Next Article in Journal
Structural Investigation of Diclofenac Binding to Ovine, Caprine, and Leporine Serum Albumins
Next Article in Special Issue
Sambucus ebulus (Elderberry) Fruits Modulate Inflammation and Complement System Activity in Humans
Previous Article in Journal
Developing Novel Experimental Models of m-TORopathic Epilepsy and Related Neuropathologies: Translational Insights from Zebrafish
Previous Article in Special Issue
High-Yield Production of Chimeric Hepatitis E Virus-Like Particles Bearing the M2e Influenza Epitope and Receptor Binding Domain of SARS-CoV-2 in Plants Using Viral Vectors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Plant Viruses and Molecular Farming: How Beneficial They Might Be for Human and Animal Health?

1
Department of Plant Physiology and Molecular Biology, University of Plovdiv, 4000 Plovdiv, Bulgaria
2
Department of Technology Transfer and IP Management, Center of Plant Systems Biology and Biotechnology, 4000 Plovdiv, Bulgaria
3
Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan
4
Institute of Molecular Biology and Biotechnologies, 4108 Markovo, Bulgaria
5
Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
6
Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3T 2M9, Canada
7
Faculty of Sciences, Brigham Young University–Hawaii, Laie, HI 96762, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(2), 1533; https://doi.org/10.3390/ijms24021533
Submission received: 18 December 2022 / Revised: 4 January 2023 / Accepted: 6 January 2023 / Published: 12 January 2023
(This article belongs to the Special Issue Plant-Derived Biologics and Other High-Value Compounds)

Abstract

:
Plant viruses have traditionally been studied as pathogens in the context of understanding the molecular and cellular mechanisms of a particular disease affecting crops. In recent years, viruses have emerged as a new alternative for producing biological nanomaterials and chimeric vaccines. Plant viruses were also used to generate highly efficient expression vectors, revolutionizing plant molecular farming (PMF). Several biological products, including recombinant vaccines, monoclonal antibodies, diagnostic reagents, and other pharmaceutical products produced in plants, have passed their clinical trials and are in their market implementation stage. PMF offers opportunities for fast, adaptive, and low-cost technology to meet ever-growing and critical global health needs. In this review, we summarized the advancements in the virus-like particles-based (VLPs-based) nanotechnologies and the role they played in the production of advanced vaccines, drugs, diagnostic bio-nanomaterials, and other bioactive cargos. We also highlighted various applications and advantages plant-produced vaccines have and their relevance for treating human and animal illnesses. Furthermore, we summarized the plant-based biologics that have passed through clinical trials, the unique challenges they faced, and the challenges they will face to qualify, become available, and succeed on the market.

1. Introduction

Since plant viruses were discovered in the late nineteenth century, their impact has changed from harmful pathogens to useful molecular machines with applications in plant molecular farming (PMF) and bio-nanotechnology. Although public perception of a “virus” is often associated with harm, viruses can easily be reorganized into environmentally friendly and safe structures [1,2,3]. The expansion of the overall knowledge and understanding of viral genomes, architecture, and biophysical properties has allowed the use of plant viruses as vectors for recombinant protein expression and the production of virus-based nanoparticles (VNPs) [4]. VLPs are self-assembled, naturally occurring nanomaterials that structurally and morphologically resemble the 3-dimensional structures of virions, but without the viral genome. VLPs modified to carry additional useful payload are defined as VNPs. Some of the many biological applications of VNPs, which are generated from the capsid structural components of various viruses, include vaccine production and innovative delivery mechanisms for therapeutic compounds, diagnostic reagents, enzymes, pesticides, immunogenic peptides, and many others [5].
In the present review, we will cover the latest developments in PMF and the usefulness of plant viruses in the production of bio-nanomaterials for human and animal health. We will also discuss virus-like nanoparticle-based technologies for vaccine production in plants and the possibility of using unpurified materials for oral treatment. Furthermore, we highlight the usefulness of plant-derived VNPs as templates, carriers, containers, and scaffolds for drug delivery and application in nanomedicine.

Historical Review of Plant Molecular Farming

Over the past three decades, several plant systems have been used to produce recombinant proteins. Much interest has been paid to plant-derived antibodies, vaccines, enzymes, microbicides, and viral protein nanocages (Figure 1).
Plant molecular farming was born in 1980 when the marker gene for β-glucuronidase (GUS) was successfully transformed in higher plants [6]. Later, β-glucuronidase became one of the successful commercial products of plant molecular farming, together with two other industrial enzymes, avidin and trypsin [7,8,9]. In 1986, human growth hormone (HGH) was produced in transgenic tobacco and sunflowers [10]. In 1988, the first production of human antibodies in plants was reported [11]. The concept of using plants for the production of vaccines, more specifically, edible vaccines, was introduced by Dr. Charles Arntzen in the 1990s [12]. Studies were focused on this topic for many years [13,14,15] until the incident of seed contamination with a transgene expressing a bovine vaccine candidate [16,17]. The new trends in plant molecular farming are to use plants in controlled conditions to produce recombinant products that can either be injected or used orally [18,19,20,21,22].
The initial plant-derived biopharmaceuticals were expressed in stable transgenic plants. This technological approach has three main disadvantages: a long developmental period, low yield, and public concerns about GMO plants. Transient expressions via plant expression virus vectors offer a way to avoid these limitations [23].
Although the production of recombinant proteins in plants by stable gene integration and expression has some limitations, the initial commercialized therapeutic recombinant proteins were produced by this technology. In 2006, stably transformed N. benthamiana cell cultures were used for the production of the first USDA-approved injectable vaccine against the Newcastle disease virus for poultry [24]. In 2012, the FDA approved plant-derived recombinant human β-glucocerebrosidase (marketed as Elelyso®) for the treatment of Gaucher’s disease type 1. The recombinant human β-glucocerebrosidase enzyme was produced in a stably transformed carrot cell suspension culture by Protalix Biotherapeutics Inc. [25] and licensed to Pfizer. In 2022, Health Canada approved the first plant-derived SARS-CoV-2 vaccine, Covifenz [26]. The strategy included transient expression in N. benthamiana of the modified recombinant SARS-CoV-2 S protein with stabilizing point mutations. Medicago Inc. successfully produced and purified modified SARS-CoV-2 S VLPs built from the S protein. Using the same technology, Medicago Inc. successfully produced a vaccine against seasonal influenza [27]. Currently, there are few examples of commercialized plant molecular farming products, and plants cannot replace industrial expression systems. However, PMF has been proven cost-effective and valuable in the production of vaccines, antibodies, enzymes, and biological VNPs under conditions of urgency, such as emerging pandemic viruses or for treating rare diseases.

2. Important Plant Viruses for Plant Biotechnology

Approximately 900 plant viruses, identified mainly in crops, are known [28]. Several of them, such as the tobacco mosaic virus (TMV), potato virus X (PVX), cowpea mosaic virus (CPMV), cowpea chlorotic mottle virus (CCMV), brome mosaic virus (BMV), cucumber mosaic virus (CMV), plum pox virus (PPV), alfalfa mosaic virus (AIMV), papaya mosaic virus (PapMV), tomato bushy stunt virus (TBSV), and many others, are utilized in biotechnology and plant viral-based nanotechnology research and industry (Figure 2) [29,30].
Plant viruses have very simple (rod-shaped, spherical, quasi-spherical, or filamentous) structures, consisting of multiple copies of one or a few capsid protein subunits, forming a protein coat around the viral genome. The capsid proteins have the ability to self-assemble, which offers the opportunity to generate a great variety of natural bio-nanomaterials. A better understanding of the structure and biophysical characteristics of plant viruses is vital for developing and producing VNPs, which can be used as nanoparticles for the surface presentation of antigens for recombinant VNP vaccines or as nanocages for “cargo” delivery. VNPs are naturally occurring biological entities, making them both biocompatible and biodegradable. Furthermore, VNPs can be targeted to particular tissue or cells. Finally, the production and purification of VNPs from plants are rapid, cost-effective, and environmentally safe (compared with conventional nanoparticles) and can easily be scaled up depending on demand [31]. There are two main strategies for VNPs production in plants: using a virus infection, allowing the cultivation of plant viruses in their natural host, or the biotechnology approach, using transient expression of the viral genes encoding the capsid proteins in heterologous expression systems [32,33,34].

2.1. Tobacco Mosaic Virus (TMV)

TMV is a member of the genus Tobamovirus in the family of Virgaviridae. TMV has a rod-shaped virion with a diameter of 18 nm, a modal length of 300 nm with a central channel in width, and the viral RNA intercalated between the coat protein turns [35]. TMV coat protein (CP) could self-assemble into the form of a rigid helical RNA-free tube [36]. Tobacco mosaic virus can be easily cultivated in plants, and the yields can be very high, ~4 g/kg wet weight of tobacco. The purified TMV rod-shaped virions can be disassembled in vitro into protein subunits and RNA [37,38]. When nucleic acid is absent, the coat protein can self-assemble into several types of VNPs. The polymerization of the TMV coat protein is temperature and concentration-dependent [39,40]. TMV VNPs are stable and can be used as scaffolds for chemical modifications, coating with biologically active peptides, or loading with drugs [17,41,42,43,44,45,46]. These modifications will be discussed in greater detail in Section 6 of this review. TMV has been widely used as a full-vector and also as a deconstructed virus vector for recombinant protein expression [47].

2.2. Cowpea Mosaic Virus (CPMV)

CPMV belongs to the genus Comovirus in the Comoviridae family. It is a non-enveloped, icosahedral virus with nanoscale dimensions (30 nm). The CPMV protein shell is comprised of 60 copies of the large (L) coat proteins with two domains and 60 copies of the small (S) coat proteins with one domain [48]. The three domains together form the asymmetric unit of the CPMV capsid, with 2 nm channels enabling the exchange of molecules from the exterior to the interior. The internal viral cavity encapsulates two single-stranded RNA molecules [49,50].
This CPMV virus has been widely used in bio-nanotechnology because its structure and particle architecture have been well studied, and the genome can easily be manipulated [51]. Virus particles, which contain viral genomic RNAs, can be produced rapidly in high yield (1–2 g/kg) through the infection of plants [48]. These viral particles have been used for selective attachment of various moieties [51,52,53,54,55] and to display immunogenic epitopes on their surface [56,57]. The drawback is that the CPMV particles containing genomic RNAs raise biosafety and regulatory concerns. They are also ineffective drug containers because they are hard to load with foreign materials such as therapeutic agents or heterologous RNA molecules. Prof. Lomonossoff’s laboratory addressed these problems by developing RNA-free, empty virus-like particles (eVLPs) based on the transient expression of VP60 (precursor of L and S coat proteins) along with the 24K viral proteinase in Nicotiana benthamiana [58,59,60,61]. In addition, the eVLPs are very stable under various conditions, which extends their bio-nanotechnology application range [2,61,62,63]. Lomonossoff’s lab further improved CPMV viral particle packaging, allowing the production of VLPs that can pack up to 6 kb of artificial RNA [64]. Furthermore, CPMV was used for the expression of recombinant proteins by applying full-virus and deconstructed virus strategies [65].

2.3. Cowpea Chlorotic Mottle Virus (CCMV)

CCMV belongs to the Bromoviridae family. The CCMV capsid is composed of 180 identical capsid proteins (CP) that form a ~28 nm diameter icosahedral shell and an 18 nm diameter inner cavity [66]. Native CCMV is stable at pH 5.0. The native CCMV virion can be disassembled, and the RNA genome can be removed by centrifugation under high salt concentrations at neutral pH [67]. In vitro, at pH 5.0, the CCMV capsid proteins can be self-assembled into empty nanoscale structures [68]. CCMV is one of the first viruses used in bio-nanotechnology as a tool for developing drug-delivery vehicles due to its well-studied structure, simple capsid with good biocompatibility, and low toxicity [69]. VNPs based on CCMV have been used to encapsulate guest molecules, such as negatively charged polymers, enzymes, and organic aggregates [70]. A wide variety of ligands, such as small peptides, biotin, fluorescent dyes, organometallic photosensitizers, and intact IgG antibodies, can be attached on the outer surface of the capsid [71,72,73,74].

2.4. Brome Mosaic Virus (BMV)

BMV, a member of the family Bromoviridae, is very similar to CCMV [75]. The native virion has a diameter of 28 nm with T = 3 icosahedral symmetry, and it is built of 180 identical capsid proteins with different conformations (A, B, and C) [76,77]. The A conformation of the coat protein forms pentameric capsomers with small 0.5 nm pores. The B and C conformations of the capsid protein form hexameric capsomer with 0.6 nm pores [76]. The stability of the capsid depends on the interaction between the positively charged inner cavity and the negatively charged encapsulated RNA [78]. BMV, similarly to CCMV, is widely used in bio-nanotechnology due to its well-examined structure and biophysical properties.

3. Plant Viruses as a Source of Vectors for Transient Expression

The use of plant viruses as vectors for delivering foreign genes into plants began in the 1980s [79,80]. Understanding the viral genomes’ structure and the strategies plant viruses use for their gene expression is essential for constructing and improving virus-based expression systems. The plant virus-based expression systems possess many advantages, including rapid and high-level gene expression and a reduced risk for contamination with transgenes. Cauliflower mosaic virus (CaMV) was used to construct the first plant virus expression systems. The limitation of these expression systems was their small capacity to integrate foreign genes after removing large regions of their viral DNA [81].
A major breakthrough in the development of plant virus-based expression systems was the use of single-stranded RNA viruses and Agrobacterium tumefaciens to insert viral genes into the plant cell [82]. The potential of many plant viruses to become a source of useful vectors for plant genome engineering has been investigated (Table 1). The most well-known of these are TMV [83], PVX [84], CPMV [85], tobacco rattle virus (TRV) [86], barley stripe mosaic virus (BSMV) [87], and many others.
The plant virus-based vectors can be divided into first-generation (gene substitution vectors, gene insertion vectors, and gene display vectors) and second-generation (deconstructed vector systems) expression vectors.
First-generation expression vectors use the entire viral genome. The gene of interest is expressed as a substitute coat protein, fused to a coat protein, or separated into another open reading frame (ORF) under the control of a potent subgenomic promoter [97]. The use of gene replacement vectors is limited because of their inability to induce systemic infections and cell-to-cell movement due to the substitution of endogenous viral sequences of coat proteins with heterologous genes of interest [47]. Gene insertion vectors contain complete functional viruses with additional ORFs for recombinant proteins. These vectors can support cell-to-cell movement and induce systemic infection. TMV and PVX are the two viruses most commonly used to create gene insertion vectors [98,99]. Gene fusion vectors are constructed by fusing a recombinant peptide of interest with a coat protein. The early works with genetically modified coat proteins of several plant viruses supporting heterologous peptide fusions and exposing them on the virion surface have been reviewed [100,101].
With the introduction of second-generation virus vectors, transient expression effectiveness has improved drastically. These vectors contain deconstructed viral genomes, keeping only essential and some additional components needed for effective protein expression [83,102]. The constructed modular system consists of the main viral components separated into distinct parts and inserted into binary vectors within Agrobacterium tumefacience. By shortening the vector size, the length of the desired gene is extended. Since the genes essential for virus transport and assembly have been removed, deconstructed virus vectors are delivered to plants primarily via vacuum agroinfiltration, leading to simultaneous expression of the protein of interest in all plant tissues with an optimal harvest yield [103]. In addition, the co-expression of vectors containing different structural parts can result in the construction of macromolecular protein structures such as empty virus-like particles (eVLPs) [59].
At present, numerous recombinant proteins used as human and veterinary pharmaceuticals have been produced using plant-based transient expression systems such as magnICON® [104], potato virus X-based vectors [105,106], and the cowpea mosaic virus-based vector pEAQ-HT [107,108,109,110,111]. These vectors made it possible for recombinant protein expression levels to reach more than 40% of total soluble protein [112].

4. Functionalization Methods of VNPs

Plant viruses are intriguing because their coat proteins may construct robust, biodegradable delivery systems for a wide range of chemicals by self-assembling around natural and synthetic molecules of interest. Viruses employ various strategies to self-assemble coat proteins around cargo moieties. For several viral platforms, encapsulating foreign materials is dependent on the interaction of coat proteins with a particular sequence of the viral RNA (i.e., red clover necrotic mosaic virus (RCNMV)) or a negatively charged substance (i.e., BMV, CCMV, Hibiscus chlorotic virus (HCRSV)) to replace the negatively charged RNA [113]. Disassembly and reassembly strategies can be used to release the viral genome and encapsulate new cargo (Figure 3).
The viral genome can be released from the capsid by altering the in vitro buffer conditions, pH, and ionic strength [114,115]. Using buffer exchange techniques, viral capsids are disassembled and then reassembled to enclose the target payload. To give just one example, CCMV disassembles and releases its core RNA at physiological pH or high ionic strength (1M) but reassembles in a buffer with a pH between 3 and 6 and low ionic strength (0.1 M) [116]. Like the creation of micelles, reassembly can be induced by combining the cargo with coat proteins at a concentration higher than the critical assembly concentration of the VLPs. Cargo encapsulation success is influenced by a variety of parameters, including, but not limited to, cargo size and surface charge, electrostatic interactions, hydrophobicity, and hydrophilicity [117].
However, with the simple co-expression of necessary viral proteins in plant cells, certain virus particles (CPMV) may form hollow capsids in the absence of genetic/external material. Molecules that are small enough and can pass through the pores of the viral capsid can also be encapsulated [118]. However, this mechanism depends on the size of the molecules and the diameter of the pores, meaning that various viruses will have different packing abilities. For some viruses, pore development can be stimulated by removing divalent ions from inside the capsid [119,120,121,122]. This feature of RCNMV was illustrated by Lommel et al. [123] by removing calcium and magnesium ions from the solvent using chelators, which resulted in the development of channels with a diameter of 11–13 Å. Re-addition of the ions closed the created channels.
By manipulating the buffer conditions, CCMV may be produced in an open and expanded configuration, creating holes of 2 nm that are suitable for cargo loading. Using a process called “gating,” the cargo is secured within by switching back to a closed and condensed shape [124]. This diffusion method has also been used with the artichoke mottled crinkle virus (AMCV) [125] and Johnson grass chlorotic stripe mosaic virus (JCSMV) [126] (Figure 3).
Other VNP-based strategies utilize the expression of fusion proteins on the exterior surface of VNP particles [127]. These chimeric proteins have been generated by fusing the coding sequences of a coat protein and a protein of interest (immunogenic peptides, receptors, tags, linkers, and others). While these gene-altering approaches generate new surface proteins, they do not disturb self-assembling during the expression in heterologous systems. Gene-engineered coat proteins are suitable for the bio-conjugation method for functionalizing the capsid. Bio-conjugation is joining biomolecules to other molecules forming a new complex of hybrid material. Targets for bio-conjugation are surface groups of reactive amino acid side chains, most commonly that of lysine (Lys), cysteine (Cys), or tyrosine (Tyr) aa, or linked carbohydrates of capsid proteins [128]. Bio-conjugation was used to functionalize VNPs for applications in drug delivery, imaging, theranostic, and immunotherapy [2,30,129,130].

5. Application of Plant VNPs for Delivering Bioactive Cargos

5.1. Anticancer Drugs

While conventional cancer therapies such as surgery, radiotherapy, and chemotherapy have a good track record of effectiveness, they also have some significant drawbacks. Although effective, conventional chemotherapy often has devastating side effects on healthy cells. The therapeutic outcomes of chemotherapy would be greatly improved if the chemotherapeutic agents were delivered in a targeted manner inside the tumor tissues. Such specific delivery of chemotherapeutics into the tumor cells would lead to a maximum therapeutic site with minimum toxic side effects. Combining conventional chemotherapy with biological treatments can not only deliver drugs in a highly targeted manner but can also boost antitumor immunity [131].
VNPs and VLPs have been used effectively in the delivery of the anticancer medication doxorubicin (DOX). DOX has been effectively delivered using TMV- and PVX-derived VLPs and VNPs. VNPs have shown a lot of promise since the simple adsorption of DOX on their surface, which effectively suppressed tumor development and progression [2]. RCNMV was coupled to a CD46-targeting polypeptide and packed it with DOX via passive diffusion of DOX through the constructed channels during the capsid’s “open” configuration. In vitro, HeLa cells were highly sensitive to the cytotoxicity of RCNMV NPs that had been targeted and loaded with DOX [132]. Peptides with targeted therapeutic effects against different malignancies can be administered through TMV. Phenanthriplatinn, reported as PhenPt-TMV, is an anticancer medication loaded into a hollow TMV vehicle. PhenPt-TMV is an example of a stimuli-responsive system in which the acidic microenvironment of the tumor drives the drug release. This is consistent with the findings of Tian et al., who showed that a Transacting Activation Transduction peptide conjugated to the TMV surface improved both internalization and the virus’s propensity to evade degradation in the endo- and lysosomal systems [133].
Platinum-derived medicines are used in half of the chemotherapy regimens. Targeted delivery of platinum-based anticancer drugs through plant virus VNPs and the effectiveness of TMV in transporting the platinum-based therapeutics cisplatin and phenanthriplatin have been reported. Both charge-driven interactions and the synthesis of stable covalent adducts were used to deliver the medicines into the TMV VNP cavity. It has been demonstrated in in vitro systems employing HepG2 and MCF-7 cancer cell lines that a TMV-based drug delivery system enables superior, targeted cytotoxicity and greater ease of absorption by cancer cells [134]. Mitoxanthrone, a topoisomerase II inhibitor, is another anticancer medication that has been reported to be encapsulated by TMV. Although direct administration of mitoxanthrone can have devastating cardiac side effects, VNPs carrying mitoxanthrone have been shown to significantly reduce tumor development in mouse cancer models [135]. Another example of a VNP-based anticancer drug delivery approach is the TMV VNPs specific for non-Hodgkin’s lymphoma that have the anticancer agent valine-citrulline monomethyl auristatin E (vcMMAE) attached to their surface. The efficacy of this approach was demonstrated in vitro against the Karpas 299 non-Hodgkin’s lymphoma cell line [45]. PVX displaying TNF related apoptosis induces ligand (TRAIL) stimulated death receptor recruitment and activation in HCC-38 primary ductal carcinoma, BT-549 ductal carcinoma, and MDA-MB-231 breast cancer cell lines [136].

5.2. Imaging Agents

VNPs can be developed for smart tissue-specific targeting of imaging agents. VNPs are nanoparticles that possess multivalent functional side chains of exposed amino acids that can be used as anchoring points for imaging modalities. Imaging molecules can be attached to the outside surface or inside cavity of VNPs, through genetic engineering, by adding bioluminescent protein sequences to specific epitopes on the capsid, using bioconjugation chemistry, or infusion method.
Numerous in vivo imaging modalities are now in development. The most used in clinical practice are computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). Magnetic resonance imaging and PET are used more frequently for cardiovascular applications and provide improved soft tissue imaging compared to CT. Both MRI and PET need the use of image-contrast agents—expensive isotopes such as 18F, 11C, or 15O in clinical applications and metals such as Gadolinium (GdIII) or iron for MRI—to achieve high sensitivity [137]. Using VNPs as frameworks for creating multivalent imaging sensors has several benefits: they can easily be functionalized through their surfaces’ exposed amino acid reactive groups to have better sensitivity and specificity, and possibly quicker response in medical analyses.
The indocyanine green near-infrared imaging agent was encapsulated using VLPs based on BMV. The BMV capsid was doped with indocyanine green and used to image human bronchial epithelial cells with preserved cellular bioavailability over 3 h of incubation [138].
The fusion of the green fluorescent protein (GFP) or the red fluorescent protein, mCherry, with the N-termini of the PVX viral coat protein demonstrated that PVX can be used as an optical imaging agent in human cancer cells and mouse model studies [139,140]. TMV nanoparticles were modified to carry the oligopeptides (VHPKQHRAEEA-Lys(PEG7-N3)-NH2), which specifically recognized vascular cell adhesion molecule (VCAM)-1 on endothelial cells at atherosclerotic plaques. These modified TMV VLPs were loaded with near-infrared dyes and (Gd(DOTA)) for MIR. Using submicromolar dosages (400 times lower than the average therapeutic dose), the modified VLPs successfully recognized atherosclerotic plaques [141].
Labeling CPMV nanoparticles with the fluorescence fluorophore, AlexaFluor 555, at a ratio of 120 dyes per particle allowed scientists to image the vascular system in living animals. The resulting modified particle is brighter than orange-fluorescent microspheres of the same dimension and much brighter than fluorescent dextran. The entire circulatory system of a developing mouse was illuminated after the injection of labeled CPMV nanoparticles into the capillaries of the yolk sac. The injection of CPMV nanoparticles into a living chick embryo visualized the whole vascular system of the animal and allowed the observation of its vascular development over several days [142]. Fluorescently labeled CPMV particles, injected into a chick’s chorioallantoic membrane, visualized the developing vasculature of a tumor for at least 2 days. In addition, sequential inoculations with various fluorophore-enabled CPMV can visualize the developmental stages of a newly formed vasculature [142].
T2 imaging has also documented the development of viral scaffolds. The low signal strength of T2 shortening agents, in contrast to the vivid signal produced by T1 shortening agents, is a hallmark of these compounds. Nanoparticles consisting of iron oxide have been incorporated with VNPs as contrast agents. To accomplish imaging in the biomedical field, iron oxide NPs were encapsulated inside VLPs generated from BMV [143].

5.3. Nucleic Acid

Nucleic acid-based therapeutics are a promising technology for treating several diseases and turned out to be the fastest and most effective way to create vaccines against emerging diseases. There are several categories of nucleic acid therapeutics: DNA/RNA coding functional proteins could overcome loss-of-function mutations; messenger RNA (mRNA) could induce translation and production of proteins; small interfering RNAs (siRNAs) and antisense oligonucleotides could induce gene silencing; and microRNAs (miRNAs) could modulate gene expression patterns [144,145]. siRNAs and miRNAs have a tremendous impact on cancer therapy and gene regulation. siRNAs became the first FDA-approved nucleic acid drugs [146]. Several delivery platforms have been used for nuclei acid therapeutics. They can be characterized into two groups: viral and non-viral nucleic acid delivery systems. Common vectors utilized for gene therapy are based on mammalian viruses, such as adenoviruses, adeno-associated viruses, lentiviruses, and retroviruses [147]. Non-viral delivery systems (liposomes, dendrimers, lipid nanoparticles (LNPs), and polymeric nanoparticles) are widely used due to their safety, easy modification, cell internalization, and allowing controlled releasing of the encapsulated therapeutics [148,149]. Viral and non-viral delivery systems have some limitations. Non-viral systems generally are more unstable, less accurate, and less effective than viral systems. The disadvantages of the viral vectors include the possibility of gene integration and pre-existing or innate immunogenicity [150,151,152]. Plant virus-based VNPs are easily reprogrammable nanoparticles with desired biological, chemical, and physical properties for effective nucleic acid delivery [153,154]. Using the disassembling/reassembling method, Steinmetz lab demonstrated that CCMV can encapsulate CpG oligodeoxynucleotides (ODNs). The direct injection of such particles into a tumor induced a robust antitumor response by increasing the phagocytic activity of macrophages [155].
Recently, CPMV coat protein was used to encapsulate and stabilize RNA molecules that can act as positive qRT-PCR controls for diagnosing viral diseases, including COVID-19 [156]. Such RNAs could either be used to induce gene silencing or act as mRNA to transiently produce proteins within the transfected cells.
CCMV VNPs were loaded with heterologous RNA from mammalian Sindbis virus for delivering nucleic acids into mammalian cells [157]. Others proved that CCMV-based VNPs can transfect and deliver heterologous genes for translation into mammalian cells [158]. The CCMV coat protein was used to encapsulate and deliver siRNAs into mammalian cells for gene knockdown [159]. Furthermore, CCMV was used as a platform for delivery of self-amplifying mRNA vaccine [160].
TMV was used to create a probe containing scrambled EBOV RNA sequences that can be used as a positive control in Ebola RT-PCR diagnostics assay [161]. TMV was also used for direct gene delivery and expression into HeLa cells with proven expression of TMV coat protein [162].

5.4. Non-Biological Synthetic Nanoparticles

Material science and medicine can greatly benefit from chemically synthesized NPs (metallic, magnetic, and semiconducting polymers). The ideal NPs for human and animal use should be safe, biocompatible, biodegradable, and tailored to the area in need of improvement. Covering these man-made NPs with proteins found on the outside of plant virus coats can make them more biocompatible, improve their delivery, and get rid of them faster.
The red clover necrotic mosaic virus (RCNMV) has been reported to encapsulate tiny gold, cobalt ferric oxide, and cadmium selenide nanoparticles (varying in size from 3 to 15 nm) [163]. The oligonucleotides mimicking the DNA framework and origin of assembly (OAS) were used to join the metal nanoparticles. The assembly of VLPs is initiated by OAS when interacting with viral coat proteins. Loo et al. introduced RCNMV RNA-1 using synthetic oligonucleotides. This led to the formation of OAS and the assembly of RCNMV CPs around metal nanoparticles, leading to the formation of VNPs, as shown in Figure 4.
Using polyethylene glycol (PEG)ylated phospholipids containing terminal carboxyl groups, Huang et al. documented how iron oxide NPs of about 20.1 nm in size can create VNPs. The capsid proteins of the Brome mosaic virus (BMV) were able to wrap metal NPs in a way that made the VNPs’ size 41 nm [143]. This research also revealed that BMV capsids can be used to incorporate foreign material into plant cells. Using VNPs as MRI probes to track vital plant functions is another area pioneered by Huang’s team [143].
Prussian blue (PB) was encapsulated by CCMV capsid proteins due to electrostatic repulsion. PB, a metal complex with a negative charge, was encapsulated via a disassembly/reassembly approach in which the pH was dropped from 7.5 to 5.2. Bimetallic nanoparticles were efficiently self-organized because of PB’s encapsulation and crystallization in CCMV [164].
The CCMV coat protein was expressed in the yeast Pichia pastoris and purified before being reassembled into CCMV empty capsids. The CCMV virus itself contains a metal-binding domain that has been found to bind to Terbium (III). It was shown that the particles could bind Gd3+, creating paramagnetic nanoparticles with relaxivity measurements 5–10 times greater than any of those recorded for Gd3+-albumin or Gd3+-dendrimers [165,166].
Unique nanomaterials and nanotechnologies can be created by depositing materials, including Ag, Ni, Co, Cu, Pt, and Fe-Pt alloy, into the 4 nm core cavity of TMV [167,168,169]. Even more so, Kobayashi et al. employed genetically engineered tobamoviruses to generate magnetized 3 nm tube-shaped VNPs. Tomato mosaic virus (ToMV) central channel amino acid residues were genetically modified to increase the amount of positive charge nucleation sites to absorb precursor cations. This led to the incorporation of a CoPt alloy into the inner channels within the ToMV [170]. Tobamovirus VNPs are an exciting new way to make a wide variety of nanoscale building blocks that can be used to make electronics.
The templated manufacturing of highly dispersed gold nanoparticles describes an innovative use of polyelectrolyte-modified Cowpea mosaic virus (CPMV). Poly(allylamine) hydrochloride (PAH), a cationic polyelectrolyte, is electrostatically attached to the exterior of the virus capsid, where it facilitates the adsorption of anionic gold complexes that can be readily reduced under mild conditions to generate a metallic gold coating. Thiol reagents allow for further modification of the templated gold nanoparticles. When polyelectrolyte-modified CPMV (CPMV-PA) reacts with already-formed gold nanoparticles, large, hexagonally-packed, tessellated spheres form on their own [171].
Their distinctive material properties and larger surface area may provide higher activity over comparable bulk materials, making them useful in a variety of industrial applications [172]. Altering the catalyst’s positioning, size, and spatial density is also a powerful tool for managing the reactions that take place during catalysis. PVNs have served as a novel scaffold for the fabrication, anchoring, and exposition of essential nanocatalysts. TMV PVN was used by Yang et al. to generate and distribute Pd nanoparticles with a predetermined size (between 5 and 15 nm) [173]. When used to break down hexavalent chromium (environmental pollutant), the TMV-patterned Pd nanocatalysts had much higher catalytic activity, per unit of Pd bulk, than commercial Pd carbon systems.
The application of plant viruses in the production of nanomaterials and nanodevices has been reviewed in several papers [124,172,174].

5.5. Plant-Derived VNPs Loading Enzymes

Conjugation of enzymes with plant-derived VNPs is a highly beneficial approach for biosensing as it provides good accessibility of the active centers for the analyte molecules while ensuring temperature, pH, and proteases protection [174].
For example, TMV proved to be a suitable enzyme nanocarrier when biotin-streptavidin [SA] bioaffinity binding was applied to load glucose oxidase for amperometric detection of glucose [175]. The same principle was used to install a two-enzyme system, of glucose oxidase (GOx) and horseradish peroxidase (HRP), for the colorimetric detection of glucose. TMV tubes improved catalytic activities up to 45-fold compared to control samples with the same input of free enzymes [176]. Additionally, TMV nanorods loaded with [SA]-penicillinase (Pen) were successfully used for antibiotic detection because the integration increased the reusability and stability of the enzyme [177].
Potyvirus turnip mosaic virus (TuMV) also has great potential as a foreign epitope carrier: the virion’s external surface is rich in functional groups susceptible to chemical conjugations. In particular, Candida antarctica lipase B (CALB) conjugated onto the external surface of the TuMV demonstrated increased specific activity compared to the non-immobilized enzyme [178].
In addition, loaded VNPs with catalytic utilities can be used to activate drugs. For example, CCMV carrying CYPBM3 (a cytochrome P450 variant) is capable of transforming and activating the chemotherapeutic prodrug tamoxifen, the prodrug resveratrol, or similar products [179].

5.6. Plant-Derived VNPs Decorated with Antibodies and Nanobodies

Therapeutic and diagnostic applications of monoclonal antibodies (mAbs) are growing worldwide. The mAbs market is the largest sector of the global biopharmaceutical market, with sales exceeding over USD 100 billion worldwide [180], and is projected to reach USD 425 billion by 2028 [181]. Plants are an alternative source of recombinant mAbs [182]. Transgenic plants can provide large-scale production of recombinant mAbs, which are used for passive immunotherapy [182,183,184]. In addition, because of their high specificity and affinity binding, antibodies can be used to recognize and specifically bind to other molecules and, as such, become building blocks for nanomaterials. Antibodies can recognize a wide variety of targets, not only proteins and peptides but also small molecules (haptens), carbohydrates, and nucleic acids [185]. Smolenska et al. generated PVX particles linked with a scFv against the herbicide diuron [186]. The “overcoat strategy” was used to decorate the tobamovirus particles with the small protein A (33 amino acids). These modified nanoparticles were exploited for affinity purification of mAbs with a recovery yield of 50% and >90% purity [187]. Antibodies can not only be used for building simple or complex nanomaterials, but they can also add specific functional components to these materials. For example, with the help of antibodies, nanomaterials can gain the ability to bind and deliver specific structures to target cells, activate and recruit effector cells, or present specific enzymatic or sensor activities.
A new alternative to mAbs are the nanobodies, which are heavy-chain only antibodies (HcAbs) with a single variable domain (VHH, ~15kDa), produced in camels [188]. Nanobodies have great potential in therapeutics because they bind their epitopes with high specificity and strong affinity [189]. Plant-derived genetically modified VNPs from zucchini yellow mosaic virus (ZYMV) and tobacco etch virus (TEV) were used as scaffolds for nanobodies against the green fluorescent protein. The recombinant VNPs decorated with anti-GFP nanobodies were able to bind the GFP, demonstrating the efficacy of this technology [190]. This research demonstrated the sustainable production of plant nanoparticles decorated with nanobodies for diagnostic and therapeutic purposes.

5.7. Plant Viruses and VNPs Used for Cancer Immunotherapy

VLPs are evolutionarily conserved structures that mimic pathogens and can act as pathogen-associated molecular patterns (PAMPs). They are non-infectious and can induce potent cellular and humoral immune response by triggering multiple signaling pathways [191]. They are an ideal platform for vaccine development and immunotherapy. Plant viruses and their VLPs can be immunogenic for mammals. The cell-surface Toll-like receptors (TLR2 and TLR4) bind mainly viral capsid proteins, while intracellular TLRs (TLR3, TLR7, TLR8, and TLR9) detect nucleic acids and trigger the innate immune response [192]. Plant viruses and their capsid proteins, as eVLPs, can also be used as cancer vaccines when administered intratumorally [193].
A functioning immune system is crucial for immunosurveillance and protection against malignant cells within the host organism, in addition to its obvious importance in fending off intruders from outside the body. Lymphoma, Kaposi’s sarcoma, and cervical carcinoma are only some of the cancers that are more likely to occur if the human immune system is compromised [194]. Tumors can evade the immune system in several ways, including by changing their antigenic profile, suppressing their expression of major histocompatibility complex (MHC) molecules, releasing antigenic peptides into the surrounding environment, or secreting immunosuppressive chemicals [195]. Physiological immune regulatory pathways are used in some of the tumor immune evasion strategies. To suppress a T-cell-mediated immune response, it is sometimes necessary to target T-cells expressing cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or programmed cell death protein-1 (PD-1). Tumor cells can also trigger apoptosis in T-cells which exhibit the Fas receptor by expressing the Fas ligand (FasL, CD95) [196].
In the case of non-Hodgkin’s B-cell lymphomas (NHL), a novel and effective drug delivery method was discovered using PVX’s binding affinity towards malignant B cells. In a mouse model of NHL, the progression of the disease was slowed when monomethyl auristatin, loaded on PVX, was administered into regions that contained malignant B cells [197]. Using a biotin/streptavidin linker, Jobsri et al. (2015) reported that PVX attached to an idiotypic (Id) tumor-associated antigen (TAA) recombinant protein resulted in a 7-fold stronger anti-Id IgG response than Id alone in a mouse B-cell lymphoma model. TLR7 was shown to be necessary for viral RNA recognition. The cytokine profile of these animals showed that IFN- and IL-12 were induced [198]. Trastuzumab (Herceptin) is a human epidermal growth factor receptor two (HER2) binding site-targeting monoclonal antibody that inhibits the development of cancer cells. Immunogenic trastuzumab-binding peptides (TBP) can stimulate the body to make antibodies that block the development of HER2-positive cancer cells. Thus, TMV particles exhibiting TBP have been engineered to stimulate such immunogenicity [199]. Trastuzumab has been shown to successfully trigger apoptosis in breast cancer cell lines once loaded onto PVX nanofilaments [200,201].
A protein called vimentin is present on the surface of most cells, and it has been suggested that CPMV nanoparticles might adhere to it. Due to its upregulation during tumor growth, vimentin is a promising therapeutic target for treating cancers. Stainmetz et al. showed that surface vimentin expression linked with CPMV absorption substantiated CPMV’s efficacy in detecting invasive cancer cells [202]. In mouse models of lung melanoma, ovarian cancer, colon cancer, and breast cancer, CPMV VLP nanoparticles were found to block tumor development. It was shown that CPMV reprograms the tumor microenvironment by attracting natural killer cells and neutrophils and allowing M2 to M1 antitumor macrophages to switch to their more effective M1 state [203]. Cell lysis occurs because of this innate immune cell population’s fight against the tumor. Mao et al. (2021) have recently established which Toll-like receptors (TLRs) were accountable for such features [193]. Effective lymphatic delivery of the human epidermal growth factor receptor 2 (HER2) epitope was accomplished after conjugation to the icosahedral CPMV, resulting in increased absorption and activation of APCs and a consequently increased anti-HER2 immune response. In animal research, the CPMV HER2 candidate vaccination prolonged longevity by decreasing tumor growth and metastasis. Notably, in mouse models, CPMV-HER2 elicited a largely Th1 immune response, while Sesbania Mosaic Virus-HER2 and CCMV-HER2 elicited a predominantly Th2 response, demonstrating that the type of the epitope carrier itself plays a crucial role in determining the Th1/Th2 bias [204]. The variation in epitope presentation between the VNPs’ surface and the capsid may be too accountable for this phenomenon [205]. The immunological response to TACAs may be improved if plant viruses were utilized to deliver these molecules. The Tn antigen (GalNAc—O-Ser/Thr)-targeting CPMV-TACA conjugates were shown to generate higher IgG titers in mouse models, suggesting stronger T-cell mediated immunity and antibody isotype switching. In studies where mouse sera were added to breast cancer cell lines, IgG binding to the Tn antigens was detected [206]. Wang and Steinmetz (2019) delved deep and discovered that a protein called CD47, which is extensively expressed on tumor cells, inhibits the activity of T cells and phagocytic cells. To stimulate an antitumor immune response, the researchers utilized a combined treatment using CD47-blocking antibodies and CPMV nanoparticles. The same research team also utilized combination treatment with modest concentrations of cyclophosphamide (CPA) and CPMV nanoparticles to effectively shrink mice tumors in vivo [207].

5.8. Plant Viruses VLPs-Based Display Platform for Immunogenic Peptides

Plant viruses displaying heterologous epitopes have been used in numerous animal and human clinical studies with positive results. Each of the test animals was successfully protected from a challenge with the virulent mink enteritis virus thanks to CPMV particles expressing a 17-mer neutralizing epitope from the VP2 capsid protein [57]. Antibody responses were enhanced when the peptide was presented on the interface of both the L and S coat protein subunits in a modified construct, as opposed to only the L subunit. Hemocyanin-conjugated keyhole limpet predominant IgG-2a levels pointed to early stimulation of T-helper type 1 cells (TH1) [208]. The proliferation and interferon-production of murine cells treated in vitro with virus particles corroborated these findings. The immune response in the nasal mucosa after intranasal vaccination was higher than the reaction in the blood serum. These results demonstrate that presenting peptides on viral particles can change the immune response’s bias toward a TH1 response (activation of macrophages and cytotoxic T cells) and that recombinant viruses can provide protection against systemic and mucosal infections. Predominant IgG-2a levels pointed to early stimulation of T-helper type 1 cells (TH1) [209,210].
Bacterial pathogen epitopes, such as those found in Pseudomonas aeruginosa and Staphylococcus aureus, have also been presented using CPMV particles. Subcutaneous immunization of mice and rats with CPMV particles showing the D2 peptide from the S. aureus fibronectin-binding protein elicited significant titers of fibronectin-binding protein-specific antibodies. Serum from immunized mice decreased the recombinant fibronectin-binding protein’s ability to bind to immobilized fibronectin, while rat serum was able to prevent S. aureus from adhering to fibronectin [211,212]. PVX was also used for displaying S. aureus protein A fragments on its surface. The protein A coated PVX was easily functionalized with IgG and used in biosensing of plant viruses [213].
Another example is the use of genetically engineered VLPs of Papaya mosaic virus as a seasonal flu trivalent vaccine [214,215].
These results demonstrate the potential efficacy of recombinant plant virus vaccines in preventing invasive endocarditis, septicemia, peritonitis, and bovine mastitis caused by S. aureus. C57BL/6 mice immunized with CPMV particles presenting a linear B-cell epitope from P. aeruginosa outer membrane protein F generated peptide-specific antibodies that bound complement receptors and enhanced phagocytosis of P. aeruginosa by human neutrophils in vitro [212,216].
Tobacco mosaic VLPs have been used as epitope display systems for the first time in the production of a polio vaccine [217]. Since then, they have also been utilized in the production of various vaccines, such as malaria, human papillomavirus, rabies, hepatitis B, influenza, HIV, norovirus, and foot and mouth disease virus [211,218,219,220,221].

6. Methods of Application of Vaccines Produced in Plants

Different vaccine delivery methods include injection, oral, or intranasal administration. In general, the delivery of human vaccines is by injection. In veterinary medicine, when immunizing large groups of animals, many factors determine the most effective immunization method. The vaccines for the big farm animals such as cattle, sheep, goats, and pigs are delivered parenterally. Those for use in poultry are delivered by injection or orally. Oral immunization is the most preferred vaccination method for fish and other aquaculture animals. Generally, oral vaccination provides a time-saving and non-stressful application for both animals and humans [222]. However, the oral administration of vaccines has some disadvantages. Delivering a specific dose of the vaccinogen is difficult, especially when it is a non-purified recombinant immunogenic protein within a living plant [223,224]. Additionally, the vaccinogen must pass through the stomach with its low pH and digestive enzymes and retain its immunogenicity. VLPs or other highly organized particles have been shown to be more resistant to the stomach’s digestive enzymes. They are more likely to retain their immunogenicity, inducing a strong immune response in small animals [225,226]. VLPs produced in plants may represent a cost-effective approach to induce mucosal immunity by oral delivery [227]. Although the commercial implementation of this strategy in the developed world faces several regulatory challenges, this strategy may offer an attractive option for veterinary vaccine delivery in both the developed and developing world [213]. Multiple reports have shown that plant expression systems offer very good solutions for the production of vaccines for veterinary use [17,228,229,230,231,232].

Can Plant-Based Oral Vaccines Help the One Health Approach?

The concept of One Health aims to achieve optimal health outcomes by recognizing the interconnection between people, animals, plants, and their shared environment (CDC). The idea of producing oral vaccines in edible plants carries the potential to change the pharmaceutical industry by reducing production costs by eliminating expensive downstream purifications and cold storage. However, despite the enormous research conducted to invent edible vaccines, no licensed commercial product has been approved for human or veterinary use [233]. The possible cross-contamination of nearby growing food crops with recombinant genes stifled the efforts to produce vaccines in edible crops [18]. Efforts then turned to microalgae and certain plant species, which are generally recognized as safe (GRAS) by the FDA, resurrecting the hope that oral vaccines can be produced in photosynthetic organisms without processing [234,235,236]. Edible vaccines can stimulate mucosal immunity and might be the solution for reducing zoonotic diseases distributed by wild animals and an efficient method for vaccinating large groups of animals, especially aquatic animals [228,237,238,239].
Many immunogenic proteins (viral proteins and bacterial toxins) have been expressed in plants and algae. Animal immunization studies have demonstrated the efficiency of edible vaccines in stimulating the immune system. Table 2 summarizes the achievements in producing edible vaccines in plants and algae for veterinary application.

7. Products, Market Size with Some Examples

From 2018 to 2026, the worldwide plant-based biologics market is expected to rise at a compound annual growth rate (CAGR) of 6.1%, reaching USD 162.4 million by 2026. The European plant-based biologics market is forecasted to grow at a CAGR of 8.1% for the same period as mentioned above [252]. The plant-based biologics products are the result of state-of-the-art studies and possess numerous advantages in terms of product safety, production scalability, easy storage, fast research and development process, and provide the opportunity to supply low-cost biologics to the low income countries. The growing demand for biological products cannot be fully met by current production platforms such as yeast, E. coli, insect cells, Chinese hamster ovary (CHO) cells, and embryonated hen’s eggs (EHE), due to limitations in scalability and in some cases higher cost. The plant expression systems imposed themselves as a superior alternative to conventional production systems due to increased yield of recombinant proteins, good stability, improved glycosylation patterns, and downstream processing [253,254,255,256,257,258].
The rapid development of novel biologics (vaccines, diagnostic reagents, and therapeutics) are among the main objectives of companies such as Protalix Biotherapeutics (Carmiel, Israel); Medicago Inc. (Québec, Canada); iBio/Caliber Therapeutics (Bryan, TX, USA); Kentucky BioProcessing Inc. (Owensboro, KY, USA; Fraunhofer USA (Plymouth, MI, USA); Pfizer (New York, NY, USA); Baiya Phytopharm (Khwaeng Prawet, Prawet, Bangkok, Thailand); Leaf Expression Systems (Norwich Research Park, Norwich, UK), Cape Biologix Technologies (Ndabeni, Cape Town, South Africa); Zea Biosciences (Walpole, MA, USA); Planet Biotechnology Inc. (Hayward, CA, USA); Mapp Biopharmaceutical Inc. (San Diego, CA, USA); and others. The breakthrough in plant-made vaccines was made by Dow AgroSciences, receiving the first USDA approval for a vaccine against NDV, based on the HN protein produced in an N. benthamiana cell culture [24]. Protalix Biotherapeutics received the first FDA approval for commercial use in humans of their plant-made recombinant human enzyme β-glucocerebrosidase for Gaucher disease. Pfizer received the license for this product and marketed it as Elelyso® [259]. Mapp Biopharmaceutical Inc. was granted fast-track designation by the FDA for their plant-made cocktails of three monoclonal antibodies (ZMapp) against Ebola virus disease [260,261]. We have even witnessed the first licensed plant-derived vaccines Covifenz against influenza and SARS-CoV-2 developed by Medicago [26].
The major players in PMF have a number of biopharmaceutical products in clinical trials or products that are awaiting approval from regulatory authorities. The American biotech company iBio Inc. has four plant-based biologics in preclinical trials. IBIO-100 targets fibrosis, IBIO-200 is a virus-like particle (VLP) vaccine against SARS-CoV-2, IBIO-201 is a vaccine targeting COVID-19 using the patented LicKM platform, and IBIO-400 is a vaccine targeting classical swine fever (CSF) disease [262]. Furthermore, KBP [263] and Baiya Phytopharm [264] announced that they have produced subunit vaccines against SARS-CoV-2 in plants [265].
Proteins with antibacterial properties can be used as medications and/or as antibacterial food additives. Using GRAS (Generally Recognized As Safe) regulatory procedures, Nomad Bioscience GmbH (Nomad) has been allowed by the United States Food and Drug Administration (FDA) to use Escherichia-derived (colicins) and Salmonella-derived (salmocins) antibacterial proteins in treatments of fresh or processed fruits and vegetables (colicins), or meat, poultry, fish, and whole eggs (salmocins). These bacteriocins, produced in plant expression systems, have been proven highly effective, even when used in low concentrations, for controlling pathogenic bacteria in food products and avoiding food poisoning. In addition, their production is also financially justifiable [266,267].
Clinical trials of innovative biological treatments such as nanobodies, recombinant vaccines, fusion proteins, antisense RNAi therapies, and gene and cell therapy are underway. Hence, such product developments are expected to positively impact the growth of the global plant-based market.

8. Challenges in Developing Biopharmaceuticals in Plants

Among the main challenges of using plants and different plant viruses as expression platforms for human pharmaceuticals are the plant glycosylation pathways resulting in distinct plant-specific glycans from those in humans or for that matter in most mammalian glycoproteins. The concern is that epitopes carrying non-human glycoforms (β1,2-xylose and α1,3-fucose) would be regarded as foreign by the immune system and hence deemed to be more immunogenic in humans resulting in mild or hypersensitive allergic response [268]. However, it should be noted that these concerns were alleviated to some extent by recent safety data on Medicago Inc. influenza and SARS-CoV-2 vaccines clinical trials phase 3. Volunteers, some of whom had existing plant allergies, were followed for six months and none developed allergic or hypersensitivity reactions.
Potentially, the impact of non-human glycoforms may be more relevant with plant monoclonals as they are usually administered intravenously in mg quantities compared to ug in vaccines or other pharmaceuticals. Still, it is not known if repeated immunizations, as is the case with the annual flu vaccine, or repeat potential boosters, for instance with plant-derived Medicago SARS-CoV-2 vaccine, may have undesirable immune responses due to plant glycans. On the other hand, the potential immunogenicity of plant glycans in some cases could be beneficial, for instance with cancer vaccines/immunotherapeutics enhancing their effect [269].
On the opposite spectre of unwelcome impact due to the expanded immunogenicity of plant glycoproteins is their potential rapid clearance by the immune system which would diminish their efficacy as pharmaceuticals. Conversely, some native, complex mammalian glycoproteins may have a detrimental impact on the plant cellular glycosylation mechanisms leading to endoplasmic reticulum-related stress response demonstrated by leaf necrosis that ultimately results in low yield of recombinant proteins. To alleviate the majority of the above mentioned drawbacks, different glycoengineering strategies have been recently developed to undo undesirable immunogenic N-glycan modifications in plants such as (i) “humanization” of plant N-glycans by production of knock-out lines removing genes responsible for plant-specific glycosylation by using the CRISPR/Cas9 system, (ii) introduction of humanized N-glycosylation pathways in plants, and (iii) co-expression of mammalian native chaperons and folding enzymes compatible with the plant endogenous chaperone machinery. This is an incomplete set of glycoengineering tools to modify plant glycosylation pathways in order to improve tomorrow’s plant pharmaceuticals.

9. Conclusions

Since their discovery, plant viruses have been transformed into a major molecular tool used for recombinant protein expressions, vaccine manufacturing, and as nonoagents for drug delivery. Whole virions, naturally occurring empty capsids of plant viruses, nanoparticles generated by reassembly of coat proteins, and VLPs displaying heterologous peptides or even whole proteins on their surface or inside their cavity are all examples of plant-made nanoparticles used for vaccine and biologics production, and in nanoscale drug delivery applications. Unlike the mainstream expression platforms, the production of plant-based biologics has its advantages: absence of major safety concerns, no need for expensive equipment and continuous temperature control, easy scalability, and affordability. Several promising plant-derived biologics are being tested in clinical trials, which should lead to cutting-edge therapies in the rapidly growing field of VNP- and VLP-based drug delivery applications.

Author Contributions

Conceptualization, G.Z., A.A.A. and A.P.A.; formal analysis, G.Z.; data curation, K.T. and V.T.; writing—original draft preparation, G.Z., K.T., A.A.A., A.P.A., M.M.T. and I.M.; writing—review and editing; G.L.L. and G.Z.; funding acquisition, G.Z. and I.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the European Union’s Horizon 2020 research and innovation programme, project PlantaSYST (SGA-CSA No. 739582 under FPA No. 664620) and by the European Regional Development Fund through the Bulgarian “Science and Education for Smart Growth” Operational Programme (project BG05M2OP001-1.003-001-C01), Research Fund at the University of Plovdiv, competition “Young Scientists and Doctoral Students” MU21-BF-022.

Acknowledgments

The authors would like to thank Valeria Tonova (bachelor student, University of Plovdiv) for helping with the graphic figures’ preparation, the program “Young scientists and Postdoctoral candidates—2” from the Bulgarian Ministry of Education and Science for funding Katerina Takova as a postdoctoral scientist, and L’Oréal and UNESCO initiative “For Women in Science 2022” for supporting Katerina Takova’s research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Steinmetz, N.F.; Lin, T.; Lomonossoff, G.P.; Johnson, J.E. Structure-Based Engineering of an Icosahedral Virus for Nanomedicine and Nanotechnology. In Viruses and Nanotech; Manchester, M., Steinmetz, N.F., Eds.; Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 2009; pp. 23–58. ISBN 978-3-540-69379-6. [Google Scholar]
  2. Aljabali, A.A.A.; Shukla, S.; Lomonossoff, G.P.; Steinmetz, N.F.; Evans, D.J. CPMV-DOX Delivers. Mol. Pharm. 2013, 10, 3–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zeng, Q.; Wen, H.; Wen, Q.; Chen, X.; Wang, Y.; Xuan, W.; Liang, J.; Wan, S. Cucumber Mosaic Virus as Drug Delivery Vehicle for Doxorubicin. Biomaterials 2013, 34, 4632–4642. [Google Scholar] [CrossRef] [PubMed]
  4. Ibrahim, A.; Odon, V.; Kormelink, R. Plant Viruses in Plant Molecular Pharming: Toward the Use of Enveloped Viruses. Front. Plant Sci. 2019, 10, 803. [Google Scholar] [CrossRef]
  5. Nooraei, S.; Bahrulolum, H.; Hoseini, Z.S.; Katalani, C.; Hajizade, A.; Easton, A.J.; Ahmadian, G. Virus-like Particles: Preparation, Immunogenicity and Their Roles as Nanovaccines and Drug Nanocarriers. J. Nanobiotechnol. 2021, 19, 59. [Google Scholar] [CrossRef]
  6. Fraley, R.T.; Rogers, S.G.; Horsch, R.B.; Sanders, P.R.; Flick, J.S.; Adams, S.P.; Bittner, M.L.; Brand, L.A.; Fink, C.L.; Fry, J.S.; et al. Expression of Bacterial Genes in Plant Cells. Proc. Natl. Acad. Sci. USA 1983, 80, 4803–4807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Witcher, D.R.; Hood, E.E.; Peterson, D.; Bailey, M.; Bond, D.; Kusnadi, A.; Evangelista, R.; Nikolov, Z.; Wooge, C.; Mehigh, R.; et al. Commercial Production of β-Glucuronidase (GUS): A Model System for the Production of Proteins in Plants. Mol. Breed. 1998, 4, 301–312. [Google Scholar] [CrossRef]
  8. Hood, E.E.; Witcher, D.R.; Maddock, S.; Meyer, T.; Baszczynski, C.; Bailey, M.; Flynn, P.; Register, J.; Marshall, L.; Bond, D.; et al. Commercial Production of Avidin from Transgenic Maize: Characterization of Transformant, Production, Processing, Extraction and Purification. Mol. Breed. 1997, 3, 291–306. [Google Scholar] [CrossRef]
  9. Woodard, S.L.; Mayor, J.M.; Bailey, M.R.; Barker, D.K.; Love, R.T.; Lane, J.R.; Delaney, D.E.; McComas-Wagner, J.M.; Mallubhotla, H.D.; Hood, E.E.; et al. Maize (Zea Mays)-Derived Bovine Trypsin: Characterization of the First Large-Scale, Commercial Protein Product from Transgenic Plants. Biotechnol. Appl. Biochem. 2003, 38, 123–130. [Google Scholar] [CrossRef] [PubMed]
  10. Barta, A.; Sommergruber, K.; Thompson, D.; Hartmuth, K.; Matzke, M.A.; Matzke, A.J.M. The Expression of a Nopaline Synthase—Human Growth Hormone Chimaeric Gene in Transformed Tobacco and Sunflower Callus Tissue. Plant Mol Biol 1986, 6, 347–357. [Google Scholar] [CrossRef] [PubMed]
  11. During, K. Wound-Inducible Expression and Secretion of T4 Lysozyme and Monoclonal Antibodies in Nicotiana Tabacum. Ph.D. Thesis, Mathematisch-Naturwissenschaftlichen Fakultat der Universität zu Köln, Köln, Germany, 1988. [Google Scholar]
  12. Arntzen, C. Plant-made Pharmaceuticals: From ‘Edible Vaccines’ to Ebola Therapeutics. Plant Biotechnol. J. 2015, 13, 1013–1016. [Google Scholar] [CrossRef]
  13. Loza-Rubio, E.; Rojas, E.; Gómez, L.; Olivera, M.T.J.; Gómez-Lim, M.A. Development of an Edible Rabies Vaccine in Maize Using the Vnukovo Strain. Dev. Biol. 2008, 131, 477–482. [Google Scholar]
  14. Zahmanova, G.; Falzarano, D.; Naimov, S.; Kostova, M.; Boncheva, R.; Dukiandjiev, S.; Minkov, I.; Andonov, A. Oral immunization with truncated hepatitis B virus nucleocapsid expressed in transgenic potatoes. Comptes Rendus L’acade’mie Bulg. Des Sci. 2008, 61, 1293–1300. [Google Scholar]
  15. Arntzen, C.; Plotkin, S.; Dodet, B. Plant-Derived Vaccines and Antibodies: Potential and Limitations. Vaccine 2005, 23, 1753–1756. [Google Scholar] [CrossRef] [PubMed]
  16. Noncompliance History. Available online: https://www.aphis.usda.gov/aphis/ourfocus/biotechnology/compliance-and-inspections/CT_Compliance_history (accessed on 8 December 2022).
  17. Rybicki, E.P. Plant-Made Vaccines for Humans and Animals. Plant Biotechnol. J. 2010, 8, 620–637. [Google Scholar] [CrossRef]
  18. Rybicki, E.P. Plant-Produced Vaccines: Promise and Reality. Drug Discov. Today 2009, 14, 16–24. [Google Scholar] [CrossRef] [PubMed]
  19. Zahmanova, G.; Mazalovska, M.; Toneva, V.; Minkov, I.; Lomonossoff, G. Production of Chimeric Virus-like Particles Bearing M2e Influenza Epitope in Nicotiana Benthamiana Plants. J. Biotechnol. 2015, 208, S109. [Google Scholar] [CrossRef]
  20. Mardanova, E.S.; Kotlyarov, R.Y.; Stuchinskaya, M.D.; Nikolaeva, L.I.; Zahmanova, G.; Ravin, N.V. High-Yield Production of Chimeric Hepatitis E Virus-Like Particles Bearing the M2e Influenza Epitope and Receptor Binding Domain of SARS-CoV-2 in Plants Using Viral Vectors. Int. J. Mol. Sci. 2022, 23, 15684. [Google Scholar] [CrossRef] [PubMed]
  21. Rebelo, B.A.; Folgado, A.; Ferreira, A.C.; Abranches, R. Production of the SARS-CoV-2 Spike Protein and Its Receptor Binding Domain in Plant Cell Suspension Cultures. Front. Plant Sci. 2022, 13, 995429. [Google Scholar] [CrossRef]
  22. Jung, J.-W.; Zahmanova, G.; Minkov, I.; Lomonossoff, G.P. Plant-Based Expression and Characterization of SARS-CoV-2 Virus-like Particles Presenting a Native Spike Protein. Plant Biotechnol. J. 2022, 20, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  23. Kopertekh, L.; Schiemann, J. Transient Production of Recombinant Pharmaceutical Proteins in Plants: Evolution and Perspectives. Curr. Med. Chem. 2019, 26, 365–380. [Google Scholar] [CrossRef] [PubMed]
  24. Vermij, P.; Waltz, E. USDA Approves the First Plant-Based Vaccine. Nat. Biotechnol. 2006, 24, 233–234. [Google Scholar]
  25. Maxmen, A. Drug-Making Plant Blooms. Nature 2012, 485, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Medicago Inc Medicago and GSK Announce the Approval by Health Canada of COVIFENZ®, an Adjuvanted Plant-Based COVID-19 Vaccine. Available online: https://medicago.com/en/press-release/covifenz/ (accessed on 26 February 2022).
  27. Ward, B.J.; Makarkov, A.; Séguin, A.; Pillet, S.; Trépanier, S.; Dhaliwall, J.; Libman, M.D.; Vesikari, T.; Landry, N. Efficacy, Immunogenicity, and Safety of a Plant-Derived, Quadrivalent, Virus-like Particle Influenza Vaccine in Adults (18–64 Years) and Older Adults (≥65 Years): Two Multicentre, Randomised Phase 3 Trials. Lancet 2020, 396, 1491–1503. [Google Scholar] [CrossRef]
  28. Roossinck, M.J. Plant Virus Ecology. PLoS Pathog. 2013, 9, e1003304. [Google Scholar] [CrossRef]
  29. Scholthof, K.-B.G.; Adkins, S.; Czosnek, H.; Palukaitis, P.; Jacquot, E.; Hohn, T.; Hohn, B.; Saunders, K.; Candresse, T.; Ahlquist, P.; et al. Top 10 Plant Viruses in Molecular Plant Pathology. Mol. Plant Pathol. 2011, 12, 938–954. [Google Scholar] [CrossRef] [PubMed]
  30. Balke, I.; Zeltins, A. Use of Plant Viruses and Virus-like Particles for the Creation of Novel Vaccines. Adv. Drug Deliv. Rev. 2019, 145, 119–129. [Google Scholar] [CrossRef]
  31. Love, A.J.; Makarov, V.; Yaminsky, I.; Kalinina, N.O.; Taliansky, M.E. The Use of Tobacco Mosaic Virus and Cowpea Mosaic Virus for the Production of Novel Metal Nanomaterials. Virology 2014, 449, 133–139. [Google Scholar] [CrossRef] [Green Version]
  32. Marsian, J.; Lomonossoff, G.P. Molecular Pharming—VLPs Made in Plants. Curr Opin Biotechnol 2016, 37, 201–206. [Google Scholar] [CrossRef] [Green Version]
  33. Evans, D.J. The Bionanoscience of Plant Viruses: Templates and Synthons for New Materials. J. Mater. Chem. 2008, 18, 3746–3754. [Google Scholar] [CrossRef]
  34. Sainsbury, F.; Lomonossoff, G.P. Transient Expressions of Synthetic Biology in Plants. Curr. Opin. Plant Biol. 2014, 19, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Clare, D.K.; Orlova, E.V. 4.6Å Cryo-EM Reconstruction of Tobacco Mosaic Virus from Images Recorded at 300keV on a 4k×4k CCD Camera. J. Struct. Biol. 2010, 171, 303–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Schramm, G.; Schumacher, G.; Zillig, W. An Infectious Nucleoprotein from Tobacco Mosaic Virus. Nature 1955, 175, 549–550. [Google Scholar] [CrossRef]
  37. Harrison, B.D.; Wilson, T.M.A.; Butler, P.J.G. Self–Assembly of Tobacco Mosaic Virus: The Role of an Intermediate Aggregate in Generating Both Specificity and Speed. Philos. Trans. R. Soc. London Ser. B Biol. Sci. 1999, 354, 537–550. [Google Scholar] [CrossRef]
  38. Okada, Y. Molecular Assembly of Tobacco Mosaic Virus in Vitro. Adv. Biophys. 1986, 22, 95–149. [Google Scholar] [CrossRef] [PubMed]
  39. Shire, S.J.; Steckert, J.J.; Adams, M.L.; Schuster, T.M. Kinetics and Mechanism of Tobacco Mosaic Virus Assembly: Direct Measurement of Relative Rates of Incorporation of 4S and 20S Protein. Proc. Natl. Acad. Sci. USA 1979, 76, 2745–2749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kegel, W.K.; van der Schoot, P. Physical Regulation of the Self-Assembly of Tobacco Mosaic Virus Coat Protein. Biophys. J. 2006, 91, 1501–1512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Pitek, A.S.; Hu, H.; Shukla, S.; Steinmetz, N.F. Cancer Theranostic Applications of Albumin-Coated Tobacco Mosaic Virus Nanoparticles. ACS Appl. Mater. Interfaces 2018, 10, 39468–39477. [Google Scholar] [CrossRef]
  42. Bruckman, M.A.; Steinmetz, N.F. Chemical Modification of the Inner and Outer Surfaces of Tobacco Mosaic Virus (TMV). Methods Mol. Biol. 2014, 1108, 173–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Bruckman, M.A.; Hern, S.; Jiang, K.; Flask, C.A.; Yu, X.; Steinmetz, N.F. Tobacco Mosaic Virus Rods and Spheres as Supramolecular High-Relaxivity MRI Contrast Agents. J. Mater. Chem. B 2013, 1, 1482–1490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Bruckman, M.A.; Czapar, A.E.; VanMeter, A.; Randolph, L.N.; Steinmetz, N.F. Tobacco Mosaic Virus-Based Protein Nanoparticles and Nanorods for Chemotherapy Delivery Targeting Breast Cancer. J. Control. Release 2016, 231, 103–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kernan, D.L.; Wen, A.M.; Pitek, A.S.; Steinmetz, N.F. Featured Article: Delivery of Chemotherapeutic VcMMAE Using Tobacco Mosaic Virus Nanoparticles. Exp. Biol. Med. 2017, 242, 1405–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Lee, K.L.; Carpenter, B.L.; Wen, A.M.; Ghiladi, R.A.; Steinmetz, N.F. High Aspect Ratio Nanotubes Formed by Tobacco Mosaic Virus for Delivery of Photodynamic Agents Targeting Melanoma. ACS Biomater. Sci. Eng. 2016, 2, 838–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Takamatsu, N.; Ishikawa, M.; Meshi, T.; Okada, Y. Expression of Bacterial Chloramphenicol Acetyltransferase Gene in Tobacco Plants Mediated by TMV-RNA. EMBO J. 1987, 6, 307–311. [Google Scholar] [CrossRef]
  48. Lomonossoff, G.P. Cowpea Mosaic Virus. In Encyclopedia of Virology, 3rd ed.; Mahy, B.W.J., Van Regenmortel, M.H.V., Eds.; Academic Press: Oxford, UK, 2008; pp. 569–574. ISBN 978-0-12-374410-4. [Google Scholar]
  49. Lomonossoff, G.P.; Johnson, J.E. The Synthesis and Structure of Comovirus Capsids. Prog. Biophys. Mol. Biol. 1991, 55, 107–137. [Google Scholar] [CrossRef] [PubMed]
  50. Lin, T.; Chen, Z.; Usha, R.; Stauffacher, C.V.; Dai, J.B.; Schmidt, T.; Johnson, J.E. The Refined Crystal Structure of Cowpea Mosaic Virus at 2.8 A Resolution. Virology 1999, 265, 20–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Sainsbury, F.; Cañizares, M.C.; Lomonossoff, G.P. Cowpea Mosaic Virus: The Plant Virus–Based Biotechnology Workhorse. Annu. Rev. Phytopathol. 2010, 48, 437–455. [Google Scholar] [CrossRef] [Green Version]
  52. Aljabali, A.A.A.; Barclay, J.E.; Steinmetz, N.F.; Lomonossoff, G.P.; Evans, D.J. Controlled Immobilisation of Active Enzymes on the Cowpea Mosaic Virus Capsid. Nanoscale 2012, 4, 5640–5645. [Google Scholar] [CrossRef] [Green Version]
  53. Aljabali, A.A.A.; Barclay, J.E.; Lomonossoff, G.P.; Evans, D.J. Virus Templated Metallic Nanoparticles. Nanoscale 2010, 2, 2596–2600. [Google Scholar] [CrossRef]
  54. Evans, D.J. Bionanoscience at the Plant Virus–Inorganic Chemistry Interface. Inorg. Chim. Acta 2010, 363, 1070–1076. [Google Scholar] [CrossRef]
  55. Steinmetz, N.F.; Evans, D.J. Utilisation of Plant Viruses in Bionanotechnology. Org. Biomol. Chem. 2007, 5, 2891–2902. [Google Scholar] [CrossRef]
  56. Porta, C.; Spall, V.E.; Loveland, J.; Johnson, J.E.; Barker, P.J.; Lomonossoff, G.P. Development of Cowpea Mosaic Virus as a High-Yielding System for the Presentation of Foreign Peptides. Virology 1994, 202, 949–955. [Google Scholar] [CrossRef] [PubMed]
  57. Dalsgaard, K.; Uttenthal, Å.; Jones, T.D.; Xu, F.; Merryweather, A.; Hamilton, W.D.O.; Langeveld, J.P.M.; Boshuizen, R.S.; Kamstrup, S.; Lomonossoff, G.P.; et al. Plant–Derived Vaccine Protects Target Animals against a Viral Disease. Nat. Biotechnol. 1997, 15, 248–252. [Google Scholar] [CrossRef] [PubMed]
  58. Montague, N.P.; Thuenemann, E.C.; Saxena, P.; Saunders, K.; Lenzi, P.; Lomonossoff, G.P. Recent Advances of Cowpea Mosaic Virus-Based Particle Technology. Hum. Vaccines 2011, 7, 383–390. [Google Scholar] [CrossRef]
  59. Saunders, K.; Sainsbury, F.; Lomonossoff, G.P. Efficient Generation of Cowpea Mosaicvirus Empty Virus-like Particles by the Proteolytic Processing of Precursors in Insect Cells and Plants. Virology 2009, 393, 329–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Hesketh, E.L.; Meshcheriakova, Y.; Thompson, R.F.; Lomonossoff, G.P.; Ranson, N.A. The Structures of a Naturally Empty Cowpea Mosaic Virus Particle and Its Genome-Containing Counterpart by Cryo-Electron Microscopy. Sci. Rep. 2017, 7, 539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Huynh, N.T.; Hesketh, E.L.; Saxena, P.; Meshcheriakova, Y.; Ku, Y.-C.; Hoang, L.T.; Johnson, J.E.; Ranson, N.A.; Lomonossoff, G.P.; Reddy, V.S. Crystal Structure and Proteomics Analysis of Empty Virus-like Particles of Cowpea Mosaic Virus. Structure 2016, 24, 567–575. [Google Scholar] [CrossRef] [Green Version]
  62. Wang, Q.; Kaltgrad, E.; Lin, T.; Johnson, J.E.; Finn, M.G. Natural Supramolecular Building Blocks. Wild-Type Cowpea Mosaic Virus. Chem. Biol. 2002, 9, 805–811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Tiu, B.D.B.; Advincula, R.C.; Steinmetz, N.F. Nanomanufacture of Free-Standing, Porous, Janus-Type Films of Polymer-Plant Virus Nanoparticle Arrays. Methods Mol. Biol. 2018, 1776, 143–157. [Google Scholar] [CrossRef] [PubMed]
  64. Kruse, I.; Peyret, H.; Saxena, P.; Lomonossoff, G.P. Encapsidation of Viral RNA in Picornavirales: Studies on Cowpea Mosaic Virus Demonstrate Dependence on Viral Replication. J. Virol. 2019, 93, e01520-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Peyret, H.; Lomonossoff, G.P. When Plant Virology Met Agrobacterium: The Rise of the Deconstructed Clones. Plant Biotechnol. J. 2015, 13, 1121–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Caspar, D.L.; Klug, A. Physical Principles in the Construction of Regular Viruses. Cold Spring Harb. Symp. Quant. Biol. 1962, 27, 1–24. [Google Scholar] [CrossRef]
  67. Douglas, T.; Young, M. Host–Guest Encapsulation of Materials by Assembled Virus Protein Cages. Nature 1998, 393, 152–155. [Google Scholar] [CrossRef]
  68. Wilts, B.D.; Schaap, I.A.T.; Schmidt, C.F. Swelling and Softening of the Cowpea Chlorotic Mottle Virus in Response to PH Shifts. Biophys. J. 2015, 108, 2541–2549. [Google Scholar] [CrossRef]
  69. Hema, M.; Vishnu Vardhan, G.P.; Savithri, H.S.; Murthy, M.R.N. Chapter 6—Emerging Trends in the Development of Plant Virus-Based Nanoparticles and Their Biomedical Applications. In Recent Developments in Applied Microbiology and Biochemistry; Buddolla, V., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 61–82. ISBN 978-0-12-816328-3. [Google Scholar]
  70. Minten, I.J.; Ma, Y.; Hempenius, M.A.; Vancso, G.J.; Nolte, R.J.M.; Cornelissen, J.J.L.M. CCMV Capsid Formation Induced by a Functional Negatively Charged Polymer. Org. Biomol. Chem. 2009, 7, 4685–4688. [Google Scholar] [CrossRef]
  71. Lomonossoff, G.P.; Evans, D.J. Applications of Plant Viruses in Bionanotechnology. In Plant Viral Vectors; Palmer, K., Gleba, Y., Eds.; Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 2014; pp. 61–87. ISBN 978-3-642-40829-8. [Google Scholar]
  72. Young, M.; Debbie, W.; Uchida, M.; Douglas, T. Plant Viruses as Biotemplates for Materials and Their Use in Nanotechnology. Annu. Rev. Phytopathol. 2008, 46, 361–384. [Google Scholar] [CrossRef]
  73. Wen, A.M.; Lee, K.L.; Cao, P.; Pangilinan, K.; Carpenter, B.L.; Lam, P.; Veliz, F.A.; Ghiladi, R.A.; Advincula, R.C.; Steinmetz, N.F. Utilizing Viral Nanoparticle/Dendron Hybrid Conjugates in Photodynamic Therapy for Dual Delivery to Macrophages and Cancer Cells. Bioconjug. Chem. 2016, 27, 1227–1235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Narayanan, K.B.; Han, S.S. Icosahedral Plant Viral Nanoparticles—Bioinspired Synthesis of Nanomaterials/Nanostructures. Adv. Colloid Interface Sci. 2017, 248, 1–19. [Google Scholar] [CrossRef] [PubMed]
  75. Van Regenmortel, M.H.V.; Fauquet, C.M.; Bishop, D.H.L.; Carstens, E.B.; Estes, M.K.; Lemon, S.M.; Maniloff, J.; Mayo, M.A.; McGeoch, D.J.; Pringle, C.R.; et al. Classification and Nomenclature of Viruses. In Virus Taxonomy Seventh Report of the International Committee on Taxonomy of Viruses; Academic Press: Cambridge, MA, USA, 2000. [Google Scholar]
  76. Lucas, R.W.; Larson, S.B.; McPherson, A. The Crystallographic Structure of Brome Mosaic Virus, Edited by I. A. Wilson. J. Mol. Biol. 2002, 317, 95–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Cuillel, M.; Zulauf, M.; Jacrot, B. Self-Assembly of Brome Mosaic Virus Protein into Capsids: Initial and Final States of Aggregation. J. Mol. Biol. 1983, 164, 589–603. [Google Scholar] [CrossRef] [PubMed]
  78. Incardona, N.L.; Kaesberg, P. A PH-Induced Structural Change in Bromegrass Mosaic Virus. Biophys. J. 1964, 4, 11–21. [Google Scholar] [CrossRef] [Green Version]
  79. Porta, C.; Lomonossoff, G.P. Viruses as Vectors for the Expression of Foreign Sequences in Plants. Biotechnol. Genet. Eng. Rev. 2002, 19, 245–292. [Google Scholar] [CrossRef]
  80. Howell, S.H.; Walker, L.L.; Dudley, R.K. Cloned Cauliflower Mosaic Virus DNA Infects Turnips (Brassica Rapa). Science 1980, 208, 1265–1267. [Google Scholar] [CrossRef] [PubMed]
  81. Lico, C.; Chen, Q.; Santi, L. Viral Vectors for Production of Recombinant Proteins in Plants. J. Cell. Physiol. 2008, 216, 366–377. [Google Scholar] [CrossRef]
  82. Grimsley, N.; Hohn, B.; Hohn, T.; Walden, R. “Agroinfection,” an Alternative Route for Viral Infection of Plants by Using the Ti Plasmid. Proc. Natl. Acad. Sci. USA 1986, 83, 3282–3286. [Google Scholar] [CrossRef] [Green Version]
  83. Gleba, Y.; Klimyuk, V.; Marillonnet, S. Magnifection—A New Platform for Expressing Recombinant Vaccines in Plants. Vaccine 2005, 23, 2042–2048. [Google Scholar] [CrossRef] [PubMed]
  84. Mardanova, E.S.; Blokhina, E.A.; Tsybalova, L.M.; Peyret, H.; Lomonossoff, G.P.; Ravin, N.V. Efficient Transient Expression of Recombinant Proteins in Plants by the Novel PEff Vector Based on the Genome of Potato Virus X. Front. Plant Sci. 2017, 8, 247. [Google Scholar] [CrossRef] [Green Version]
  85. Sainsbury, F.; Lavoie, P.-O.; D’Aoust, M.-A.; Vézina, L.-P.; Lomonossoff, G.P. Expression of Multiple Proteins Using Full-Length and Deleted Versions of Cowpea Mosaic Virus RNA-2. Plant Biotechnol. J. 2008, 6, 82–92. [Google Scholar] [CrossRef]
  86. Zeng, H.; Xie, Y.; Liu, G.; Wei, Y.; Hu, W.; Shi, H. Agrobacterium-Mediated Gene Transient Overexpression and Tobacco Rattle Virus (TRV)-Based Gene Silencing in Cassava. Int. J. Mol. Sci. 2019, 20, 3976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lee, W.-S.; Hammond-Kosack, K.E.; Kanyuka, K. Barley Stripe Mosaic Virus-Mediated Tools for Investigating Gene Function in Cereal Plants and Their Pathogens: Virus-Induced Gene Silencing, Host-Mediated Gene Silencing, and Virus-Mediated Overexpression of Heterologous Protein. Plant Physiol. 2012, 160, 582–590. [Google Scholar] [CrossRef] [Green Version]
  88. Marillonnet, S.; Giritch, A.; Gils, M.; Kandzia, R.; Klimyuk, V.; Gleba, Y. In Planta Engineering of Viral RNA Replicons: Efficient Assembly by Recombination of DNA Modules Delivered by Agrobacterium. Proc. Natl. Acad. Sci. USA 2004, 101, 6852–6857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Wang, L.-Y.; Lin, S.-S.; Hung, T.-H.; Li, T.-K.; Lin, N.-C.; Shen, T.-L. Multiple Domains of the Tobacco Mosaic Virus P126 Protein Can Independently Suppress Local and Systemic RNA Silencing. Mol. Plant Microbe Interact. 2012, 25, 648–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Liu, Z.; Kearney, C.M. An Efficient Foxtail Mosaic Virus Vector System with Reduced Environmental Risk. BMC Biotechnol. 2010, 10, 88. [Google Scholar] [CrossRef] [Green Version]
  91. Dugdale, B.; Mortimer, C.L.; Kato, M.; James, T.A.; Harding, R.M.; Dale, J.L. In Plant Activation: An Inducible, Hyperexpression Platform for Recombinant Protein Production in Plants. Plant Cell 2013, 25, 2429–2443. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, Q.; He, J.; Phoolcharoen, W.; Mason, H.S. Geminiviral Vectors Based on Bean Yellow Dwarf Virus for Production of Vaccine Antigens and Monoclonal Antibodies in Plants. Hum. Vaccines 2011, 7, 331–338. [Google Scholar] [CrossRef] [Green Version]
  93. Regnard, G.L.; Halley-Stott, R.P.; Tanzer, F.L.; Hitzeroth, I.I.; Rybicki, E.P. High Level Protein Expression in Plants through the Use of a Novel Autonomously Replicating Geminivirus Shuttle Vector. Plant Biotechnol. J. 2010, 8, 38–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. MacFarlane, S.A.; Popovich, A.H. Efficient Expression of Foreign Proteins in Roots from Tobravirus Vectors. Virology 2000, 267, 29–35. [Google Scholar] [CrossRef] [Green Version]
  95. Cañizares, M.C.; Liu, L.; Perrin, Y.; Tsakiris, E.; Lomonossoff, G.P. A Bipartite System for the Constitutive and Inducible Expression of High Levels of Foreign Proteins in Plants. Plant Biotechnol. J. 2006, 4, 183–193. [Google Scholar] [CrossRef]
  96. Sainsbury, F.; Thuenemann, E.C.; Lomonossoff, G.P. PEAQ: Versatile Expression Vectors for Easy and Quick Transient Expression of Heterologous Proteins in Plants. Plant Biotechnol. J. 2009, 7, 682–693. [Google Scholar] [CrossRef] [PubMed]
  97. Hefferon, K. Plant Virus Expression Vectors: A Powerhouse for Global Health. Biomedicines 2017, 5, 44. [Google Scholar] [CrossRef] [Green Version]
  98. Chapman, S.; Kavanagh, T.; Baulcombe, D. Potato Virus X as a Vector for Gene Expression in Plants. Plant J. 1992, 2, 549–557. [Google Scholar] [CrossRef]
  99. Harrison, B.D.; Wilson, T.M.A.; Turpen, T.H. Tobacco Mosaic Virus and the Virescence of Biotechnology. Philos. Trans. R. Soc. London Ser. B Biol. Sci. 1999, 354, 665–673. [Google Scholar] [CrossRef] [PubMed]
  100. Johnson, J.; Lin, T.; Lomonossoff, G. PRESENTATION OF HETEROLOGOUS PEPTIDES ON PLANT VIRUSES: Genetics, Structure, and Function. Annu. Rev. Phytopathol. 1997, 35, 67–86. [Google Scholar] [CrossRef] [PubMed]
  101. Porta, C.; Lomonossoff, G.P. Scope for Using Plant Viruses to Present Epitopes from Animal Pathogens. Rev. Med. Virol. 1998, 8, 25–41. [Google Scholar] [CrossRef]
  102. Gleba, Y.; Marillonnet, S.; Klimyuk, V. Engineering Viral Expression Vectors for Plants: The “full Virus” and the “Deconstructed Virus” Strategies. Curr. Opin. Plant Biol. 2004, 7, 182–188. [Google Scholar] [CrossRef] [PubMed]
  103. Leuzinger, K.; Dent, M.; Hurtado, J.; Stahnke, J.; Lai, H.; Zhou, X.; Chen, Q. Efficient Agroinfiltration of Plants for High-Level Transient Expression of Recombinant Proteins. J. Vis. Exp. 2013, 23, 50521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Klimyuk, V.; Pogue, G.; Herz, S.; Butler, J.; Haydon, H. Production of Recombinant Antigens and Antibodies in Nicotiana Benthamiana Using ‘Magnifection’ Technology: GMP-Compliant Facilities for Small-and Large-Scale Manufacturing. In Plant Viral Vectors; Palmer, K., Gleba, Y., Eds.; Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 2014; pp. 127–154. ISBN 978-3-642-40829-8. [Google Scholar]
  105. Mardanova, E.S.; Takova, K.H.; Toneva, V.T.; Zahmanova, G.G.; Tsybalova, L.M.; Ravin, N.V. A Plant-Based Transient Expression System for the Rapid Production of Highly Immunogenic Hepatitis E Virus-like Particles. Biotechnol. Lett. 2020, 42, 2441–2446. [Google Scholar] [CrossRef]
  106. Mardanova, E.S.; Ravin, N.V. Chapter Ten—Transient Expression of Recombinant Proteins in Plants Using Potato Virus X Based Vectors. In Recombinant Protein Expression: Eukaryotic Hosts; O’Dell, W.B., Kelman, Z., Eds.; Academic Press: Cambridge, MA, USA, 2021; Volume 660, pp. 205–222. [Google Scholar]
  107. Takova, K.; Koynarski, T.; Minkov, G.; Toneva, V.; Mardanova, E.; Ravin, N.; Lukov, G.L.; Zahmanova, G. Development and Optimization of an Enzyme Immunoassay to Detect Serum Antibodies against the Hepatitis E Virus in Pigs, Using Plant-Derived ORF2 Recombinant Protein. Vaccines 2021, 9, 991. [Google Scholar] [CrossRef] [PubMed]
  108. Zahmanova, G.G.; Mazalovska, M.; Takova, K.H.; Toneva, V.T.; Minkov, I.N.; Mardanova, E.S.; Ravin, N.V.; Lomonossoff, G.P. Rapid High-Yield Transient Expression of Swine Hepatitis E ORF2 Capsid Proteins in Nicotiana Benthamiana Plants and Production of Chimeric Hepatitis E Virus-Like Particles Bearing the M2e Influenza Epitope. Plants 2020, 9, 29. [Google Scholar] [CrossRef] [Green Version]
  109. Thuenemann, E.C.; Lenzi, P.; Love, A.J.; Taliansky, M.; Bécares, M.; Zuñiga, S.; Enjuanes, L.; Zahmanova, G.G.; Minkov, I.N.; Matić, S.; et al. The Use of Transient Expression Systems for the Rapid Production of Virus-like Particles in Plants. Curr. Pharm. Des. 2013, 19, 5564–5573. [Google Scholar] [CrossRef] [PubMed]
  110. Zahmanova, G.; Mazalovska, M.; Takova, K.; Toneva, V.; Minkov, I.; Peyret, H.; Lomonossoff, G. Efficient Production of Chimeric Hepatitis B Virus-Like Particles Bearing an Epitope of Hepatitis E Virus Capsid by Transient Expression in Nicotiana Benthamiana. Life 2021, 11, 64. [Google Scholar] [CrossRef]
  111. Zahmanova, G.; Naimov, S.; Mazalovska, M.; Valkova, R.; Minkov, I. Transient Expression of Modified Hepatitis B Capsid Protein in Nicotiana Benthamiana Plants for Viral Nanoparticles Production. J. BioSci. Biotechnol. 2014, 3, SE/ONLINE: 11–16. [Google Scholar]
  112. Lindbo, J.A. TRBO: A High-Efficiency Tobacco Mosaic Virus RNA-Based Overexpression Vector. Plant Physiol. 2007, 145, 1232–1240. [Google Scholar] [CrossRef] [Green Version]
  113. Minten, I.J.; Hendriks, L.J.A.; Nolte, R.J.M.; Cornelissen, J.J.L.M. Controlled Encapsulation of Multiple Proteins in Virus Capsids. J. Am. Chem. Soc. 2009, 131, 17771–17773. [Google Scholar] [CrossRef] [PubMed]
  114. Steele, J.F.C.; Peyret, H.; Saunders, K.; Castells-Graells, R.; Marsian, J.; Meshcheriakova, Y.; Lomonossoff, G.P. Synthetic Plant Virology for Nanobiotechnology and Nanomedicine. WIREs Nanomed. Nanobiotechnol. 2017, 9, e1447. [Google Scholar] [CrossRef]
  115. Zhang, J.; Zhou, K.; Wang, Q. Tailoring the Self-Assembly Behaviors of Recombinant Tobacco Mosaic Virus by Rationally Introducing Covalent Bonding at the Protein–Protein Interface. Small 2016, 12, 4955–4959. [Google Scholar] [CrossRef] [PubMed]
  116. Lavelle, L.; Michel, J.-P.; Gingery, M. The Disassembly, Reassembly and Stability of CCMV Protein Capsids. J. Virol. Methods 2007, 146, 311–316. [Google Scholar] [CrossRef]
  117. Li, L.; Xu, C.; Zhang, W.; Secundo, F.; Li, C.; Zhang, Z.-P.; Zhang, X.-E.; Li, F. Cargo-Compatible Encapsulation in Virus-Based Nanoparticles. Nano Lett. 2019, 19, 2700–2706. [Google Scholar] [CrossRef]
  118. Miao, Y.; Johnson, J.E.; Ortoleva, P.J. All-Atom Multiscale Simulation of Cowpea Chlorotic Mottle Virus Capsid Swelling. J. Phys. Chem. B 2010, 114, 11181–11195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Loo, L.; Guenther, R.H.; Lommel, S.A.; Franzen, S. Infusion of Dye Molecules into Red Clover Necrotic Mosaic Virus. Chem. Commun. 2008, 1, 88–90. [Google Scholar] [CrossRef]
  120. Aljabali, A.A.; Alzoubi, L.; Hamzat, Y.; Alqudah, A.; Obeid, M.A.; Al Zoubi, M.S.; Ennab, R.M.; Alshaer, W.; Albatayneh, K.; Al-Trad, B.; et al. A Potential MRI Agent and an Anticancer Drug Encapsulated within CPMV Virus-Like Particles. Comb. Chem. High Throughput Screen. 2021, 24, 1557–1571. [Google Scholar] [CrossRef] [PubMed]
  121. Aljabali, A.A.; Hassan, S.S.; Pabari, R.M.; Shahcheraghi, S.H.; Mishra, V.; Charbe, N.B.; Chellappan, D.K.; Dureja, H.; Gupta, G.; Almutary, A.G.; et al. The Viral Capsid as Novel Nanomaterials for Drug Delivery. Future Sci. OA 2021, 7, FSO744. [Google Scholar] [CrossRef] [PubMed]
  122. Cao, J.; Guenther, R.H.; Sit, T.L.; Opperman, C.H.; Lommel, S.A.; Willoughby, J.A. Loading and Release Mechanism of Red Clover Necrotic Mosaic Virus Derived Plant Viral Nanoparticles for Drug Delivery of Doxorubicin. Small 2014, 10, 5126–5136. [Google Scholar] [CrossRef] [PubMed]
  123. Sherman, M.B.; Guenther, R.H.; Tama, F.; Sit, T.L.; Brooks, C.L.; Mikhailov, A.M.; Orlova, E.V.; Baker, T.S.; Lommel, S.A. Removal of Divalent Cations Induces Structural Transitions in Red Clover Necrotic Mosaic Virus, Revealing a Potential Mechanism for RNA Release. J. Virol. 2006, 80, 10395–10406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Zhang, Y.; Dong, Y.; Zhou, J.; Li, X.; Wang, F. Application of Plant Viruses as a Biotemplate for Nanomaterial Fabrication. Molecules 2018, 23, 2311. [Google Scholar] [CrossRef]
  125. Arcangeli, C.; Circelli, P.; Donini, M.; Aljabali, A.A.A.; Benvenuto, E.; Lomonossoff, G.P.; Marusic, C. Structure-Based Design and Experimental Engineering of a Plant Virus Nanoparticle for the Presentation of Immunogenic Epitopes and as a Drug Carrier. J. Biomol. Struct. Dyn. 2014, 32, 630–647. [Google Scholar] [CrossRef]
  126. Alemzadeh, E.; Izadpanah, K.; Ahmadi, F. Generation of Recombinant Protein Shells of Johnson Grass Chlorotic Stripe Mosaic Virus in Tobacco Plants and Their Use as Drug Carrier. J. Virol. Methods 2017, 248, 148–153. [Google Scholar] [CrossRef] [PubMed]
  127. Chung, Y.H.; Cai, H.; Steinmetz, N.F. Viral Nanoparticles for Drug Delivery, Imaging, Immunotherapy, and Theranostic Applications. Adv. Drug Deliv. Rev. 2020, 156, 214–235. [Google Scholar] [CrossRef]
  128. Steinmetz, N.F.; Manchester, M. Viral Nanoparticles: Tools for Materials Science & Biomedicine; Jenny Stanford Publishing: New York, NY, USA, 2019; ISBN 978-0-429-06745-7. [Google Scholar]
  129. Esfandiari, N.; Arzanani, M.K.; Soleimani, M.; Kohi-Habibi, M.; Svendsen, W.E. A New Application of Plant Virus Nanoparticles as Drug Delivery in Breast Cancer. Tumour Biol. 2016, 37, 1229–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Masarapu, H.; Patel, B.K.; Chariou, P.L.; Hu, H.; Gulati, N.M.; Carpenter, B.L.; Ghiladi, R.A.; Shukla, S.; Steinmetz, N.F. Physalis Mottle Virus-Like Particles as Nanocarriers for Imaging Reagents and Drugs. Biomacromolecules 2017, 18, 4141–4153. [Google Scholar] [CrossRef] [Green Version]
  131. Baxevanis, C.N.; Perez, S.A.; Papamichail, M. Combinatorial Treatments Including Vaccines, Chemotherapy and Monoclonal Antibodies for Cancer Therapy. Cancer Immunol. Immunother. 2009, 58, 317–324. [Google Scholar] [CrossRef]
  132. Lockney, D.M.; Guenther, R.N.; Loo, L.; Overton, W.; Antonelli, R.; Clark, J.; Hu, M.; Luft, C.; Lommel, S.A.; Franzen, S. The Red Clover Necrotic Mosaic Virus Capsid as a Multifunctional Cell Targeting Plant Viral Nanoparticle. Bioconjug. Chem. 2011, 22, 67–73. [Google Scholar] [CrossRef]
  133. Tian, Y.; Zhou, M.; Shi, H.; Gao, S.; Xie, G.; Zhu, M.; Wu, M.; Chen, J.; Niu, Z. Integration of Cell-Penetrating Peptides with Rod-like Bionanoparticles: Virus-Inspired Gene-Silencing Technology. Nano Lett. 2018, 18, 5453–5460. [Google Scholar] [CrossRef] [PubMed]
  134. Liu, X.; Wu, F.; Tian, Y.; Wu, M.; Zhou, Q.; Jiang, S.; Niu, Z. Size Dependent Cellular Uptake of Rod-like Bionanoparticles with Different Aspect Ratios. Sci. Rep. 2016, 6, 24567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Lin, R.D.; Steinmetz, N.F. Tobacco Mosaic Virus Delivery of Mitoxantrone for Cancer Therapy. Nanoscale 2018, 10, 16307–16313. [Google Scholar] [CrossRef]
  136. Le, D.H.T.; Commandeur, U.; Steinmetz, N.F. Presentation and Delivery of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand via Elongated Plant Viral Nanoparticle Enhances Antitumor Efficacy. ACS Nano 2019, 13, 2501–2510. [Google Scholar] [CrossRef] [PubMed]
  137. Rudin, M.; Weissleder, R. Molecular Imaging in Drug Discovery and Development. Nat. Rev. Drug Discov. 2003, 2, 123–131. [Google Scholar] [CrossRef]
  138. Jung, B.; Rao, A.L.N.; Anvari, B. Optical Nano-Constructs Composed of Genome-Depleted Brome Mosaic Virus Doped with a near Infrared Chromophore for Potential Biomedical Applications. ACS Nano 2011, 5, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  139. Röder, J.; Dickmeis, C.; Fischer, R.; Commandeur, U. Systemic Infection of Nicotiana Benthamiana with Potato Virus X Nanoparticles Presenting a Fluorescent ILOV Polypeptide Fused Directly to the Coat Protein. BioMed Res. Int. 2018, 2018, 9328671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Shukla, S.; Dickmeis, C.; Nagarajan, A.S.; Fischer, R.; Commandeur, U.; Steinmetz, N.F. Molecular Farming of Fluorescent Virus-Based Nanoparticles for Optical Imaging in Plants, Human Cells and Mouse Models. Biomater. Sci. 2014, 2, 784–797. [Google Scholar] [CrossRef] [PubMed]
  141. Bruckman, M.A.; Jiang, K.; Simpson, E.J.; Randolph, L.N.; Luyt, L.G.; Yu, X.; Steinmetz, N.F. Dual-Modal Magnetic Resonance and Fluorescence Imaging of Atherosclerotic Plaques in Vivo Using VCAM-1 Targeted Tobacco Mosaic Virus. Nano Lett. 2014, 14, 1551–1558. [Google Scholar] [CrossRef]
  142. Lewis, J.D.; Destito, G.; Zijlstra, A.; Gonzalez, M.J.; Quigley, J.P.; Manchester, M.; Stuhlmann, H. Viral Nanoparticles as Tools for Intravital Vascular Imaging. Nat. Med. 2006, 12, 354–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Huang, X.; Stein, B.D.; Cheng, H.; Malyutin, A.; Tsvetkova, I.B.; Baxter, D.V.; Remmes, N.B.; Verchot, J.; Kao, C.; Bronstein, L.M.; et al. Magnetic Virus-like Nanoparticles in N. Benthamiana Plants: A New Paradigm for Environmental and Agronomic Biotechnological Research. ACS Nano 2011, 5, 4037–4045. [Google Scholar] [CrossRef] [Green Version]
  144. Davidson, B.L.; McCray, P.B. Current Prospects for RNA Interference-Based Therapies. Nat. Rev. Genet. 2011, 12, 329–340. [Google Scholar] [CrossRef] [PubMed]
  145. Karlsson, J.; Luly, K.M.; Tzeng, S.Y.; Green, J.J. Nanoparticle Designs for Delivery of Nucleic Acid Therapeutics as Brain Cancer Therapies. Adv. Drug Deliv. Rev. 2021, 179, 113999. [Google Scholar] [CrossRef] [PubMed]
  146. Padda, I.S.; Mahtani, A.U.; Parmar, M. Small Interfering RNA (SiRNA) Based Therapy. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  147. Lundstrom, K. Viral Vectors in Gene Therapy. Diseases 2018, 6, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Yan, Y.; Liu, X.-Y.; Lu, A.; Wang, X.-Y.; Jiang, L.-X.; Wang, J.-C. Non-Viral Vectors for RNA Delivery. J. Control. Release 2022, 342, 241–279. [Google Scholar] [CrossRef] [PubMed]
  149. Yin, H.; Kanasty, R.L.; Eltoukhy, A.A.; Vegas, A.J.; Dorkin, J.R.; Anderson, D.G. Non-Viral Vectors for Gene-Based Therapy. Nat. Rev. Genet. 2014, 15, 541–555. [Google Scholar] [CrossRef] [PubMed]
  150. Hacein-Bey-Abina, S.; Garrigue, A.; Wang, G.P.; Soulier, J.; Lim, A.; Morillon, E.; Clappier, E.; Caccavelli, L.; Delabesse, E.; Beldjord, K.; et al. Insertional Oncogenesis in 4 Patients after Retrovirus-Mediated Gene Therapy of SCID-X1. J. Clin. Invest. 2008, 118, 3132–3142. [Google Scholar] [CrossRef]
  151. Vorburger, S.A.; Hunt, K.K. Adenoviral Gene Therapy. Oncologist 2002, 7, 46–59. [Google Scholar] [CrossRef] [PubMed]
  152. Zaiss, A.K.; Machado, H.B.; Herschman, H.R. The Influence of Innate and Pre-Existing Immunity on Adenovirus Therapy. J. Cell. Biochem. 2009, 108, 778–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Steinmetz, N.F. Viral Nanoparticles as Platforms for Next-Generation Therapeutics and Imaging Devices. Nanomedicine 2010, 6, 634–641. [Google Scholar] [CrossRef] [Green Version]
  154. Alemzadeh, E.; Dehshahri, A.; Izadpanah, K.; Ahmadi, F. Plant Virus Nanoparticles: Novel and Robust Nanocarriers for Drug Delivery and Imaging. Colloids Surf. B Biointerfaces 2018, 167, 20–27. [Google Scholar] [CrossRef]
  155. Cai, H.; Shukla, S.; Steinmetz, N.F. The Antitumor Efficacy of CpG Oligonucleotides Is Improved by Encapsulation in Plant Virus-Like Particles. Adv. Funct. Mater. 2020, 30, 1908743. [Google Scholar] [CrossRef]
  156. Peyret, H.; Groppelli, E.; Clark, D.; Eckersley, N.; Planche, T.; Ma, J.; Lomonossoff, G.P. Production and Use of Encapsidated RNA Mimics as Positive Control Reagents for SARS-CoV-2 RT-QPCR Diagnostics. J. Virol. Methods 2022, 300, 114372. [Google Scholar] [CrossRef] [PubMed]
  157. Azizgolshani, O.; Garmann, R.F.; Cadena-Nava, R.; Knobler, C.M.; Gelbart, W.M. Reconstituted Plant Viral Capsids Can Release Genes to Mammalian Cells. Virology 2013, 441, 12–17. [Google Scholar] [CrossRef] [Green Version]
  158. Villagrana-Escareño, M.V.; Reynaga-Hernández, E.; Galicia-Cruz, O.G.; Durán-Meza, A.L.; De la Cruz-González, V.; Hernández-Carballo, C.Y.; Ruíz-García, J. VLPs Derived from the CCMV Plant Virus Can Directly Transfect and Deliver Heterologous Genes for Translation into Mammalian Cells. BioMed Res. Int. 2019, 2019, e4630891. [Google Scholar] [CrossRef]
  159. Lam, P.; Steinmetz, N.F. Delivery of SiRNA Therapeutics Using Cowpea Chlorotic Mottle Virus-like Particles. Biomater. Sci. 2019, 7, 3138–3142. [Google Scholar] [CrossRef]
  160. Biddlecome, A.; Habte, H.H.; McGrath, K.M.; Sambanthamoorthy, S.; Wurm, M.; Sykora, M.M.; Knobler, C.M.; Lorenz, I.C.; Lasaro, M.; Elbers, K.; et al. Delivery of Self-Amplifying RNA Vaccines in in Vitro Reconstituted Virus-like Particles. PLoS ONE 2019, 14, e0215031. [Google Scholar] [CrossRef] [Green Version]
  161. Lam, P.; Gulati, N.M.; Stewart, P.L.; Keri, R.A.; Steinmetz, N.F. Bioengineering of Tobacco Mosaic Virus to Create a Non-Infectious Positive Control for Ebola Diagnostic Assays. Sci. Rep. 2016, 6, 23803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Li, L.; Wang, L.; Xiao, R.; Zhu, G.; Li, Y.; Liu, C.; Yang, R.; Tang, Z.; Li, J.; Huang, W.; et al. The Invasion of Tobacco Mosaic Virus RNA Induces Endoplasmic Reticulum Stress-Related Autophagy in HeLa Cells. Biosci. Rep. 2012, 32, 171–184. [Google Scholar] [CrossRef] [Green Version]
  163. Loo, L.; Guenther, R.H.; Lommel, S.A.; Franzen, S. Encapsidation of Nanoparticles by Red Clover Necrotic Mosaic Virus. J. Am. Chem. Soc. 2007, 129, 11111–11117. [Google Scholar] [CrossRef]
  164. Wu, Y.; Yang, H.; Shin, H.-J. Encapsulation and Crystallization of Prussian Blue Nanoparticles by Cowpea Chlorotic Mottle Virus Capsids. Biotechnol. Lett. 2014, 36, 515–521. [Google Scholar] [CrossRef]
  165. Basu, G.; Allen, M.; Willits, D.; Young, M.; Douglas, T. Metal Binding to Cowpea Chlorotic Mottle Virus Using Terbium(III) Fluorescence. J. Biol. Inorg. Chem. 2003, 8, 721–725. [Google Scholar] [CrossRef] [PubMed]
  166. Allen, M.; Bulte, J.W.M.; Liepold, L.; Basu, G.; Zywicke, H.A.; Frank, J.A.; Young, M.; Douglas, T. Paramagnetic Viral Nanoparticles as Potential High-Relaxivity Magnetic Resonance Contrast Agents. Magn. Reason. Med. 2005, 54, 807–812. [Google Scholar] [CrossRef] [PubMed]
  167. Balci, S.; Bittner, A.M.; Hahn, K.; Scheu, C.; Knez, M.; Kadri, A.; Wege, C.; Jeske, H.; Kern, K. Copper Nanowires within the Central Channel of Tobacco Mosaic Virus Particles. Electrochim. Acta 2006, 51, 6251–6257. [Google Scholar] [CrossRef]
  168. Dujardin, E.; Peet, C.; Stubbs, G.; Culver, J.N.; Mann, S. Organization of Metallic Nanoparticles Using Tobacco Mosaic Virus Templates. Nano Lett. 2003, 3, 413–417. [Google Scholar] [CrossRef]
  169. Knez, M.; Bittner, A.M.; Boes, F.; Wege, C.; Jeske, H.; Maiß, E.; Kern, K. Biotemplate Synthesis of 3-Nm Nickel and Cobalt Nanowires. Nano Lett. 2003, 3, 1079–1082. [Google Scholar] [CrossRef] [Green Version]
  170. Kobayashi, M.; Seki, M.; Tabata, H.; Watanabe, Y.; Yamashita, I. Fabrication of Aligned Magnetic Nanoparticles Using Tobamoviruses. Nano Lett. 2010, 10, 773–776. [Google Scholar] [CrossRef] [PubMed]
  171. Aljabali, A.A.A.; Lomonossoff, G.P.; Evans, D.J. CPMV-Polyelectrolyte-Templated Gold Nanoparticles. Biomacromolecules 2011, 12, 2723–2728. [Google Scholar] [CrossRef] [PubMed]
  172. Culver, J.N.; Brown, A.D.; Zang, F.; Gnerlich, M.; Gerasopoulos, K.; Ghodssi, R. Plant Virus Directed Fabrication of Nanoscale Materials and Devices. Virology 2015, 479–480, 200–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Yang, C.; Meldon, J.H.; Lee, B.; Yi, H. Investigation on the Catalytic Reduction Kinetics of Hexavalent Chromium by Viral-Templated Palladium Nanocatalysts. Catal. Today 2014, 233, 108–116. [Google Scholar] [CrossRef]
  174. Shahgolzari, M.; Pazhouhandeh, M.; Milani, M.; Yari Khosroushahi, A.; Fiering, S. Plant Viral Nanoparticles for Packaging and in Vivo Delivery of Bioactive Cargos. WIREs Nanomed. Nanobiotechnol. 2020, 12, e1629. [Google Scholar] [CrossRef] [PubMed]
  175. Bäcker, M.; Koch, C.; Eiben, S.; Geiger, F.; Eber, F.; Gliemann, H.; Poghossian, A.; Wege, C.; Schöning, M.J. A New Class of Biosensors Based on Tobacco Mosaic Virus and Coat Proteins as Enzyme Nanocarrier. Procedia Eng. 2016, 168, 618–621. [Google Scholar] [CrossRef]
  176. Koch, C.; Wabbel, K.; Eber, F.J.; Krolla-Sidenstein, P.; Azucena, C.; Gliemann, H.; Eiben, S.; Geiger, F.; Wege, C. Modified TMV Particles as Beneficial Scaffolds to Present Sensor Enzymes. Front. Plant Sci. 2015, 6, 1137. [Google Scholar] [CrossRef] [PubMed]
  177. Koch, C.; Poghossian, A.; Schöning, M.J.; Wege, C. Penicillin Detection by Tobacco Mosaic Virus-Assisted Colorimetric Biosensors. Nanotheranostics 2018, 2, 184–196. [Google Scholar] [CrossRef] [Green Version]
  178. Cuenca, S.; Mansilla, C.; Aguado, M.; Yuste-Calvo, C.; Sánchez, F.; Sánchez-Montero, J.M.; Ponz, F. Nanonets Derived from Turnip Mosaic Virus as Scaffolds for Increased Enzymatic Activity of Immobilized Candida Antarctica Lipase B. Front. Plant Sci. 2016, 7, 464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Sánchez-Sánchez, L.; Cadena-Nava, R.D.; Palomares, L.A.; Ruiz-Garcia, J.; Koay, M.S.T.; Cornelissen, J.J.M.T.; Vazquez-Duhalt, R. Chemotherapy Pro-Drug Activation by Biocatalytic Virus-like Nanoparticles Containing Cytochrome P450. Enzym. Microb. Technol. 2014, 60, 24–31. [Google Scholar] [CrossRef]
  180. Grilo, A.L.; Mantalaris, A. The Increasingly Human and Profitable Monoclonal Antibody Market. Trends Biotechnol. 2019, 37, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Monoclonal Antibodies Market to Hit USD 425 Billion by 2028, Says Global Market Insights Inc. 2022. Available online: Bloomberg.com (accessed on 17 May 2022).
  182. Ma, J.K.-C.; Hiatt, A.; Hein, M.; Vine, N.D.; Wang, F.; Stabila, P.; van Dolleweerd, C.; Mostov, K.; Lehner, T. Generation and Assembly of Secretory Antibodies in Plants. Science 1995, 268, 716–719. [Google Scholar] [CrossRef]
  183. Diamos, A.G.; Hunter, J.G.L.; Pardhe, M.D.; Rosenthal, S.H.; Sun, H.; Foster, B.C.; DiPalma, M.P.; Chen, Q.; Mason, H.S. High Level Production of Monoclonal Antibodies Using an Optimized Plant Expression System. Front. Bioeng. Biotechnol. 2019, 7, 474. [Google Scholar] [CrossRef]
  184. Malaquias, A.D.M.; Marques, L.E.C.; Pereira, S.S.; de Freitas Fernandes, C.; Maranhão, A.Q.; Stabeli, R.G.; Florean, E.O.P.T.; Guedes, M.I.F.; Fernandes, C.F.C. A Review of Plant-Based Expression Systems as a Platform for Single-Domain Recombinant Antibody Production. Int. J. Biol. Macromol. 2021, 193, 1130–1137. [Google Scholar] [CrossRef] [PubMed]
  185. Edgue, G.; Twyman, R.M.; Beiss, V.; Fischer, R.; Sack, M. Antibodies from Plants for Bionanomaterials. WIREs Nanomed. Nanobiotechnol. 2017, 9, e1462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Smolenska, L.; Roberts, I.M.; Learmonth, D.; Porter, A.J.; Harris, W.J.; Wilson, T.M.A.; Cruz, S.S. Production of a Functional Single Chain Antibody Attached to the Surface of a Plant Virus. FEBS Lett. 1998, 441, 379–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Werner, S.; Marillonnet, S.; Hause, G.; Klimyuk, V.; Gleba, Y. Immunoabsorbent Nanoparticles Based on a Tobamovirus Displaying Protein A. Proc. Natl. Acad. Sci. USA 2006, 103, 17678–17683. [Google Scholar] [CrossRef]
  188. Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hamers, C.; Songa, E.B.; Bendahman, N.; Hamers, R. Naturally Occurring Antibodies Devoid of Light Chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef] [PubMed]
  189. Mitchell, L.S.; Colwell, L.J. Comparative Analysis of Nanobody Sequence and Structure Data. Proteins Struct. Funct. Bioinform. 2018, 86, 697–706. [Google Scholar] [CrossRef]
  190. Martí, M.; Merwaiss, F.; Butković, A.; Daròs, J.-A. Production of Potyvirus-Derived Nanoparticles Decorated with a Nanobody in Biofactory Plants. Front. Bioeng. Biotechnol. 2022, 10, 877363. [Google Scholar] [CrossRef] [PubMed]
  191. Mohsen, M.O.; Gomes, A.C.; Vogel, M.; Bachmann, M.F. Interaction of Viral Capsid-Derived Virus-Like Particles (VLPs) with the Innate Immune System. Vaccines 2018, 6, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Lester, S.N.; Li, K. Toll-like Receptors in Antiviral Innate Immunity. J. Mol. Biol. 2014, 426, 1246–1264. [Google Scholar] [CrossRef] [PubMed]
  193. Mao, C.; Beiss, V.; Fields, J.; Steinmetz, N.F.; Fiering, S. Cowpea Mosaic Virus Stimulates Antitumor Immunity through Recognition by Multiple MYD88-Dependent Toll-like Receptors. Biomaterials 2021, 275, 120914. [Google Scholar] [CrossRef] [PubMed]
  194. Reid, E.; Suneja, G.; Ambinder, R.F.; Ard, K.; Baiocchi, R.; Barta, S.K.; Carchman, E.; Cohen, A.; Gupta, N.; Johung, K.L.; et al. Cancer in People Living With HIV, Version 1.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2018, 16, 986–1017. [Google Scholar] [CrossRef] [Green Version]
  195. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune Evasion in Cancer: Mechanistic Basis and Therapeutic Strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef] [PubMed]
  196. Khong, H.T.; Restifo, N.P. Natural Selection of Tumor Variants in the Generation of “Tumor Escape” Phenotypes. Nat. Immunol. 2002, 3, 999–1005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Shukla, S.; Roe, A.J.; Liu, R.; Veliz, F.A.; Commandeur, U.; Wald, D.N.; Steinmetz, N.F. Affinity of Plant Viral Nanoparticle Potato Virus X (PVX) towards Malignant B Cells Enables Cancer Drug Delivery. Biomater. Sci. 2020, 8, 3935–3943. [Google Scholar] [CrossRef] [PubMed]
  198. Jobsri, J.; Allen, A.; Rajagopal, D.; Shipton, M.; Kanyuka, K.; Lomonossoff, G.P.; Ottensmeier, C.; Diebold, S.S.; Stevenson, F.K.; Savelyeva, N. Plant Virus Particles Carrying Tumour Antigen Activate TLR7 and Induce High Levels of Protective Antibody. PLoS ONE 2015, 10, e0118096. [Google Scholar] [CrossRef]
  199. Tyulkina, L.G.; Skurat, E.V.; Frolova, O.Y.; Komarova, T.V.; Karger, E.M.; Atabekov, I.G. New Viral Vector for Superproduction of Epitopes of Vaccine Proteins in Plants. Acta Nat. 2011, 3, 73–82. [Google Scholar] [CrossRef]
  200. Esfandiari, N. Targeting Breast Cancer With Bio-Inspired Virus Nanoparticles. Arch Breast Cancer 2018, 5, 90–95. [Google Scholar] [CrossRef]
  201. Esfandiari, N.; Arzanani, M.K.; Koohi-Habibi, M. The Study of Toxicity and Pathogenicity Risk of Potato Virus X/Herceptin Nanoparticles as Agents for Cancer Therapy. Cancer Nanotechnol. 2018, 9, 1. [Google Scholar] [CrossRef] [Green Version]
  202. Steinmetz, N.F.; Cho, C.-F.; Ablack, A.; Lewis, J.D.; Manchester, M. Cowpea Mosaic Virus Nanoparticles Target Surface Vimentin on Cancer Cells. Nanomedicine 2011, 6, 351–364. [Google Scholar] [CrossRef] [Green Version]
  203. Wang, C.; Beiss, V.; Steinmetz, N.F. Cowpea Mosaic Virus Nanoparticles and Empty Virus-Like Particles Show Distinct but Overlapping Immunostimulatory Properties. J. Virol. 2019, 93, e00129-19. [Google Scholar] [CrossRef] [PubMed]
  204. Cai, H.; Shukla, S.; Wang, C.; Masarapu, H.; Steinmetz, N.F. Heterologous Prime-Boost Enhances the Antitumor Immune Response Elicited by Plant-Virus-Based Cancer Vaccine. J. Am. Chem. Soc. 2019, 141, 6509–6518. [Google Scholar] [CrossRef] [PubMed]
  205. Shukla, S.; Ablack, A.L.; Wen, A.M.; Lee, K.L.; Lewis, J.D.; Steinmetz, N.F. Increased Tumor Homing and Tissue Penetration of the Filamentous Plant Viral Nanoparticle Potato Virus X. Mol. Pharm. 2013, 10, 33–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Yin, Z.; Nguyen, H.G.; Chowdhury, S.; Bentley, P.; Bruckman, M.A.; Miermont, A.; Gildersleeve, J.C.; Wang, Q.; Huang, X. Tobacco Mosaic Virus as a New Carrier for Tumor Associated Carbohydrate Antigens. Bioconjug. Chem. 2012, 23, 1694–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Wang, C.; Steinmetz, N.F. CD47 Blockade and Cowpea Mosaic Virus Nanoparticle In Situ Vaccination Triggers Phagocytosis and Tumor Killing. Adv Health Mater 2019, 8, e1801288. [Google Scholar] [CrossRef] [PubMed]
  208. Nicholas, B.L.; Brennan, F.R.; Martinez-Torrecuadrada, J.L.; Casal, J.I.; Hamilton, W.D.; Wakelin, D. Characterization of the Immune Response to Canine Parvovirus Induced by Vaccination with Chimaeric Plant Viruses. Vaccine 2002, 20, 2727–2734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Yusibov, V.; Rabindran, S.; Commandeur, U.; Twyman, R.M.; Fischer, R. The Potential of Plant Virus Vectors for Vaccine Production. Drugs R D 2006, 7, 203–217. [Google Scholar] [CrossRef] [PubMed]
  210. Staczek, J.; Bendahmane, M.; Gilleland, L.B.; Beachy, R.N.; Gilleland, H.E. Immunization with a Chimeric Tobacco Mosaic Virus Containing an Epitope of Outer Membrane Protein F of Pseudomonas Aeruginosa Provides Protection against Challenge with P. Aeruginosa. Vaccine 2000, 18, 2266–2274. [Google Scholar] [CrossRef]
  211. Bendahmane, M.; Koo, M.; Karrer, E.; Beachy, R.N. Display of Epitopes on the Surface of Tobacco Mosaic Virus: Impact of Charge and Isoelectric Point of the Epitope on Virus-Host Interactions. J. Mol. Biol. 1999, 290, 9–20. [Google Scholar] [CrossRef]
  212. Porta, C.; Spall, V.E.; Findlay, K.C.; Gergerich, R.C.; Farrance, C.E.; Lomonossoff, G.P. 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. [Google Scholar] [CrossRef] [Green Version]
  213. Uhde-Holzem, K.; McBurney, M.; Tiu, B.D.B.; Advincula, R.C.; Fischer, R.; Commandeur, U.; Steinmetz, N.F. Production of Immunoabsorbent Nanoparticles by Displaying Single-Domain Protein A on Potato Virus X. Macromol. Biosci. 2016, 16, 231–241. [Google Scholar] [CrossRef]
  214. Carignan, D.; Thérien, A.; Rioux, G.; Paquet, G.; Gagné, M.-È.L.; Bolduc, M.; Savard, P.; Leclerc, D. Engineering of the PapMV Vaccine Platform with a Shortened M2e Peptide Leads to an Effective One Dose Influenza Vaccine. Vaccine 2015, 33, 7245–7253. [Google Scholar] [CrossRef]
  215. Balke, I.; Zeltins, A. Recent Advances in the Use of Plant Virus-Like Particles as Vaccines. Viruses 2020, 12, 270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Lomonossoff, G.P.; Hamilton, W.D.O. Cowpea Mosaic Virus-Based Vaccines. In Plant Biotechnology: New Products and Applications; Hammond, J., McGarvey, P., Yusibov, V., Eds.; Current Topics in Microbiology and Immunology; Springer: Berlin/Heidelberg, Germany, 2000; pp. 177–189. ISBN 978-3-642-60234-4. [Google Scholar]
  217. Haynes, J.R.; Cunningham, J.; von Seefried, A.; Lennick, M.; Garvin, R.T.; Shen, S.-H. Development of a Genetically-Engineered, Candidate Polio Vaccine Employing the Self-Assembling Properties of the Tobacco Mosaic Virus Coat Protein. Biotechnology 1986, 4, 637–641. [Google Scholar] [CrossRef] [PubMed]
  218. Usha, R.; Rohll, J.B.; Spall, V.E.; Shanks, M.; Maule, A.J.; Johnson, J.E.; Lomonossoff, G.P. Expression of an Animal Virus Antigenic Site on the Surface of a Plant Virus Particle. Virology 1993, 197, 366–374. [Google Scholar] [CrossRef]
  219. Turpen, T.H.; Reinl, S.J.; Charoenvit, Y.; Hoffman, S.L.; Fallarme, V.; Grill, L.K. Malaria Epitopes Expressed on the Surface of Recombinant Tobacco Mosaic Virus. Nat. Biotechnol. 1995, 13, 53–57. [Google Scholar] [CrossRef] [PubMed]
  220. Sugiyama, Y.; Hamamoto, H.; Takemoto, S.; Watanabe, Y.; Okada, Y. Systemic Production of Foreign Peptides on the Particle Surface of Tobacco Mosaic Virus. FEBS Lett. 1995, 359, 247–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  221. Lomonossoff, G.P.; Johnson, J.E. Use of Macromolecular Assemblies as Expression Systems for Peptides and Synthetic Vaccines. Curr. Opin. Struct. Biol. 1996, 6, 176–182. [Google Scholar] [CrossRef] [PubMed]
  222. Embregts, C.W.; Forlenza, M. Oral Vaccination of Fish: Lessons from Humans and Veterinary Species. Dev. Comp. Immunol. 2016, 64, 118–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Chen, Q. Expression and Purification of Pharmaceutical Proteins in Plants. Biol. Eng. Trans. 2008, 1, 291–321. [Google Scholar] [CrossRef]
  224. Lai, H.; Chen, Q. Bioprocessing of Plant-Derived Virus-like Particles of Norwalk Virus Capsid Protein under Current Good Manufacture Practice Regulations. Plant Cell Rep. 2012, 31, 573–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Chen, Q.; Lai, H. Plant-Derived Virus-like Particles as Vaccines. Hum. Vaccines Immunother. 2013, 9, 26–49. [Google Scholar] [CrossRef] [PubMed]
  226. Potter, A.; Gerdts, V. Veterinary Vaccines: Alternatives to Antibiotics? Anim. Health Res. Rev. 2008, 9, 187–199. [Google Scholar] [CrossRef]
  227. Chen, T.-H.; Hu, C.-C.; Liao, J.-T.; Lee, Y.-L.; Huang, Y.-W.; Lin, N.-S.; Lin, Y.-L.; Hsu, Y.-H. Production of Japanese Encephalitis Virus Antigens in Plants Using Bamboo Mosaic Virus-Based Vector. Front. Microbiol. 2017, 8, 788. [Google Scholar] [CrossRef] [PubMed]
  228. Kolotilin, I.; Topp, E.; Cox, E.; Devriendt, B.; Conrad, U.; Joensuu, J.; Stöger, E.; Warzecha, H.; McAllister, T.; Potter, A.; et al. Plant-Based Solutions for Veterinary Immunotherapeutics and Prophylactics. Vet. Res. 2014, 45, 117. [Google Scholar] [CrossRef] [PubMed]
  229. Zahmanova, G.; Takova, K.; Valkova, R.; Toneva, V.; Minkov, I.; Andonov, A.; Lukov, G.L. Plant-Derived Recombinant Vaccines against Zoonotic Viruses. Life 2022, 12, 156. [Google Scholar] [CrossRef] [PubMed]
  230. Jacob, S.S.; Cherian, S.; Sumithra, T.G.; Raina, O.K.; Sankar, M. Edible Vaccines against Veterinary Parasitic Diseases—Current Status and Future Prospects. Vaccine 2013, 31, 1879–1885. [Google Scholar] [CrossRef]
  231. Su, H.; Yakovlev, I.A.; van Eerde, A.; Su, J.; Clarke, J.L. Plant-Produced Vaccines: Future Applications in Aquaculture. Front. Plant Sci. 2021, 12, 718775. [Google Scholar] [CrossRef] [PubMed]
  232. Joensuu, J.J.; Niklander-Teeri, V.; Brandle, J.E. Transgenic Plants for Animal Health: Plant-Made Vaccine Antigens for Animal Infectious Disease Control. Phytochem. Rev. 2008, 7, 553–577. [Google Scholar] [CrossRef]
  233. Howard, J.A. Commercialization of Plant-Based Vaccines from Research and Development to Manufacturing. Anim. Health Res. Rev. 2004, 5, 243–245. [Google Scholar] [CrossRef]
  234. Sahoo, A.; Mandal, A.K.; Dwivedi, K.; Kumar, V. A Cross Talk between the Immunization and Edible Vaccine: Current Challenges and Future Prospects. Life Sci. 2020, 261, 118343. [Google Scholar] [CrossRef]
  235. Mičúchová, A.; Piačková, V.; Frébort, I.; Korytář, T. Molecular Farming: Expanding the Field of Edible Vaccines for Sustainable Fish Aquaculture. Rev. Aquac. 2022, 14, 1978–2001. [Google Scholar] [CrossRef]
  236. Specht, E.; Mayfield, S. Algae-Based Oral Recombinant Vaccines. Front. Microbiol. 2014, 5, 60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  237. Park, E.-J.; Kim, M.-N.; Park, J.-Y.; Cha, J.-H.; Chung, H.-J. Edible vaccine for aquacultured fish: Present and prospect. J. Plant Biotechnol. 2010, 37, 269–274. [Google Scholar] [CrossRef] [Green Version]
  238. Ma, K.; Bao, Q.; Wu, Y.; Chen, S.; Zhao, S.; Wu, H.; Fan, J. Evaluation of Microalgae as Immunostimulants and Recombinant Vaccines for Diseases Prevention and Control in Aquaculture. Front. Bioeng. Biotechnol. 2020, 8, 590431. [Google Scholar] [CrossRef]
  239. Rybicki, E. History and Promise of Plant-Made Vaccines for Animals. In Prospects of Plant-Based Vaccines in Veterinary Medicine; MacDonald, J., Ed.; Springer International Publishing: Cham, Germany, 2018; pp. 1–22. ISBN 978-3-319-90137-4. [Google Scholar]
  240. Zhang, X.; Buehner, N.A.; Hutson, A.M.; Estes, M.K.; Mason, H.S. Tomato Is a Highly Effective Vehicle for Expression and Oral Immunization with Norwalk Virus Capsid Protein. Plant Biotechnol. J. 2006, 4, 419–432. [Google Scholar] [CrossRef]
  241. Tacket, C.O.; Mason, H.S.; Losonsky, G.; Estes, M.K.; Levine, M.M.; Arntzen, C.J. Human Immune Responses to a Novel Norwalk Virus Vaccine Delivered in Transgenic Potatoes. J. Infect. Dis. 2000, 182, 302–305. [Google Scholar] [CrossRef]
  242. Yusibov, V.; Hooper, D.C.; Spitsin, S.V.; Fleysh, N.; Kean, R.B.; Mikheeva, T.; Deka, D.; Karasev, A.; Cox, S.; Randall, J.; et al. Expression in Plants and Immunogenicity of Plant Virus-Based Experimental Rabies Vaccine. Vaccine 2002, 20, 3155–3164. [Google Scholar] [CrossRef] [PubMed]
  243. Hahn, B.-S.; Jeon, I.-S.; Jung, Y.-J.; Kim, J.-B.; Park, J.-S.; Ha, S.-H.; Kim, K.-H.; Kim, H.-M.; Yang, J.-S.; Kim, Y.-H. Expression of Hemagglutinin-Neuraminidase Protein of Newcastle Disease Virus in Transgenic Tobacco. Plant Biotechnol. Rep. 2007, 1, 85–92. [Google Scholar] [CrossRef]
  244. Guerrero-Andrade, O.; Loza-Rubio, E.; Olivera-Flores, T.; Fehérvári-Bone, T.; Gómez-Lim, M.A. Expression of the Newcastle Disease Virus Fusion Protein in Transgenic Maize and Immunological Studies. Transgenic Res. 2006, 15, 455–463. [Google Scholar] [CrossRef] [PubMed]
  245. Shahid, N.; Samiullah, T.R.; Shakoor, S.; Latif, A.; Yasmeen, A.; Azam, S.; Shahid, A.A.; Husnain, T.; Rao, A.Q. Early Stage Development of a Newcastle Disease Vaccine Candidate in Corn. Front. Vet. Sci. 2020, 7, 499. [Google Scholar] [CrossRef] [PubMed]
  246. Motamedi, M.J.; Ebrahimi, M.M.; Shahsavandi, S.; Amani, J.; Kazemi, R.; Jafari, M.; Salmanian, A.-H. The Immunogenicity of a Novel Chimeric Hemagglutinin-Neuraminidase-Fusion Antigen from Newcastle Disease Virus by Oral Delivery of Transgenic Canola Seeds to Chickens. Mol. Biotechnol. 2020, 62, 344–354. [Google Scholar] [CrossRef]
  247. Berinstein, A.; Vazquez-Rovere, C.; Asurmendi, S.; Gómez, E.; Zanetti, F.; Zabal, O.; Tozzini, A.; Conte Grand, D.; Taboga, O.; Calamante, G.; et al. Mucosal and Systemic Immunization Elicited by Newcastle Disease Virus (NDV) Transgenic Plants as Antigens. Vaccine 2005, 23, 5583–5589. [Google Scholar] [CrossRef] [PubMed]
  248. Dreesen, I.A.; Charpin-El Hamri, G.; Fussenegger, M. Heat-Stable Oral Alga-Based Vaccine Protects Mice from Staphylococcus Aureus Infection. J. Biotechnol. 2010, 145, 273–280. [Google Scholar] [CrossRef] [PubMed]
  249. Wigdorovitz, A.; Mozgovoj, M.; Santos, M.J.D.; Parreño, V.; Gómez, C.; Pérez-Filgueira, D.M.; Trono, K.G.; Ríos, R.D.; Franzone, P.M.; Fernández, F.; et al. Protective Lactogenic Immunity Conferred by an Edible Peptide Vaccine to Bovine Rotavirus Produced in Transgenic Plants. J. Gen. Virol. 2004, 85, 1825–1832. [Google Scholar] [CrossRef]
  250. Pogrebnyak, N.; Golovkin, M.; Andrianov, V.; Spitsin, S.; Smirnov, Y.; Egolf, R.; Koprowski, H. Severe Acute Respiratory Syndrome (SARS) S Protein Production in Plants: Development of Recombinant Vaccine. Proc. Natl. Acad. Sci. USA 2005, 102, 9062–9067. [Google Scholar] [CrossRef] [Green Version]
  251. Legocki, A.B.; Miedzinska, K.; Czaplińska, M.; Płucieniczak, A.; Wędrychowicz, H. Immunoprotective Properties of Transgenic Plants Expressing E2 Glycoprotein from CSFV and Cysteine Protease from Fasciola Hepatica. Vaccine 2005, 23, 1844–1846. [Google Scholar] [CrossRef]
  252. Plant-Based Biologics Market Size 2026 | Revised in a New Report. Available online: https://www.researchdive.com/150/plant-based-biologics-market (accessed on 5 December 2022).
  253. Peyret, H.; Brown, J.K.M.; Lomonossoff, G.P. Improving Plant Transient Expression through the Rational Design of Synthetic 5′ and 3′ Untranslated Regions. Plant Methods 2019, 15, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Montero-Morales, L.; Steinkellner, H. Advanced Plant-Based Glycan Engineering. Front. Bioeng. Biotechnol. 2018, 6, 81. [Google Scholar] [CrossRef]
  255. Drossard, J. Downstream Processing of Plant-Derived Recombinant Therapeutic Proteins. In Molecular Farming; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2004; pp. 217–231. ISBN 978-3-527-60363-3. [Google Scholar]
  256. Streatfield, S.J. Approaches to Achieve High-Level Heterologous Protein Production in Plants. Plant Biotechnol. J. 2007, 5, 2–15. [Google Scholar] [CrossRef]
  257. Jutras, P.V.; D’Aoust, M.-A.; Couture, M.M.-J.; Vézina, L.-P.; Goulet, M.-C.; Michaud, D.; Sainsbury, F. Modulating Secretory Pathway PH by Proton Channel Co-Expression Can Increase Recombinant Protein Stability in Plants. Biotechnol. J. 2015, 10, 1478–1486. [Google Scholar] [CrossRef] [PubMed]
  258. Jutras, P.V.; Dodds, I.; van der Hoorn, R.A. Proteases of Nicotiana Benthamiana: An Emerging Battle for Molecular Farming. Curr. Opin. Biotechnol. 2020, 61, 60–65. [Google Scholar] [CrossRef] [PubMed]
  259. Elelyso® for Gaucher Disease. Available online: https://protalix.com/about/elelyso/ (accessed on 5 December 2022).
  260. Tran, E.E.H.; Nelson, E.A.; Bonagiri, P.; Simmons, J.A.; Shoemaker, C.J.; Schmaljohn, C.S.; Kobinger, G.P.; Zeitlin, L.; Subramaniam, S.; White, J.M. Mapping of Ebolavirus Neutralization by Monoclonal Antibodies in the ZMapp Cocktail Using Cryo-Electron Tomography and Studies of Cellular Entry. J. Virol. 2016, 90, 7618–7627. [Google Scholar] [CrossRef] [Green Version]
  261. Reuters Staff Mapp Biopharma’s Ebola Drug Gets FDA Fast Track Status. Reuters 2015. Available online: https://www.reuters.com/article/mapp-biopharmaceutical-fda-idUSL4N11N4K520150917 (accessed on 17 November 2022).
  262. IBio Inc. IBio Reports Successful Preclinical Immunization Studies with Next-Gen Nucleocapsid COVID-19 Vaccine Candidate. Available online: https://ibioinc.com/ibio-reports-successful-preclinical-immunization-studies-with-next-gen-nucleocapsid-covid-19-vaccine-candidate/ (accessed on 20 November 2021).
  263. British American Tobacco—BAT Makes Progress on COVID-19 Vaccine Provides Community Support. Available online: https://www.bat.com/group/sites/UK__9D9KCY.nsf/vwPagesWebLive/DOBPMBZC# (accessed on 20 November 2021).
  264. Baiya Phytopharm Co Ltd.: Baiya SARS-CoV-2 Vax 1 Vaccine—COVID19 Vaccine Tracker. Available online: https://covid19.trackvaccines.org/vaccines/130/ (accessed on 20 November 2021).
  265. Hardy, A. IBIO-201 Demonstrates Ability to Elicit Anti-SARS-CoV-2 Immune Response in Preclinical Studies. Available online: https://biotuesdays.com/2020/08/10/ibio-updates-ibio-201-covid-19-vaccine-candidate/ (accessed on 20 November 2021).
  266. Hahn-Löbmann, S.; Stephan, A.; Schulz, S.; Schneider, T.; Shaverskyi, A.; Tusé, D.; Giritch, A.; Gleba, Y. Colicins and Salmocins —New Classes of Plant-Made Non-Antibiotic Food Antibacterials. Front. Plant Sci. 2019, 10, 437. [Google Scholar] [CrossRef]
  267. Starkevič, U.; Bortesi, L.; Virgailis, M.; Ružauskas, M.; Giritch, A.; Ražanskienė, A. High-Yield Production of a Functional Bacteriophage Lysin with Antipneumococcal Activity Using a Plant Virus-Based Expression System. J. Biotechnol. 2015, 200, 10–16. [Google Scholar] [CrossRef]
  268. Gomord, V.; Fitchette, A.-C.; Menu-Bouaouiche, L.; Saint-Jore-Dupas, C.; Plasson, C.; Michaud, D.; Faye, L. Plant-Specific Glycosylation Patterns in the Context of Therapeutic Protein Production. Plant Biotechnol. J. 2010, 8, 564–587. [Google Scholar] [CrossRef] [PubMed]
  269. Rosales-Mendoza, S.; Salazar-González, J.A.; Decker, E.L.; Reski, R. Implications of Plant Glycans in the Development of Innovative Vaccines. Expert Rev. Vaccines 2016, 15, 915–925. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biologics produced in plants.
Figure 1. Biologics produced in plants.
Ijms 24 01533 g001
Figure 2. Important plant VLPs with application in biotechnology and plant viral-based nanotechnology. Plant viruses with their corresponding protein databank identification number—CPMV (PDB ID: 1NY7), CCMV (PDB ID: 1CW7), CMV (PDB ID: 1F15), BMV (PDB ID: 1JS9), TBSV (PDB ID: 2TBV), AIMV (PDB ID: 7EPP), PVX (PDB ID: 6R7G), TMV (PDB ID: 7Q22).
Figure 2. Important plant VLPs with application in biotechnology and plant viral-based nanotechnology. Plant viruses with their corresponding protein databank identification number—CPMV (PDB ID: 1NY7), CCMV (PDB ID: 1CW7), CMV (PDB ID: 1F15), BMV (PDB ID: 1JS9), TBSV (PDB ID: 2TBV), AIMV (PDB ID: 7EPP), PVX (PDB ID: 6R7G), TMV (PDB ID: 7Q22).
Ijms 24 01533 g002
Figure 3. Strategies for functionalization of VNPs.
Figure 3. Strategies for functionalization of VNPs.
Ijms 24 01533 g003
Figure 4. Mechanism of reassembly of RCNMV coat protein around metal nanoparticles [163]. (1) metal nanoparticles were linked with synthetic oligonucleotides (DNA-2) mimicking the OAS, (2) RCNMV RNA-1 was used to interact with DNA-2, (3) OAS formation, (4) reassembly of RCNMV coat proteins [163].
Figure 4. Mechanism of reassembly of RCNMV coat protein around metal nanoparticles [163]. (1) metal nanoparticles were linked with synthetic oligonucleotides (DNA-2) mimicking the OAS, (2) RCNMV RNA-1 was used to interact with DNA-2, (3) OAS formation, (4) reassembly of RCNMV coat proteins [163].
Ijms 24 01533 g004
Table 1. List of some viruses used for the design of viral vectors.
Table 1. List of some viruses used for the design of viral vectors.
Virus Classification/Genome OrganizationVirusVectorAchieved High YieldRecombinant
Proteins
Reference
Tobamovirus + ssRNAtobacco mosaic virus (TMV)magnICON5 mg/g FWTGFP[88]
TRBO5.5 mg/g FWTGFP[89]
Potexvirus + ssRNAfoxtail mosaic virus (FoMV)FECT/40 vector1.7 mg/g FWTGFP[90]
potato virus X (PVX)pEff1 mg/g FWTGFP[84]
Geminivirusss circular DNA tobacco yellow dwarf mastrevirus (TYDV)INPACT0.1 mg/g FWThuman Vitronectin[91]
bean yellow dwarf
virus (BeYDV)
pBYRp190.5 mg/g FWTmAb 6D8 against Ebola virus[92]
pRIC0.55 mg/g FWTHPV CP L1[93]
Tobravirus + ssRNAtobacco rattle virus (TRV)TRV- based vector0.01 mg/g of fresh weight root tissueGNA lectin protein[94]
Comovirus + ss RNA cowpea mosaic virus (CPMV)delRNA-2-GFP[95]
pEAQ-HT1.5 mg/gGFP[96]
Table 2. List of known to be edible vaccines against animal pathogens.
Table 2. List of known to be edible vaccines against animal pathogens.
Disease/Infectious AgentsAntigenSpeciesYieldImmunogenicityReference
Norwalk virusCapsid protein (NVCP)Tomatoup to 8% of TSPFreeze-dried tomato (40 µg VLPs) induced NV-specific serum IgG and mucosal IgA in ≥80% of mice.[240]
Potato-19/20 human volunteers developed an immune response after oral immunization with VLPs.[241]
Rabies virusG and N proteins fused to AlMV CPTobacco and spinach0.4 ± 0.07 mg/g of fresh leaf tissueImmunized mice were protected against challenge infection. Human volunteers previously non-immunized demonstrated significant antibody responses after fed.[242]
Newcastle Disease Virus (NDV)Hemagglutinin-neuraminidase protein (HN)Tobacco0.069% of TSPSpecific immune response after oral administration of chicken was induced.[243]
Fusion (F)Maize0.9–3% TSP[244]
Fusion (F) and hemagglutinin-neuraminidase (HN) proteinsMaize0.5–0.8% of total seed protein[245]
Canola up to 0.18% and 0.11% TSP in transgenic seeds and leaves[246]
Potato 0.3–0.6 mg/g of total leaf protein[247]
S. aureus and CholeraD2 fibronectin-binding domain with cholera toxin B subunitC. reinhardtiiUp to 0.7% TSPMice fed with whole algae showed mucosal IgA and systemic IgG responses to CTB and D2. A total of 80% survived after lethal challenge.[248]
Bovine rotavirus (BVR)eBRV4 fused to βGUSAlfalfa0.4–0.9 mg (g TPS)−1An effective anti-rotavirus antibody response was induced in mice after oral administration.[249]
CoronavirusS protein (S1)Tomato-Orally immunized mice showed significantly increased levels of SARS-CoV-specific IgA.[250]
Hog pest virus/F. hepaticaGlycoprotein E2/cysteine proteasesLettuce0.16 mg/g
dry mass
Oral immunization of mice induced specific antibodies.[251]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zahmanova, G.; Aljabali, A.A.; Takova, K.; Toneva, V.; Tambuwala, M.M.; Andonov, A.P.; Lukov, G.L.; Minkov, I. The Plant Viruses and Molecular Farming: How Beneficial They Might Be for Human and Animal Health? Int. J. Mol. Sci. 2023, 24, 1533. https://doi.org/10.3390/ijms24021533

AMA Style

Zahmanova G, Aljabali AA, Takova K, Toneva V, Tambuwala MM, Andonov AP, Lukov GL, Minkov I. The Plant Viruses and Molecular Farming: How Beneficial They Might Be for Human and Animal Health? International Journal of Molecular Sciences. 2023; 24(2):1533. https://doi.org/10.3390/ijms24021533

Chicago/Turabian Style

Zahmanova, Gergana, Alaa A. Aljabali, Katerina Takova, Valentina Toneva, Murtaza M. Tambuwala, Anton P. Andonov, Georgi L. Lukov, and Ivan Minkov. 2023. "The Plant Viruses and Molecular Farming: How Beneficial They Might Be for Human and Animal Health?" International Journal of Molecular Sciences 24, no. 2: 1533. https://doi.org/10.3390/ijms24021533

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop