies. We evaluated the immunological properties of a chimeric Cucumber mosaic virus (CMV), a plant virus engineered to express on its surface a synthetic peptide derived from many HVR1 sequences of the HCV envelope protein E2 (R9 mimotope). Evidence was obtained that the chimeric R9-CMV elicits a specific humoral response in rabbits. Furthermore, in patients with chronic HCV infection, purified preparations of R9-CMV down-modulated the lymphocyte surface density of CD3 and CD8, and induced a significant release of interferon (IFN)-gamma, interleukin (IL)-12 p70 and IL-15 by lymphomonocyte cultures. Finally, an R9 mimotope-specific CD8 T-cell response, as assessed by intracellular IFN-gamma production, was achieved in the majority of the patients studied. Our results open up new prospects for the development of effective vaccines against HCV infection. Moreover, the wide edible host range of CMV makes the production of an edible vaccine conceivable.
Collapse
Affiliation(s)
- G Piazzolla
- Department of Internal Medicine, Immunology and Infectious Diseases, Section of Internal Medicine, University of Bari, Policlinico, 70124 Bari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rodgers PB, Hamilton WD, Adair JR. The therapeutic potential of plant-derived vaccines and antibodies. Expert Opin Investig Drugs 2005; 8:211-27. [PMID: 15992073 DOI: 10.1517/13543784.8.3.211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The production of recombinant proteins in plants is reviewed with a particular focus on plant-derived vaccines and antibodies for human healthcare. Issues relating to foreign gene expression, such as protein yield, localisation and glycosylation are also considered. Emphasis is placed on reporting progress with preclinical and clinical evaluation of plant-derived vaccines and antibodies. An assessment is made of the likely future direction of research and development in this area.
Collapse
Affiliation(s)
- P B Rodgers
- Axis Genetics plc, Babraham, Cambridge, CB2 4AZ, UK.
| | | | | |
Collapse
|
32
|
Antigen Delivery Systems III: Use of Recombinant Plant Viruses. Mucosal Immunol 2005. [PMCID: PMC7149764 DOI: 10.1016/b978-012491543-5/50063-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
|
33
|
Cheung L, McLain L, Hollier MJ, Reading SA, Dimmock NJ. Part of the C-terminal tail of the envelope gp41 transmembrane glycoprotein of human immunodeficiency virus type 1 is exposed on the surface of infected cells and is involved in virus-mediated cell fusion. J Gen Virol 2005; 86:131-138. [PMID: 15604440 DOI: 10.1099/vir.0.80439-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The C-terminal tail of the gp41 transmembrane glycoprotein of the human immunodeficiency virus type 1 (HIV-1) virion is usually thought to be inside the virion, but it has been shown recently that part of the tail is exposed on the virion exterior. Here, using a panel of antibodies, it was demonstrated that the same part of the tail is exposed on the surface of HIV-1-infected C8166 lymphoblastoid cells and HeLa cells infected with a gp41-expressing vaccinia virus recombinant. Both types of infected cell failed to react with p17 matrix protein-specific IgGs until permeabilized with saponin, confirming the integrity of the plasma membrane. Cell-surface exposure of the gp41 tail was independently demonstrated by inhibition of HIV-1-mediated cell–cell fusion by one of the gp41 tail-specific antibodies. These data also implicate the exposed region of the gp41 C-terminal tail either directly or indirectly in the viral fusion process. Its surface exposure suggests that the gp41 C-terminal tail may be a candidate for immune intervention or chemotherapy of infection.
Collapse
Affiliation(s)
- Linda Cheung
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Lesley McLain
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Mark J Hollier
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Steven A Reading
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Nigel J Dimmock
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
34
|
Negrouk V, Eisner G, Midha S, Lee HI, Bascomb N, Gleba Y. Affinity purification of streptavidin using tobacco mosaic virus particles as purification tags. Anal Biochem 2004; 333:230-5. [PMID: 15450797 DOI: 10.1016/j.ab.2004.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2004] [Indexed: 10/26/2022]
Abstract
A new protein affinity purification system has been developed. Recombinant tobacco mosaic virus (TMV) was used as an affinity matrix for isolation and purification of the given protein of interest. In model experiments, streptavidin-specific heptapeptide sequence TLIAHPQ was inserted into TMV coat protein near the C end. This oligopeptide did not interfere significantly with viral replication, assembly, and movement. Recombinant TMV functioned as an epitope tag recognizing streptavidin in plant protein extracts. Plant protein extracts containing streptavidin were incubated with recombinant TMV virions. Affinity complexes of viral particles with the protein of interest were collected by centrifugation. Recombinant TMV-streptavidin complex was dissociated with 0.2M acetic acid, pH 4.6, and was passed through membrane filter Nanosep 300K by centrifugation. The filtrate contained pure streptavidin. Recombinant TMV was left on the filter. TMV particles collected from the filter could be used for at least two more purification cycles. The streptavidin-specific recombinant TMV system was applied successfully for purification of streptavidin from Streptomyces avidinii. The authors believe that the TMV-based affinity system can also be used for the purification of other proteins.
Collapse
Affiliation(s)
- Valentine Negrouk
- Icon Genetics Inc., Princeton Research Center, 1 Deer Park Drive, Suite C, Monmouth Jct., NJ 08852, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Nicholas BL, Brennan FR, Hamilton WDO, Wakelin D. Effect of priming/booster immunisation protocols on immune response to canine parvovirus peptide induced by vaccination with a chimaeric plant virus construct. Vaccine 2003; 21:2441-7. [PMID: 12744877 DOI: 10.1016/s0264-410x(03)00054-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Expression of a 17-mer peptide sequence from canine parvovirus expressed on cowpea mosaic virus (CPMV) to form chimaeric virus particles (CVPs) creates vaccine antigens that elicit strong anti-peptide immune responses in mice. Systemic (subcutaneous, s.c.) immunisation and boosting with such CVP constructs produces IgG(2a) serum antibody responses, while mucosal (intranasal, i.n.) immunisation and boosting elicits intestinal IgA responses. Combinations of systemic and mucosal routes for priming and boosting immunisations were used to examine their influence on the level, type and location of immune response generated to one of these constructs (CVP-1). In all cases, s.c. administration, whether for immunisation or boosting, generated a Th1-biased response, reflected in a predominantly IgG(2a) serum antibody isotype and secretion of IFN-gamma from in vitro-stimulated lymphocytes. Serum antibody responses were greatest in animals primed and boosted subcutaneously, and least in mucosally vaccinated mice. The i.n. exposure also led to IFN-gamma release from in vitro-stimulated cells, but serum IgG(2a) was significantly elevated only in mice primed intranasally and boosted subcutaneously. Peptide- and wild-type CPMV-specific IgA responses in gut lavage fluid were greatest in animals exposed mucosally and least in those primed and boosted subcutaneously or primed subcutaneously and boosted orally. Lymphocytes from immunised mice proliferated in response to in vitro stimulation with CPMV but not with peptide. The predominant secretion of IFN-gamma from all immunising/boosting combinations indicates that the route of vaccination and challenge does not alter the Th1 bias of the response to CVP constructs. However, optimal serum and intestinal antibody responses were achieved by combining s.c. and i.n. administration.
Collapse
Affiliation(s)
- B L Nicholas
- Department of Agriculture and Horticulture, School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire Nottingham LE12 5RD, UK.
| | | | | | | |
Collapse
|
36
|
Olszewska W, Steward MW. The molecular basis of the antigenic cross-reactivity between measles and cowpea mosaic viruses. Virology 2003; 310:183-9. [PMID: 12788642 DOI: 10.1016/s0042-6822(03)00118-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Two nonrelated viruses, cowpea mosaic virus (wtCPMV) and measles virus (MV), were found to induce cross-reactive antibodies. The nature of this cross-reactivity was studied and results are presented here demonstrating that antiserum raised against wtCPMV reacted with peptide from the fusion (F) protein of MV. Furthermore, the F protein of MV was shown to share an identical conformational B cell epitope with the small subunit of CPMV coat protein. Passive transfer of anti-wtCPMV antibodies into BALB/c mice conferred partial protection against measles virus induced encephalitis. The results are discussed in the context of cross-protection.
Collapse
Affiliation(s)
- Wieslawa Olszewska
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK.
| | | |
Collapse
|
37
|
Porta C, Spall VE, Findlay KC, Gergerich RC, Farrance CE, Lomonossoff GP. Cowpea mosaic virus-based chimaeras. Effects of inserted peptides on the phenotype, host range, and transmissibility of the modified viruses. Virology 2003; 310:50-63. [PMID: 12788630 DOI: 10.1016/s0042-6822(03)00140-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Expression of foreign peptides on the surface of cowpea mosaic virus particles leads to the creation of chimaeras with a variety of phenotypes and yields. Two factors were shown to be particularly significant in determining the properties of a given chimaera: the length of the inserted sequence and its isoelectric point. The deleterious effect of high isoelectric point on the ability of chimeras to produce a systemic infection occurs irrespective of the site of insertion of the peptide. Ultrastructural analysis of tissue infected with chimaeras with different phenotypes showed that all produced particles with a tendency to aggregate, irrespective of the size or isoelectric point of the insert. Host range and transmission studies revealed that the expression of a foreign peptide did not (1) alter the virus host range, (2) increase the rate of transmission by beetles or through seed, or (3) change the insect vector specificity. These findings have implications for both the utility and the biosafety of Cowpea mosaic virus-based chimaeras.
Collapse
|
38
|
Scholthof KBG, Mirkov TE, Scholthof HB. Plant virus gene vectors: biotechnology applications in agriculture and medicine. GENETIC ENGINEERING 2003; 24:67-85. [PMID: 12416301 DOI: 10.1007/978-1-4615-0721-5_4] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Affiliation(s)
- Karen-Beth G Scholthof
- Department of Plant Pathology and Microbiology, Texas A&M University, 2132 TAMU, College Station, TX 77843, USA
| | | | | |
Collapse
|
39
|
Cleveland SM, McLain L, Cheung L, Jones TD, Hollier M, Dimmock NJ. A region of the C-terminal tail of the gp41 envelope glycoprotein of human immunodeficiency virus type 1 contains a neutralizing epitope: evidence for its exposure on the surface of the virion. J Gen Virol 2003; 84:591-602. [PMID: 12604810 DOI: 10.1099/vir.0.18630-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The approximately 150 amino acid C-terminal tail of the gp41 transmembrane glycoprotein of human immunodeficiency virus type 1 (HIV-1) is generally thought to be located inside the virion. However, we show here that both monoclonal IgG and polyclonal epitope-purified IgG specific for the (746)ERDRD(750) epitope that lies within the C-terminal tail neutralized infectious virus. IgG was mapped to the C-terminal tail by its failure to neutralize tail-deleted virus, and by sequencing of antibody-escape mutants. The fact that antibody does not cross lipid membranes, and infectious virus is by definition intact, suggested that ERDRD was exposed on the surface of the virion. This was confirmed by reacting virus and IgG, separating virus and unbound IgG by centrifugation, and showing that virus was neutralized to essentially the same extent as virus that had been in constant contact with antibody. Epitope exposure on virions was independent of temperature and therefore constitutive. Monoclonal antibodies specific to epitopes PDRPEG and IEEE, upstream of ERDRD, also bound to virions, suggesting that they too were located externally. Protease digestion destroyed the ERDRD and PDRPEG epitopes, consistent with their proposed external location. Altogether these data are consistent with part of the C-terminal tail of gp41 being exposed on the outside of the virion. Possible models of the structure of the gp41 tail, taking these observations into account, are discussed.
Collapse
Affiliation(s)
- S Matthew Cleveland
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Lesley McLain
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Linda Cheung
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Tim D Jones
- Axis Genetics, Babraham, Cambridge CB2 4AZ, UK
| | - Mark Hollier
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| | - Nigel J Dimmock
- Department of Biological Sciences, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
40
|
Awram P, Gardner RC, Forster RL, Bellamy AR. The potential of plant viral vectors and transgenic plants for subunit vaccine production. Adv Virus Res 2003; 58:81-124. [PMID: 12205784 DOI: 10.1016/s0065-3527(02)58003-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Peter Awram
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
41
|
Affiliation(s)
- Tianwei Lin
- Department of Molecular Biology, Center for Integrative Molecular Biosciences, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
42
|
Schneemann A, Young MJ. Viral Assembly Using Heterologous Expression Systems And Cell Extracts. VIRUS STRUCTURE 2003; 64:1-36. [PMID: 13677044 DOI: 10.1016/s0065-3233(03)01001-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Anette Schneemann
- Department of Molecular Biology, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
43
|
Wang Q, Lin T, Johnson JE, Finn MG. Natural supramolecular building blocks. Cysteine-added mutants of cowpea mosaic virus. CHEMISTRY & BIOLOGY 2002; 9:813-9. [PMID: 12144925 DOI: 10.1016/s1074-5521(02)00166-7] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Wild-type Cowpea mosaic virus (CPMV) displays no cysteine side chains on the exterior capsid surface and is therefore relatively unreactive with thiol-selective reagents. Four CPMV mutants bearing cysteine residues in one of two exterior positions of the asymmetric unit were created. The mutants were shown to aggregate by virtue of disulfide bond formation in the absence of added reducing agent, bind to metallic gold, and undergo selective reactions at the introduced thiol residues. Controlled aggregation by virtue of biotin-avidin interactions was demonstrated, as was the independent derivatization of reactive lysine and cysteine positions. The ability to introduce such reactivity into a system that can be readily prepared and isolated in gram quantities should open new doors to applications in biochemistry, materials science, and catalysis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
44
|
Nicholas BL, Brennan FR, Martinez-Torrecuadrada JL, Casal JI, Hamilton WD, Wakelin D. Characterization of the immune response to canine parvovirus induced by vaccination with chimaeric plant viruses. Vaccine 2002; 20:2727-34. [PMID: 12034099 DOI: 10.1016/s0264-410x(02)00200-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
NIH mice were vaccinated subcutaneously or intranasally with chimaeric cow pea mosaic virus (CPMV) constructs expressing a 17-mer peptide sequence from canine parvovirus (CPV) as monomers or dimers on the small or large protein surface subunits. Responses to the chimaeric virus particles (CVPs) were compared with those of mice immunized with the native virus or with parvovirus peptide conjugated to keyhole limpet haemocyanin (KLH). The characteristics of the immune response to vaccination were examined by measuring serum and mucosal antibody responses in ELISA, in vitro antigen-induced spleen cell proliferation and cytokine responses. Mice made strong antibody responses to the native plant virus and peptide-specific responses to two of the four CVP constructs tested which were approximately 10-fold lower than responses to native plant virus. The immune response generated by the CVP constructs showed a marked TH1 bias, as determined by a predominantly IgG(2a) isotype peptide-specific antibody response and the release of IFN-gamma but not IL-4 or IL-5 from lymphocytes exposed to antigen in vitro. In comparison, parvovirus peptide conjugated to KLH generated an IgG(1)-biased (TH2) response. These data indicate that the presentation of peptides on viral particles could be used to bias the immune response in favor of a TH1 response.Anti-viral and anti-peptide IgA were detected in intestinal and bronchial lavage fluid of immunized mice, demonstrating that a mucosal immune response to CPV can be generated by systemic and mucosal immunization with CVP vaccines. Serum antibody from both subcutaneously-vaccinated and intranasally-vaccinated mice showed neutralizing activity against CPV in vitro.
Collapse
Affiliation(s)
- Benjamin L Nicholas
- School of Biosciences, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK.
| | | | | | | | | | | |
Collapse
|
45
|
Khor IW, Lin T, Langedijk JPM, Johnson JE, Manchester M. Novel strategy for inhibiting viral entry by use of a cellular receptor-plant virus chimera. J Virol 2002; 76:4412-9. [PMID: 11932408 PMCID: PMC155080 DOI: 10.1128/jvi.76.9.4412-4419.2002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The plant virus cowpea mosaic virus (CPMV) has recently been developed as a biomolecular platform to display heterologous peptide sequences. Such CPMV-peptide chimeras can be easily and inexpensively produced in large quantities from experimentally infected plants. This study utilized the CPMV chimera platform to create an antiviral against measles virus (MV) by displaying a peptide known to inhibit MV infection. This peptide sequence corresponds to a portion of the MV binding site on the human MV receptor CD46. The CPMV-CD46 chimera efficiently inhibited MV infection of HeLa cells in vitro, while wild-type CPMV did not. Furthermore, CPMV-CD46 protected mice from mortality induced by an intracranial challenge with MV. Our results indicate that the inhibitory CD46 peptide expressed on the surface of CPMV retains virus-binding activity and is capable of inhibiting viral entry both in vitro and in vivo. The CD46 peptide presented in the context of CPMV is also up to 100-fold more effective than the soluble CD46 peptide at inhibiting MV infection in vitro. To our knowledge, this study represents the first utilization of a plant virus chimera as an antiviral agent.
Collapse
Affiliation(s)
- Ing Wei Khor
- Department of Cell Biology, Center for Integrative Molecular Biosciences, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
46
|
Pogue GP, Lindbo JA, Garger SJ, Fitzmaurice WP. Making an ally from an enemy: plant virology and the new agriculture. ANNUAL REVIEW OF PHYTOPATHOLOGY 2002; 40:45-74. [PMID: 12147754 DOI: 10.1146/annurev.phyto.40.021102.150133] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Historically, the study of plant viruses has contributed greatly to the elucidation of eukaryotic biology. Recently, concurrent with the development of viruses into expression vectors, the biotechnology industry has developed an increasing number of disease therapies utilizing recombinant proteins. Plant virus vectors are viewed as a viable option for recombinant protein production. Employing pathogens in the process of creating added value to agriculture is, in effect, making an ally from an enemy. This review discusses the development and use of viruses as expression vectors, with special emphasis on (+) strand RNA virus systems. Further, the use of virus expression vectors in large-scale agricultural settings to produce recombinant proteins is described, and the technical challenges that need to be addressed by agriculturists and molecular virologists to fully realize the potential of this latest evolution of plant science are outlined.
Collapse
Affiliation(s)
- Gregory P Pogue
- Large Scale Biology Corporation, 3333 Vaca Valley Pkwy, Vacaville, CA 95688, USA.
| | | | | | | |
Collapse
|
47
|
Abstract
Essentially all of the currently available vaccines are based on the use of inactivated or live-attenuated pathogens. However, these vaccines have several shortcomings, such as difficulties of in vitro culturing, biohazard risks, as well as loss of efficacy due to the genetic variations seen in many viruses. These problems may potentially be solved by immunising with epitope-based vaccines consisting of rationally designed protective epitopes, appropriately presented and easy to deliver, which are capable of stimulating effective B-cell, T-cell and cytotoxic immune responses whilst avoiding potentially hazardous and undesirable effects. Furthermore, the use of a mixture of defined epitopes could lead to an effective broad range immune response which has the potential to overcome both strain specificity of the pathogen and the MHC restriction of the host. Epitope-based vaccines can be designed to involve the use of synthetic materials that can be available in unlimited quantities and posing no biohazard. Other approaches include the use of naked DNA or recombinant viruses or bacteria expressing the epitopes. An important objective in the development of such vaccines is that they should be effective when delivered via the mucosal route and effective in the presence of maternal antibodies. In this review, we present examples of the use of various epitope-based vaccine constructs, focussing particularly upon their intranasal delivery to the immune system.
Collapse
Affiliation(s)
- W Olszewska
- The Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, WC1E 7HT, London, UK
| | | |
Collapse
|
48
|
Abstract
Agrobacterium tumefaciens mediated gene transfer into the plant genome laid the groundwork for new procedures aimed at crop improvement, including resistance to pathogens, increased product yield, modified oil content, and resistance to environmental stress conditions. New developments in molecular plant virology have led to the generation of plant-based systems for transient expression of foreign sequences using plant virus vectors. In the last decade both transgenic plants and plant virus vectors have been used increasingly to produce a wide range of biomedical reagents, including vaccine antigens, in a safe and economically feasible manner. These new plant-based technologies have enormous potential for a variety of applications, including the oral delivery of vaccine antigens.
Collapse
Affiliation(s)
- H Koprowski
- Biotechnology Foundation Laboratories at Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | |
Collapse
|
49
|
Brennan FR, Jones TD, Hamilton WD. Cowpea mosaic virus as a vaccine carrier of heterologous antigens. Mol Biotechnol 2001; 17:15-26. [PMID: 11280928 DOI: 10.1385/mb:17:1:15] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The plant virus, cowpea mosaic virus (CPMV), has been developed as an expression and presentation system to display antigenic epitopes derived from a number of vaccine targets including infectious disease agents and tumors. These chimeric virus particles (CVPs) could represent a cost-effective and safe alternative to live replicating virus and bacterial vaccines. A number of CVPs have now been generated and their immunogenicity examined in a number of animal species. This review details the humoral and cellular immune responses generated by these CVPs following both parenteral and mucosal delivery and highlights the potential of CVPs to elicit protective immunity from both viral and bacterial infection.
Collapse
Affiliation(s)
- F R Brennan
- Proteom Ltd., Babraham Hall, Babrahm, Cambridge, CB2 4AT
| | | | | |
Collapse
|
50
|
|