Next Article in Journal
Green Solid-State Synthesis of Antibacterial Binary Organic Material: Crystal Growth, Physicochemical Properties, Thermal Study, Antibacterial Activity, and Hirshfeld Surface Analysis
Previous Article in Journal
Unveiling GABA and Serotonin Interactions During Neurodevelopment to Re-Open Adult Critical Periods for Neuropsychiatric Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Harnessing Transient Expression Systems with Plant Viral Vectors for the Production of Biopharmaceuticals in Nicotiana benthamiana

1
Tobacco Research Institute, Chinese Academy of Agricultural Sciences, Qingdao 266101, China
2
Graduate School of Chinese Academy of Agricultural Sciences, Beijing 100081, China
3
Division of Math and Natural Sciences, University of Pikeville, Pikeville, KY 41051, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2025, 26(12), 5510; https://doi.org/10.3390/ijms26125510
Submission received: 19 May 2025 / Revised: 5 June 2025 / Accepted: 6 June 2025 / Published: 9 June 2025
(This article belongs to the Section Molecular Pharmacology)

Abstract

Plant Molecular Farming (PMF) capitalizes on the unique properties of plants as bioreactors to efficiently produce valuable proteins, pharmaceuticals, and enzymes. This review emphasizes the critical role of transient expression systems, particularly in Nicotiana benthamiana, due to its susceptibility to various pathogens. Viral vector-based transient expression has proven essential during health emergencies like COVID-19, enabling rapid recombinant protein production. The review also evaluates different transient expression platforms and highlights their applications in biopharmaceutical production, education, synthetic biology, and gene editing. Advances in viral vector modification, hydroponics, and Controlled Environment Agriculture (CEA) are presented as transformative innovations enhancing scalability and regulatory compliance. Furthermore, glycoengineering advancements broaden the range of producible biopharmaceuticals, improving global medication access. By exploring these advancements, this review underscores the vast potential of transient expression systems to meet dynamic scientific and market demands, positioning PMF as a vital component in modern biotechnology.

1. Introduction

Plant Molecular Farming (PMF) harnesses the power of plants as bioreactors, presenting a promising avenue for the efficient production of valuable proteins, pharmaceuticals, and industrial enzymes [1,2,3,4]. Over 170 recombinant protein drugs have reached the market, targeting conditions like arthritis and cancer, with many more in development [5,6,7,8,9]. This innovative approach enables the generation of recombinant proteins through transient expression systems, plant cell-suspension culture, and stable expression. Compared to stable transformation, transient protein expression in plant cells offers a quicker and more resource-efficient alternative for developing fully transgenic plants. Utilizing various techniques such as viral vectors, agroinfiltration, hydroponic systems, and downstream processing, this method streamlines the process of gene expression in plants [3,10,11,12].
N. benthamiana, a plant species indigenous to the arid regions of Australia, has emerged as a central model organism in plant biotechnology and molecular farming [11,13]. Its prominence in research and production settings is largely attributed to its exceptional susceptibility to a wide array of plant pathogens, including viruses, bacteria, and fungi. This heightened vulnerability is due in part to a natural mutation in its RNA-dependent RNA polymerase gene, which impairs its antiviral defense mechanisms [14]. As a result, N. benthamiana serves as an ideal host for transient gene expression, a technique that allows for the rapid, temporary introduction and expression of foreign genes [15]. This makes it particularly useful for applications such as vaccine development, therapeutic protein production, and functional genomics studies, where speed and flexibility are crucial. The approval of the N. benthamiana plant-based COVID-19 vaccine, Covifenz, which meets Canadian safety, efficacy, and quality standards, exemplifies the regulatory achievements and underscores the global potential of transient expression systems [16].
Transient expression systems have evolved through extensive back and forth between various platforms, each offering distinct advantages and limitations. Early systems, such as the infiltration of Nicotiana benthamiana using Rhizobium radiobacter (formerly known as Agrobacterium tumefaciens), quickly gained popularity due to their high protein yield and scalability [17,18]. However, challenges like gene silencing and inconsistent expression led to the integration of viral vectors, such as TMV- and PVX-based systems, which improved expression levels but raised biosafety and regulatory concerns [10,19,20,21,22]. This spurred interest in deconstructed viral vectors, like the MagnICON, pEAQ, and pHREAC systems, which balanced high expression with modular design and safety [11,23,24,25,26,27,28]. More recent efforts have focused on optimizing expression timing, subcellular targeting, and co-expression of silencing suppressors to further enhance yields and functional protein recovery [25,29,30]. These iterative refinements underscore the dynamic nature of transient expression technologies as researchers continuously adapt systems to meet growing bioproduction demands.
PMF technology has improved significantly over the past decade, with yields reaching five grams per kilogram of plant biomass [31]. However, there is still a need to improve yield and downstream processing and purification, which could constitute up to 80% of total production costs. Technical and economic issues such as lignin, fibers, swelling substances, waxes, phenolic compounds, pigments, and endogenous proteases pose challenges [32]. The sheer amount of biomass poses technical and economic challenges, such as clogging chromatography columns and difficulty in removing plant pigments and phenolics from monoclonal antibody pools [33]. Other issues include scaling up filtration processes, proteolytic destruction of target proteins during downstream processing, creating an effective viral clearance method, and dealing with changing expression levels. Proper glycosylation, or “humanized” glycans, is crucial for safety and efficacy. Challenges of protein farming include public acceptance and competition with established biomanufacturing systems.
This review evaluates the strengths and limitations of various transient expression platforms in PMF and explores their considerable potential for expansion in fields such as education, synthetic biology, functional genomics, and personalized medicine.

2. Advancements and Applications of Plant Transient Expression Systems: A Historical Overview

The historical progression of plant transient expression technology has been defined by pivotal developments (Figure 1). The intersection of plant biotechnology and viral vectors has notably propelled advancements in this field. A significant milestone was the cloning and characterization of Cauliflower Mosaic Virus (CaMV) DNA in Escherichia coli, which revealed that the unencapsulated viral DNA was non-infectious in plants [34]. This finding, alongside the limitations of existing methods, helped drive the development of key DNA delivery techniques in the 1980s, such as electroporation and particle bombardment, paving the way for novel approaches to transient gene expression in plants. In a significant 1987 study, Klein et al. [35] used microprojectiles to introduce Tobacco Mosaic Virus (TMV) RNA into plant cells, successfully achieving transient expression in onion epidermal tissue. In the same decade, Marcotte et al. [36] demonstrated for the first time that a plasmid DNA containing a chimeric β-glucuronidase enzyme (GUS) reporter gene could be regulated within 60 min in rice protoplasts transformed via polyethylene glycol (PEG), enhancing transient expression technology for functional gene analysis. Concurrently, plant viruses were recognized as vectors for genetic modification, as demonstrated by the infectivity of Brome mosaic virus (BMV) cDNA in barley through in vitro assembly [37]. Additionally, Takamatsu et al. [38] documented that a TMV-based vector could produce biologically active bacterial chloramphenicol acetyltransferase in treated plant leaves ten days after inoculation.
The 1990s marked a significant era in the evolution of plant viral vectors, tailored for diverse protein productions in plant biotechnology. This decade saw the refinement of viral vector systems for specialized applications in plant science. The team led by Ow investigated the use of the Barley stripe mosaic virus (BSMV) genome for expressing foreign genes in both dicot and monocot cells, enhancing the scope of viral vectors in plant genetic engineering [39]. In a significant study by Donson et al. [40], a TMV-based vector was shown to systemically express the bacterial protein neomycin phosphotransferase across various plant systems. In 1996, Tang et al. [41] leveraged the agroinfiltration technique to transiently express the avrPto transgene in Nicotiana sylvestris cells harboring the Pto gene, inducing a defense response. This period also witnessed the application of plant virus vectors for producing various proteins, exemplified by the creation of vaccines using a Cowpea Mosaic Virus (CPMV) vector derived from Vigna unguiculata, and the expression of a monoclonal antibody (mAb) CO17-1A targeting a colon cancer antigen in N. benthamiana. Furthermore, Schob et al. [42] demonstrated the targeted delivery and expression of the P35S-CHN48 gene construct via R. radiobacter in N. sylvestris, using syringe infiltration. Notably, the CHN48 transgene was expressed exclusively in leaves transformed with the vector alone, not in those previously transformed with CHN48, which remained silent.
The 2000s marked a significant advancement in PMF with the development of various virus-based deconstructed vectors for transient expression. This innovation facilitated large-scale production of biopharmaceuticals, vaccines, and other valuable proteins, serving as an efficient alternative to traditional expression systems. It has since been widely adopted in both research and industrial applications. Key viral backbones include RNA viruses like TMV [43,44,45], Potato virus X (PVX) [46,47], Tobacco etch virus (TEV) [47], and CPMV [48], along with the DNA geminivirus Bean yellow dwarf virus (BeYDV-based vectors) [49,50,51]. Among these, the tobamovirus-based system, “magnICON®”, introduced by Dr. Yuri Gleba [23] and his team at Icon Genetics, achieved notable success. In 2005, Magnifection, a vacuum agroinfiltration technique, led to yields of up to 4 g of recombinant protein per kilogram of fresh leaf biomass in N. benthamiana and 2.5 g per kilogram in N. tabacum within just four days post-infiltration [52].
The FDA’s approval in May 2012 of the first plant-derived enzyme for clinical use, ELELYSO™ (taliglucerase alfa) by Protalix BioTherapeutics, underscored the potential of PMF. Utilizing carrot cells, ELELYSO™ treats adult patients with Gaucher disease, representing a pioneering achievement in plant-based pharmaceuticals [53]. Further advancements included the use of a transgenic N. benthamiana line lacking specific N-glycan residues to produce monoclonal antibodies (mAbs) with human-like glycoforms in 2011, enhancing the efficacy of an Ebola virus immunoprotective agent [54]. Additionally, a BeYDV-based vector produced an effective non-replicating subunit vaccine, Ebola immune complexes (EIC), which demonstrated protective efficacy in mice [55]. These efforts culminated in the protection of nonhuman primates using a cocktail of three tobacco-derived anti-Ebola mAbs, leading to the development of ZMapp for combating the Ebola outbreak in Africa [56].
During the 2020s, the plant transient expression platform demonstrated its potential in rapid vaccine development. Companies such as Medicago Inc. in Québec, Canada; iBio in Bryan, TX, USA; and Kentucky BioProcessing Inc. in Owensboro, KY, USA, rapidly developed VLP-or subunit-based COVID-19 vaccines within just a month [57]. Medicago’s plant-derived vaccine, CoVLP + AS03, underwent a comprehensive phase 3 trial across 85 centers worldwide, demonstrating its efficacy against various COVID-19 strains. The vaccine significantly reduced symptomatic cases by 69.5% and moderate-to-severe cases by 78.8% [58]. These results led to the approval of the vaccine by Health Canada on 24 February 2022 under the brand name “Covifenz”. Although Covifenz was not ultimately deployed in public vaccination campaigns, its regulatory approval marked a major milestone in plant-based pharmaceutical technology and significantly heightened both public and industry awareness of the potential of plants in modern vaccine development [59,60].

3. Plant Virus-Based Vectors in Transient Protein Expression

Transient protein expression in PMF provides a rapid and efficient alternative to stable transformation, significantly shortening production timelines—from several years to just a few weeks. It also avoids complications associated with random transgene integration and the lengthy screening processes required for transgenic lines [61]. While widely used in pharmaceutical production, its utility extends into educational environments, functional genomics, and diverse biological research, enabling accelerated experimentation and data generation across these fields. In educational settings, transient expression systems offer a hands-on, accessible platform for teaching molecular biology and biotechnology, allowing students to observe gene expression, test genetic constructs, and engage with synthetic biology in real time. In parallel, their use in functional genomics supports fast and flexible analysis of gene function, protein–protein interactions, subcellular localization, and regulatory networks—all without the need for stable integration. This versatility across both learning and research contexts not only enhances education but also drives scientific discovery, positioning transient expression platforms as essential tools for both developing scientific talent and advancing genomic understanding (Figure 2).
Advanced technologies such as deconstructed viral vectors, including the pEAQ vectors from Lomonossoff’s team [62], Lindbo’s TRBO [63], the geminiviral (BeYDV) vectors developed by Mason’s group [29,64], and the magnICON® systems devised by Gleba and colleagues at Icon Genetics, have been integrated with humanized glycosylation techniques in N. benthamiana [65,66]. These integrations facilitate rapid protein accumulation and sophisticated glycoengineering, crucial for producing high-quality therapeutics.
Various methods of transient gene expression are employed in PMF, using full viruses, separated viral genomes, or deconstructed viral genomes to introduce viral vector gene cassettes into plants (Figure 3). Achieving high yields is vital for the scalability and economic feasibility of PMF. Continual efforts aim to enhance production within transient expression systems. For instance, Lomonossoff’s team developed the pHREAC vector, which combines a synthetic 5′ UTR (5S0) with the 3′ UTR of CPMV, achieving a maximum recombinant protein yield of approximately 3 g/kg of GFP in fresh weight (FW) tissue, significantly higher than the 1.5 g/kg FW of GFP yields achieved with the pEAQ vector, also invented by his team [26].
Additionally, the TRBO vector, an efficient system for producing recombinant proteins, modifies the TMV genome to enable bacterial expansion. By replacing non-essential TMV genes with the target gene while preserving essential viral replication elements allows for viral replication within plant cells without the TMV coat protein gene. This adjustment boosts agroinfection efficiency and protein expression rates, with yields reaching 3 to 5 g/kg of FW, substantially increasing protein yield [63]. Essential DNA elements such as a promoter, two terminators, 5′ and 3′ untranslated regions (UTRs), and a matrix attachment region play a key role in transient gene expression. In a recent study by Coates et al. (2024) [67], the addition of extra DNA elements, specifically a 5′ UTR, to the pJL-TRBO vector enhanced GFP gene expression both in planta and at the protein level, yielding a 14-fold increase compared to the pJL-TRBO-eGFP vector [63], which did not include the UTR.
The magnICON® system is one of the first transient expression platforms to successfully enter the commercial market, representing a significant advancement in PMF. This system employs Rhizobium to deliver viral replicons, serving as an infectious agent to facilitate the transient amplification of deconstructed viral vectors across the plant’s cellular structure [23,68,69]. The process begins when Rhizobium carrying T-DNA that encodes RNA replicons infiltrates the plant, setting off a systemic infection. Viral vectors then enhance cell-to-cell transmission and amplify the gene expression, achieving high-level production of recombinant proteins usually within 4 to 7 days, yielding up to 3–5 g/kg of FW in N. benthamiana [52,70].
The efficacy of the magnICON® system in producing a range of proteins has been well documented, notably its role in synthesizing a prominent Ebola virus antibody [54,71]. Recent studies have further demonstrated its capability to produce high yields of functional foreign proteins. For example, the transient expression of recombinant human interferon α1b, α2b, and Gamma demonstrates significant biological activity against pathogens and cancer cells [72]. Variants of aflibercept equipped with specifically designed carbohydrates N-glycans with terminal GlcNAc (AFLIGnGn) and sialic acid residues (AFLISia) have yielded up to 2 g/kg of FW. These variants are properly assembled into dimers in glycoengineered N. benthamiana plants and exhibit binding potencies comparable to VEGF165 [73]. Modarresi et al. [74] successfully expressed a recombinant anti-VEGFR2 nanobody in N. benthamiana and N. tabacum cv. Xanthi, with the nanobody accumulating to 0.45% of total soluble protein, representing the first successful expression of a camelid anti-VEGFR2 nanobody in tobacco plants. Additionally, the system has been effective in producing immunoglobulin M (IgM), the largest antibody isotype known for its extensive glycosylation and oligomerization. Specific variants, H4 and P5C3 IgMs, of SARS-CoV-2 IgMs were generated in N. benthamiana (ΔXTFT line), yielding purified IgM at approximately 10 mg/kg and 15 mg/kg of FW for H4- and P5C3-IgM, respectively, with high glycan homogeneity. Comparative analyses revealed that, when assessing neutralizing activities, P5C3-IgM-P exhibited a roughly 12-fold increase (IC50 3 pM), and H4-IgM-P showed about a 390-fold increase (IC50 13 pM) compared to their corresponding IgG1s [75]. MagnICON®-based therapies have emerged as a promising platform in the realm of PMF, demonstrating both safety and efficacy in clinical and preclinical studies [76,77]. For instance, a bivalent norovirus-like particle vaccine produced in N. benthamiana exhibited excellent safety and robust immunogenic responses in animal models, paving the way for human trials [77].
Following the magnICON® system, other viral expression vectors like the BeYDV-based vectors have further advanced the field. The BeYDV system uses a Rhizobium-mediated approach similar to magnICON® but incorporates different viral elements (Table 1). In 2011, the Mason group engineered a high-level expression system that leverages elements from the replication machinery of a single-stranded DNA virus. This system involves the replication initiator protein (Rep), which facilitates the release and replication of a replicon from the “LSL vector”. This vector includes an expression cassette for a gene of interest, which is flanked by the virus’s cis-acting elements and was tested in tobacco NT1 cells in which the GUS expression yield that co-expressed Rep/RepA was enhanced by up to 40-fold [50,78]. A BeYDV-based single DNA replicon vector containing a built-in Rep/RepA cassette without p19 was further developed for recombinant protein expression at 0.8 g/kg FW with transient expression in N. benthamiana [49], demonstrating its potential for efficient and scalable protein production in plant-based systems.
The magnICON® system stands as a prominent example of efficient transient expression, consistently yielding 3–5 mg of foreign proteins per gram of FW within just a week [52,70]. On the other hand, vectors based on the BeYDV have traditionally produced lower levels of recombinant proteins. However, innovative modifications to enhance these yields have shown promising results. For instance, Yamamoto et al. [79] improved the BeYDV replication mechanism by implementing a double terminator system, which integrates both a heat shock protein terminator and an extensin terminator. This modification notably increased the GFP yield to approximately 3.7 mg/g of FW within three days. Further refining BeYDV’s efficiency, Diamos and Mason [29] explored genetic modifications at the molecular level. They made a single nucleotide change in the 5′ untranslated region (UTR) of BeYDV, which successfully decreased the expression of the replication initiator proteins Rep/RepA. This alteration not only reduced cell death but also enhanced the yield of the Norwalk virus capsid protein (NVCP) to 2 mg/g of FW. These advancements indicate a significant potential for improving the productivity and application scope of BeYDV-based vectors in PMF.
The BeYDV system has undergone sophisticated optimizations to enhance its ability to efficiently co-express multiple proteins, thereby significantly improving its flexibility and scalability. These strategic advancements in the vector’s design have facilitated substantial increases in expression levels. BeYDV vectors can accommodate large genetic inserts, making them highly effective for producing high yields of monoclonal antibodies and other therapeutic proteins, with productivity increases of 4–5 times the initial yields. This system has successfully co-expressed two to four fluorescent proteins within plants, achieving notable yields of 3–5 g/kg of FW, which constitutes about 50% of the total soluble protein (TSP) [64].
Moreover, the antibodies produced using the BeYDV system, yielding 1.2–1.4 g/kg of FW, have shown strong neutralizing effects against Zika virus and other flaviviruses [64]. This demonstrates the utility of the BeYDV system in rapidly and cost-effectively producing a diverse array of pharmaceutical multi-protein complexes, highlighting its potential in advancing therapeutic solutions.
Like the magnICON® system, the BeYDV system is widely used in PMF for the production of vaccines, antibodies, and various proteins in both academic and industrial settings. Rattanapisit et al. [30] demonstrated the potential of BeYDV-based vectors in N. benthamiana for producing the SARS-CoV-2 receptor binding domain (RBD) [80] and monoclonal antibody CR3022 using a transient expression system, with peak yields of 8 μg/g and 130 μg/g leaf fresh weight, respectively, at 3 days post-infiltration. The plant-produced RBD effectively bound to ACE2, while CR3022, though binding to SARS-CoV-2, lacked in vitro neutralization. This is the first demonstration of the N. benthamiana-based production of these SARS-CoV-2 proteins. In 2023, the Phoolcharoen group exhibited the efficacy of BeYDV-based vectors in N. benthamiana for the production of the anti-CTLA-4 monoclonal antibody 2C8. This antibody effectively binds both human and murine CTLA-4 and has been shown to inhibit tumor growth in vivo as effectively as Yervoy® [81]. In the same year, their work with a purified anti-PD-L1 antibody yielded 1.8 mg/g of FW within five days post-infiltration, further demonstrating the capability to inhibit tumor growth in mice [82]. Additionally, this group reported that the BeYDV-based system in N. benthamiana produced a plant-purified anti-dengue virus D54 neutralizing therapeutic antibody, achieving a yield of 0.34 mg/g of FW [83]. A comparative overview of the magnICON® and BeYDV systems, within the context of PMF, is detailed in Table 1, illustrating their pivotal roles in advancing biopharmaceutical manufacturing and research.
Table 1. A comparative overview of the magnICON® and BeYDV systems in PMF.
Table 1. A comparative overview of the magnICON® and BeYDV systems in PMF.
No.MagnICON® SystemBeYDV System
1.The magnICON® agnICON system relies on “deconstructed” viral vectors, employing Rhizobium-mediated systemic delivery for recombinant protein production [84].BeYDV, a geminivirus, employs Rhizobium-mediated delivery to plant cells utilizing expression vectors facilitated by the viral replication initiator protein (Rep) to generate high levels of recombinant protein [78].
2.Primarily functioning in tobacco, the magnICON® system faces limitations in lettuce and tomatoes due to its reliance on a TMV-based viral vector [85,86].BeYDV’s extensive host compatibility enables the efficient production of proteins at elevated levels in numerous dicotyledonous plants [87].
3.Transient expression of model proteins like GFP in the magnICON® system yielded 3–5 mg/g FM within one week in N. benthamiana [52].Achieved exceptional yields of 3–5 mg/g leaf FW in just 4–5 days, equivalent to roughly 50% of TSP [64,87].
4.The transient nature of the magnICON® system, free from stable genetic plant modifications, enables faster and more adaptable production [79].The BeYDV system offers flexibility in expressing large gene fragments and has the potential for high-volume production of recombinant proteins [88].
5.Supports systemic movement and requires low Rhizobium density, enabling cost-effective agroinfiltration and simpler downstream processing [17].Lacks systemic movement, required high Rhizobium density (OD600 = 0.2), leads to higher agroinfiltration costs and more complex purification due to endotoxins [29].
In plant biotechnology, choosing the right viral vector for transient expression is essential and depends on the host plant and the targeted protein. The TMV-based and BeYDV systems are known for their rapid production of high yields of recombinant proteins, typically within a week. TMV-based systems have exceptional efficacy in N. benthamiana, but their efficiency diminishes in other plants like lettuce and tomatoes due to species-specific interactions with TMV elements [85,86]. This limitation, which allows for the expression of only one or two genes simultaneously, restricts their application in synthetic biology, where the concurrent expression of multiple genes is often required for the synthesis of complex products.
Conversely, the BeYDV system is capable of expressing multiple genes simultaneously and managing large gene fragments across a variety of dicot plants [29,86,87]. This feature makes BeYDV highly valuable for research, gene editing, and synthetic biology. Unlike the magnICON® or TRBO systems, BeYDV does not facilitate cell-to-cell movement, necessitating higher concentrations of Rhizobium for effective delivery (OD600 = 0.2) as noted by Diamos and Mason [29], compared to OD600 = 0.015 for TRBO as per Lindbo [63]. This leads to increased costs for agroinfiltration and subsequent endotoxin removal and purification processes, challenges that are less significant in industrial-scale settings with the TRBO system.

4. Comparison of Plant-Based Transient Expression Systems with Other Expression Systems

Compared to other expression platforms, plant-based transient expression systems offer a unique balance of scalability, cost-effectiveness, and the ability to produce complex eukaryotic proteins with proper folding. While they require labor-intensive infiltration processes and controlled growth conditions, they do not need sterile environments or expensive infrastructure. Plant systems typically produce plant-specific glycosylation patterns, which differ from mammalian glycosylation but are sufficient for many applications. They are particularly well-suited for medium- to large-scale production with relatively low upstream costs [61,89,90] (Table 2).
In contrast, yeast-based expression systems (e.g., Pichia pastoris) are easier to culture and offer faster protein production. They can perform some eukaryotic post-translational modifications (PTMs) and can be engineered to mimic human-like glycosylation, although natural patterns differ from mammalian systems. Yeast systems are robust, scalable for industrial use, and less labor-intensive but may not be ideal for proteins requiring highly specific PTMs or precise folding, where plant systems may be more favorable [91,92,93,94].
Bacterial systems, especially E. coli, are fast, inexpensive, and highly scalable, making them ideal for simple proteins and research reagents. However, they lack the machinery for complex PTMs and often yield misfolded proteins or inclusion bodies, limiting their use in therapeutic applications where correct folding and modifications are critical [95].
Mammalian expression systems provide the most accurate human-like PTMs and are preferred for high-end therapeutic protein production. However, they come with high costs and require sterile conditions, specialized media, and CO2 incubators, making them the most resource-intensive option among all systems discussed [96,97,98,99].
Insect cell systems, such as those utilizing Sf9 or Sf21 with baculovirus vectors, strike a balance between mammalian and plant systems. They support proper protein folding and more mammalian-like PTMs than plants but still require sterile conditions, bioreactors, and expensive media. While they offer higher and more consistent yields, plant-based systems remain more economical and operationally simpler for large-scale applications when exact human PTMs are not essential [100,101].
Algal systems, like Chlamydomonas reinhardtii, are emerging platforms known for rapid growth, low-cost cultivation, and sustainability due to photosynthetic production. They can perform some eukaryotic PTMs, but their glycosylation pathways are less defined and often incompatible with therapeutic use. Additionally, algal expression tends to produce lower and more variable yields than plant systems, which are currently more mature and reliable for producing complex proteins at scale [102].
Table 2. Quantitative comparison of different expression systems with key insights.
Table 2. Quantitative comparison of different expression systems with key insights.
MetricPlant-BasedMammalian CellsBacterialYeast/FungiInsect CellsAlgal SystemsReferences
Expression Yield (mg/L)1.6 g/Kg FW3.2 g/L1.2 g/L1.9 g/L0.7 g/L1.14 μg/g[21,103]
Yield NotesHigh yields via agroinfiltrationHEK293/CHO provide human-like PTMs but lower yieldsHigh yield but may lack proper PTMsGood yields with some glycosylationBaculovirus system allows for complex foldingGeminiviral vector allowed for the expression of recombinant proteins in Chlorella vulgaris[89,103,104,105,106,107,108]
Time to Expression (Days)3–72–101–32–53–42[61,79,103,109,110,111,112]
Time NotesFast with agroinfiltration/viral vectorsQuick with lipid transfection, viral vectors take longerVery fast due to simple machineryModerate speed for plasmid-based systemsRequires baculovirus amplificationTime varies by strain and method[61,79,103,109,110,111,112]
Operating costLowVery highLowMediumHighLow[113]
Cost NotesCost-effective, good for scalingExpensive media and slow growthVery cheap but lacks PTMsModerate cost with better PTMs than bacteriaCostly but allows for complex proteinsSustainable but optimization needed[113]
ScalabilityHighMedium to HighVery HighHighMediumHigh[61,105,114,115,116]
Scalability NotesEasily scalable, needs greenhouse/fieldsBioreactor-based, limits scaleExtremely scalable, used industriallyOptimized bioreactors allow for large-scale useLimited by baculovirus productionLarge-scale photobioreactors possible[61,105,114,115,116]
PTM CapabilityModerateHighNoneIncorrectHighModerate[113,117,118]
PTM NotesGlycoproteins, glycosylation differs from humansComplex, human-like PTMsDoes not support most eukarotic PTMsPerforms glycosylation but differs from humansProvides PTMs similar to mammalian cellsMinimal PTMs, unsuitable for complex proteins[105,119,120,121,122,123]
Workflow ComplexityModerateHighLowModerateHighModerate[124,125,126,127,128,129]
Complexity NotesRequires cultivation and agroinfiltration expertiseNeeds sterile conditions and expensive mediaSimple, well-established protocolsRequires bioreactor optimization for high yieldBaculovirus production adds stepsCultivation methods need optimization[124,125,126,127,128,129]

5. The Role of N. benthamiana and Glycoengineering for Biopharmaceutical Production

N. benthamiana is widely recognized as an ideal host in PMF due to its rapid growth, significant biomass yield, and high susceptibility to a variety of viral vectors. Unlike N. tabacum, N. benthamiana exhibits a complex genome with extensive chromosomal rearrangements, making it uniquely receptive to genetic manipulation [130]. Additionally, N. benthamiana lacks several key antiviral silencing genes, such as RDR1, RDR6, DCL2, DCL3, and AGO2, which are present in N. tabacum. This genetic distinction not only enhances its vulnerability to viral infections but also amplifies its efficiency as a host for viral-based expression systems [131]. Utilizing the MagnICON system, the transient expression of ER-targeted human glycoproteins like interleukin-6 (IL6) in N. benthamiana leaves has achieved yields as high as 7% of TSP, significantly higher than the 1% typically observed in N. tabacum [132]. Similarly, this system has facilitated a higher protein accumulation for potential therapeutics such as the colorectal cancer vaccine GA733-2 and the fusion protein rGA733-Fc in N. benthamiana compared to N. tabacum [133].
N. benthamiana’s rapid lifecycle, transitioning from seed to maturity in just 6–8 weeks, makes it a valuable resource for fast-track projects requiring quick production of recombinant proteins. Its ability to generate abundant leaf material enables extensive protein extraction from minimal cultivation areas, boosting its utility for both research and commercial purposes [134]. The plant’s compatibility with various viral vectors, such as TMV, PVX, and BeYDV, is largely attributed due to its compromised innate immune response. This trait facilitates efficient viral replication and gene expression, crucial for the effective use of viral vectors in transient expression systems. In addition, N. benthamiana’s amenability to cutting-edge genetic engineering methods, including CRISPR/Cas9 for targeted genome editing, facilitates precise genetic alterations using recombinant negative-stranded RNA rhabdovirus vectors, resulting in plants devoid of foreign DNA [135]. These attributes have significantly contributed to the deployment of various transient expression products, such as vaccines and therapeutic proteins, in industrial settings, enhancing N. benthamiana’s role in addressing urgent global health needs (Table 3).

5.1. Enhancing the Biocompatibility of Plant-Derived Pharmaceuticals Through Glycosylation Pathway Modifications in N. benthamiana

Despite extensive clinical trials showing no adverse effects from plant-specific glycosylation [136], concerns persist in the scientific community regarding the differences between plant and mammalian glycosylation pathways. To address these concerns, researchers have developed humanized glycosylated strains of N. benthamiana to better align with regulatory standards and enhance the biocompatibility of plant-derived therapeutics [65,66]. Advanced molecular and genome-editing techniques have enabled the precise removal of specific glycosylation markers, such as β-1,2-xylose and core α-1,3-fucose, from plant-expressed glycoproteins by targeting the respective glycosyltransferase genes for knockout in N. benthamiana. A significant advancement involves using a multiplex CRISPR/Cas9 strategy to create N. benthamiana lines devoid of both β-1,2-xylosyltransferase and α-1,3-fucosyltransferase activities (FX-KO), highlighting a move towards fully human-compatible glycosylation profiles [65]. Similarly, Jansing et al. [137] effectively used CRISPR/Cas9 to eliminate six glycosyltransferase genes, successfully producing recombinant proteins devoid of these plant-specific glycosylation elements.
Research indicates that therapeutic proteins manufactured in these genetically edited lines maintain biological activities comparable to those produced in mammalian systems, which is crucial for ensuring their therapeutic efficacy and safety [138]. Recently, the Steinkellner group engineered a marker-free genome-edited N. benthamiana line (NbXF-KO) that lacks seven genes associated with β1,2-xylosyltransferase and α1,3-fucosyltransferase, producing monoclonal antibodies that exhibit human-like β1,4-galactosylated and α2,6-sialylated glycosylation patterns with enhanced biological functions [139]. Furthermore, in their study, the Steinkellner team explored the glycosylation efficiency of three popular transient expression vectors—pEAQ, magnICON®, and pTra—within a glycoengineered plant background to express the therapeutic monoclonal antibody Cetuximab. All the vectors achieved similar, human-compatible glycosylation profiles, although slight differences in product quality were observed. Compared to the other two, magnICON®-based expression yielded the highest protein levels, but the antibody showed slightly increased mannosidic and incompletely processed galactosylated/sialylated structures, likely due to stress on the secretory pathway from high expression levels, highlighting both the potential and current limitations of plant-based glycoengineering to meet stringent industrial and regulatory requirements [140]. These innovations are pivotal in refining the production processes for high-quality, safe, and effective plant-derived therapeutic proteins.

5.2. Overcoming Downstream Processing Challenges with N. benthamiana

Downstream processing in PMF using N. benthamiana has traditionally been a complex and labor-intensive task, particularly in academic settings, where the extraction and purification of proteins from intricate plant matrices present numerous obstacles [141]. Various plant-derived substances such as lignin, fibers, swelling agents (like pectins), waxes, phenolic compounds, pigments, and endogenous proteases pose significant technical and economic challenges during downstream processing [142]. These components can hinder protein extraction, increase viscosity, interfere with purification, and degrade target proteins. For example, lignin and fibers make cell disruption more difficult, while phenolics and proteases can damage or inactivate the product [142,143]. Pigments and waxes complicate purification, often requiring additional processing steps [144]. Collectively, these factors increase costs, reduce yields, and necessitate specialized equipment or additives, making downstream processing a major bottleneck in the economic viability of plant-based bioproduction. Recent advancements have significantly improved both yield and purity while addressing the reduction of undesirable alkaloid content. For instance, blanching combined with chromatography simplifies the purification of the thermostable malaria transmission-blocking vaccine candidate FQS from N. benthamiana, achieving ~72% purity and 60% recovery while mitigating protease degradation [145]. A two-step tandem affinity purification strategy employs distinct affinity tags for each component of a protein complex, enabling the recovery of highly purified complexes suitable for structural and biochemical analyses [146]. A combined pH and temperature treatment removes approximately 70% of host cell proteins from clarified tobacco extracts and intact leaves, improving the purification of biopharmaceutical proteins sensitive to extreme conditions while broadening the applicability of precipitation-based techniques by minimizing product denaturation and loss [147]. Stephan et al. introduced a cost-effective approach involving acidic extraction and a single chromatography step, achieving a high recovery yield of purified colicins while reducing nicotine content to levels comparable to those found in common nicotine-containing foods such as tomatoes or potatoes [148]. Using ultrafiltration/diafiltration (UF/DF) with a 100 kDa membrane, Opdensteinen et al. simplified the purification of the ~11 kDa HIV-neutralizing lectin cyanovirin-N from tobacco extracts, achieving ~70% recovery, a threefold purity increase, and process volume reduction before chromatography [149]. Additionally, Faye et al. described a rapid and efficient method for purifying N. benthamiana-produced antibodies using protein A magnetic beads. This technique reduces resource requirements, operational time, and equipment needs, while delivering high-purity antibodies in a single step [150].
In industrial applications, many of these challenges have been successfully addressed, as evidenced by the commercialization of products like ZMapp™, an antibody cocktail for Ebola, and plant-based COVID-19 vaccines utilizing virus-like particles (VLPs). These achievements highlight N. benthamiana’s potential for the rapid, scalable, and efficient production of complex biopharmaceuticals [151,152]. Although details of industrial advancements are often proprietary, the outcomes demonstrate substantial technological progress and process optimization, ensuring consistency, efficacy, and compliance with regulatory standards.
As regulatory approvals for plant-derived pharmaceuticals grow, the role of N. benthamiana is set to expand further, cementing its value in swiftly and effectively addressing global health challenges. This ongoing progress in PMF technology promises more innovations and broader access to crucial biopharmaceuticals, illustrating the plant’s essential role in modern medicine and biotechnology.

6. Optimizing Plant Molecular Farming Through Hydroponics and Controlled Environment Agriculture (CEA)

Hydroponics and Controlled Environment Agriculture (CEA) represent significant advancements in PMF (Figure 4), providing precise control over environmental factors that critically influence plant growth and metabolic activities. Hydroponics, the cultivation of plants in a soilless medium using nutrient-rich solutions, optimizes nutrient management and water use. This method eliminates the variability associated with soil conditions and reduces the risk of soil-borne diseases, significantly enhancing the health and productivity of plants like N. benthamiana [1,93,152].
CEA complements hydroponics by providing meticulous control over environmental conditions such as temperature, light, humidity, and CO2 levels. This controlled setting allows for the fine-tuning of conditions that stimulate specific genetic pathways, enhancing the production of targeted proteins. For instance, manipulating light spectra can trigger the expression of genes essential for synthesizing specific therapeutic proteins, optimizing protein yield and quality [93,152,153,154].
These technologies not only boost plant growth and protein yield but also meet critical regulatory compliance and safety standards necessary for pharmaceutical production. The precise conditions in hydroponics and CEA ensure adherence to Good Manufacturing Practices (GMPs), pivotal for gaining regulatory approval for plant-derived pharmaceuticals. They enhance the consistency and traceability of the production process, facilitate contamination control by reducing pesticide use, and support detailed documentation of all cultivation parameters, simplifying regulatory audits.
Furthermore, these methods improve the quality control of the final biopharmaceutical product, ensuring that the proteins produced possess the necessary post-translational modifications, such as proper glycosylation patterns, which are crucial for their efficacy and safety. By streamlining these elements, hydroponics and CEA not only enhance the scalability of PMF but also reinforce its role in producing dependable and safe biopharmaceuticals, thereby supporting the expansion of plant-based drug development.

7. New Developments in Transient Expression Systems

Transient expression systems have undergone significant advancements in recent years, driven by the need for faster, scalable, and more cost-effective production of recombinant proteins, vaccines, and therapeutic agents. These developments encompass novel methodologies, technological tools, and enhanced host systems to improve yields, efficiency, and product quality.

7.1. Plant Cell Pack (PCP) System for Transient Expression: A Promising Innovation in Plant Biotechnology

Transient expression systems using R. radiobacter infiltration of whole N. benthamiana plants have been extensively explored for academic and commercial applications. However, the reliance on whole plants presents challenges, as screening multiple constructs or production conditions becomes both costly and labor-intensive. To address these limitations, the Buyel group introduced the Plant Cell Pack (PCP) system, a novel approach involving the infusion of R. radiobacter into three-dimensional, porous plant cell aggregates [155].
This innovative system is compatible with plant species such as N. tabacum BY-2, N. benthamiana, and Daucus carota, achieving up to a 10-times-higher efficiency than liquid culture-based transient expression methods. It supports high-throughput screening, metabolic engineering, and recombinant protein production, making it a valuable tool for process optimization. By integrating PCP preparation with automated liquid-handling systems, this method significantly reduces costs, minimizes errors, and increases throughput to over 2500 samples per day while maintaining consistent protein analysis results [156]. Notably, plant virus-derived replicating vectors have been employed in tobacco BY-2 plant cell packs, enabling scalable and efficient transient protein expression [157]. With the use of single- and multi-cassette viral vectors optimized for enhanced replication and hypertranslation, protein yields of up to ~700 ng/g fresh mass have been achieved, underscoring the potential of this system in plant biotechnology.
Despite its promise, the PCP system remains a relatively new technology. Current protein yields are still modest, and further strategies are required to improve its efficiency, yield, and scalability. Continued research and optimization efforts will be crucial to fully realize the potential of PCP-based recombinant protein production systems.

7.2. Transient Expression in Hairy Root Cultures

Hairy root cultures, induced by Rhizobium rhizogenes, provide a stable and highly efficient platform for transient expression [158]. This system combines the speed of transient methods with the robustness of root-specific expression. The advantages of this system include the reduction of costs due to continuous production in bioreactors and the enhancement of protein expression by high metabolic activity in root tissues. This system is used for secondary metabolite production and the expression of therapeutic proteins like enzymes and antibodies [158,159].

7.3. Bioprinted Plant Cells

The integration of 3D bioprinting with plant cells has opened new avenues for transient expression. Bioprinting creates controlled cell structures or tissues optimized for recombinant protein production [160]. The ability to produce structured tissues that enhance protein synthesis and customized scaffolds to improve cell viability and metabolic activity make this system very attractive. It finds usefulness in small-scale, on-demand protein production for diagnostics or research and prototyping novel therapeutic compounds [161].

7.4. Multi-Host Systems for Complex Protein Production

In some cases, transient expression systems now use co-cultures of plant cells with other organisms, like yeast or bacteria, to facilitate the production of complex proteins. This system has dual-expression systems where one host synthesizes the protein backbone and another adds post-translational modifications. Also, shared metabolic engineering optimizes yield and quality. It is used for the production of bispecific antibodies or glycoengineered therapeutic proteins [162,163].

8. Limitations and Future Challenges

Harnessing transient expression systems with plant viral vectors for the production of biopharmaceuticals in N. benthamiana has opened up new avenues for rapid and scalable manufacturing. However, despite considerable advancements, several limitations and future challenges remain.
One major limitation is the variability in expression levels among different constructs and batches. Factors such as infiltration efficiency, plant physiological state, and vector design critically influence recombinant protein yields [70,89]. This batch-to-batch variability can complicate downstream processing and regulatory approval, which demands highly consistent biomanufacturing standards.
Another significant challenge is the potential for the proteolytic degradation of recombinant proteins within the plant tissues. Host proteases may recognize and cleave foreign proteins, reducing overall yields and necessitating additional strategies such as protease inhibitor co-expression or protein engineering [164,165].
Glycosylation differences also pose a barrier for plant-produced biopharmaceuticals. N. benthamiana naturally adds plant-specific glycans, which differ from human glycosylation patterns and can affect therapeutic efficacy or immunogenicity [166]. Advances in glycoengineering, including the use of glycoengineered N. benthamiana lines, are addressing this issue, but complete human-like glycosylation is not yet fully standardized.
Scalability, while demonstrated up to several thousand liters [167], also presents technical hurdles. Large-scale infiltration requires optimized equipment and methods to maintain uniformity and avoid physical damage to plants, which could reduce yield or introduce contaminants.
Furthermore, regulatory challenges remain a critical bottleneck. Although plant-based systems offer safety advantages, such as the absence of human pathogens, regulatory frameworks are still evolving and can be complex and time-consuming to navigate [168].
Future challenges will involve improving vector design for higher and more stable expression, developing robust protease control systems, perfecting human-like glycosylation, and establishing globally harmonized regulatory pathways. Continued investment in bioprocess optimization, synthetic biology tools, and regulatory science will be crucial for the widespread commercialization of plant-made biopharmaceuticals.

9. Conclusions and Prospectives

Transient expression systems in plants, particularly in N. benthamiana, offer a rapid, versatile, and cost-effective platform for the production of biomolecules, making them essential tools in both modern medicine and scientific research. Their capacity to rapidly generate target proteins makes them especially valuable in time-sensitive scenarios such as pandemics and emerging infectious disease outbreaks. This fast turnaround has facilitated the development of modular “on-demand biofactories”, which are regional production facilities equipped with vacuum infiltration systems and controlled growth chambers, enabling agile and decentralized manufacturing of vaccines and therapeutic antibodies. These facilities provide a scalable solution to global health emergencies by ensuring localized and timely access to critical biopharmaceuticals. Moreover, transient expression systems significantly contribute to personalized medicine and synthetic biology [169,170], supporting the tailored development of treatments and enabling comprehensive genomic studies. The integration of gene editing technologies with these platforms further enhances the precision and efficiency of developing novel plant traits and therapeutic agents [171], thereby directly addressing the evolving demands of modern medicine and agriculture.
A major breakthrough enhancing the utility of this platform is glycoengineering. By knocking out plant-specific glycosylation pathways (e.g., β1,2-xylose and α1,3-fucose) and introducing components of the human glycosylation machinery, N. benthamiana can produce therapeutic proteins with human-compatible glycan profiles. When coupled with transient expression using plant viral vectors, this approach allows for the flexible, precise customization of monoclonal antibody glycoforms—improving pharmacokinetics and effector functions such as antibody-dependent cellular cytotoxicity (ADCC). Future advances in plant synthetic biology are expected to yield plug-and-play glycosylation modules, enabling tailored glycoprotein production for specific therapeutic targets and expanding global access to critical medications.
As transient expression systems in plants continue to mature, future innovations in downstream processing will be critical to unlocking their full commercial and therapeutic potential. The next generation of purification technologies is likely to focus on fully integrated, automated, and modular workflows that can operate seamlessly within decentralized “on-demand biofactories”. Advancements in synthetic biology may enable the design of expression constructs that incorporate smart purification tags with programmable cleavage sites, allowing for the selective release of target proteins under specific conditions. Additionally, plant lines could be engineered to express protease inhibitors or reduce host-derived contaminants, thereby improving extract purity at the source.
Beyond therapeutics, transient expression systems are influencing broader fields such as gene editing, synthetic biology, and personalized medicine. The integration of CRISPR and other gene editing technologies enhances the precision and efficiency of engineering both plant traits and therapeutic agents. These systems also support rapid prototyping and customization of treatments, aligning with the demands of personalized medicine. Educational applications are expanding as well; for instance, the EU Horizon 2020 Pharma-Factory consortium has developed a freely available Plant Molecular Farming (PMF) kit (https://www.ispmf.org/lab-protocols accessed on 18 May 2025) to facilitate hands-on learning, allowing students and researchers to quickly explore gene function and molecular interactions.
Looking forward, the integration of artificial intelligence and digital agriculture will further optimize plant-based bioproduction. Smart growth chambers equipped with AI-based monitoring and predictive analytics will enable dynamic control of environmental variables to maximize protein yield and quality. While N. benthamiana remains the preferred host for PMF, researchers are exploring alternatives like lettuce 60, which offers advantages such as lower levels of phenolics and alkaloids and compatibility with hydroponic cultivation. Developments in plant glycoengineering expand the range of biopharmaceuticals that can be produced, increasing global access to crucial medications. As these technologies continue to evolve, their potential to meet significant healthcare and agricultural challenges is substantial, paving the way for groundbreaking biotechnological applications.

Author Contributions

S.A.A., K.Y.W. and Z.Z. conceived the manuscript; K.H., O.S.H. and M.S. assisted in collecting part of the data and references; S.A.A. and K.Y.W. wrote the manuscript; Z.Z. revised the manuscript; Y.G. conceived the project. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from Key R&D Program of Shandong Province (2022SFGC0102) and the Agricultural Science and Technology Innovation Program (ASTIP-TRIC02). This publication was also supported in part by grant P20GM103436-24 (KY INBRE) from the National Institute of General Medical Sciences, National Institutes of Health.

Acknowledgments

We thank Bingqiang Zhang for providing valuable suggestions during the preparation of the manuscript.

Conflicts of Interest

The authors declare no competing interests.

References

  1. Buyel, J. Product safety aspects of plant molecular farming. Front. Bioeng. Biotechnol. 2023, 11, 1238917. [Google Scholar] [CrossRef] [PubMed]
  2. Buyel, J. Towards a seamless product and process development workflow for recombinant proteins produced by plant molecular farming. Biotechnol. Adv. 2024, 75, 108403. [Google Scholar] [CrossRef] [PubMed]
  3. Venkataraman, S.; Khan, I.; Habibi, P.; Le, M.; Lippert, R.; Hefferon, K. Recent advances in expression and purification strategies for plant made vaccines. Front. Plant Sci. 2023, 14, 1273958. [Google Scholar] [CrossRef] [PubMed]
  4. Zahmanova, G.; Aljabali, A.A.; Takova, K.; Minkov, G.; Tambuwala, M.M.; Minkov, I.; Lomonossoff, G.P. Green biologics: Harnessing the power of plants to produce pharmaceuticals. Int. J. Mol. Sci. 2023, 24, 17575. [Google Scholar] [CrossRef]
  5. Walsh, G. Biopharmaceutical benchmarks 2018. Nat. Biotechnol. 2018, 36, 1136–1145. [Google Scholar] [CrossRef]
  6. Leader, B.; Baca, Q.J.; Golan, D.E. Protein therapeutics: A summary and pharmacological classification. Nat. Rev. Drug Discov. 2008, 7, 21–39. [Google Scholar] [CrossRef]
  7. Pham, P.V. Medical biotechnology: Techniques and applications. In Omics Technologies and Bio-Engineering: Towards Improving Quality of Life; Barh, V.K., Azevedo, V., Eds.; Academic Press: Cambridge, MA, USA, 2018; Volume 1, pp. 449–469. [Google Scholar]
  8. Batta, A.; Kalra, B.S.; Khirasaria, R. Trends in FDA drug approvals over last 2 decades: An observational study. JFMPC 2020, 9, 105–114. [Google Scholar]
  9. de La Torre, B.G.; Albericio, F. The pharmaceutical industry in 2020. An analysis of FDA drug approvals from the perspective of molecules. Molecules 2021, 26, 627. [Google Scholar] [CrossRef]
  10. Venkataraman, S.; Hefferon, K. Application of plant viruses in biotechnology, medicine, and human health. Viruses 2021, 13, 1697. [Google Scholar] [CrossRef]
  11. Eidenberger, L.; Kogelmann, B.; Steinkellner, H. Plant-based biopharmaceutical engineering. Nat. Rev. Bioeng. 2023, 1, 426–439. [Google Scholar] [CrossRef]
  12. Cabedo Díaz, P.; Covarrubias, M.P.; Handford, M. Verifying plasmid constructs via transient Agrobacterium tumefaciens–mediated plant transformation in Nicotiana benthamiana. In Agrobacterium: Methods in Molecular Biology; Klein, C.S., Ed.; Humana: New York, NY, USA, 2025; Volume 2911, pp. 21–36. [Google Scholar]
  13. Bally, J.; Jung, H.; Mortimer, C.; Naim, F.; Philips, J.G.; Hellens, R.; Bombarely, A.; Goodin, M.M.; Waterhouse, P.M. The rise and rise of Nicotiana benthamiana: A plant for all reasons. Annu. Rev. Phytopathol. 2018, 56, 405–426. [Google Scholar] [CrossRef] [PubMed]
  14. Prakash, V.; Singh, A.; Singh, A.K.; Dalmay, T.; Chakraborty, S. Tobacco RNA-dependent RNA polymerase 1 affects the expression of defence-related genes in Nicotiana benthamiana upon Tomato leaf curl Gujarat virus infection. Planta 2020, 252, 11. [Google Scholar] [CrossRef]
  15. Yoshikawa, M.; Shirai, R.; Suzuki, T.; Matsushita, Y.; Sasaki, N. Enhanced virus infection in Nicotiana benthamiana transiently overexpressing MDP92, encoding a tobacco MYB transcription factor. JGPP 2025, 91, 69–82. [Google Scholar] [CrossRef]
  16. England, C.; TrejoMartinez, J.; PerezSanchez, P.; Karki, U.; Xu, J. Plants as biofactories for therapeutic proteins and antiviral compounds to combat COVID-19. Life 2023, 13, 617. [Google Scholar] [CrossRef]
  17. Chen, Q.; Lai, H.; Hurtado, J.; Stahnke, J.; Leuzinger, K.; Dent, M. Agroinfiltration as an effective and scalable strategy of gene delivery for production of pharmaceutical proteins. Adv. Tech. Biol. Med. 2013, 1, 103. [Google Scholar] [CrossRef]
  18. Kapila, J.; De Rycke, R.; Van Montagu, M.; Angenon, G. An Agrobacterium-mediated transient gene expression system for intact leaves. Plant Sci. 1997, 122, 101–108. [Google Scholar] [CrossRef]
  19. 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]
  20. Naseri, Z.; Khezri, G.; Davarpanah, S.J.; Ofoghi, H. Virus-based vectors: A new approach for the production of recombinant proteins. Appl. Biotechnol. Rep. 2019, 6, 6–14. [Google Scholar] [CrossRef]
  21. Jadhav, R.R.; Khare, D. Green biotherapeutics: Overcoming challenges in plant-based expression platforms. Plant Biotechnol. Rep. 2024, 18, 465–486. [Google Scholar] [CrossRef]
  22. Tran, H.H. Developing a Plant Virus-Based Expression System for the Expression Of Vaccines against Porcine Reproductive and Respiratory Syndrome Virus. Ph.D. Thesis, The University of Western Ontario, London, ON, Canada, 2017. [Google Scholar]
  23. Gleba, Y.; Klimyuk, V.; Marillonnet, S. Magnifection—A new platform for expressing recombinant vaccines in plants. Vaccine 2005, 23, 2042–2048. [Google Scholar] [CrossRef]
  24. 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]
  25. Peyret, H.; Lomonossoff, G.P. The pEAQ vector series: The easy and quick way to produce recombinant proteins in plants. Plant Mol. Biol. 2013, 83, 51–58. [Google Scholar] [CrossRef]
  26. Peyret, H.; Brown, J.K.M.; Lomonossoff, G.P. Lomonossoff. Improving plant transient expression through the rational design of synthetic 5′ and 3′ untranslated regions. Plant Methods 2019, 15, 108. [Google Scholar] [CrossRef] [PubMed]
  27. Rozov, S.; Deineko, E. Recombinant VLP vaccines synthesized in plant expression systems: Current updates and prospects. Mol. Biol. 2024, 58, 402–418. [Google Scholar] [CrossRef]
  28. Mardanova, E.S.; Vasyagin, E.A.; Ravin, N.V. Virus-like particles produced in plants: A promising platform for recombinant vaccine development. Plants 2024, 13, 3564. [Google Scholar] [CrossRef]
  29. Diamos, A.G.; Mason, H.S. Modifying the replication of geminiviral vectors reduces cell death and enhances expression of biopharmaceutical proteins in Nicotiana benthamiana leaves. Front. Plant Sci. 2019, 9, 1974. [Google Scholar] [CrossRef]
  30. Rattanapisit, K.; Shanmugaraj, B.; Manopwisedjaroen, S.; Purwono, P.B.; Siriwattananon, K.; Khorattanakulchai, N.; Hanittinan, O.; Boonyayothin, W.; Thitithanyanont, A.; Smith, D.R.; et al. Rapid production of SARS-CoV-2 receptor binding domain (RBD) and spike specific monoclonal antibody CR3022 in Nicotiana benthamiana. Sci. Rep. 2020, 10, 17698. [Google Scholar] [CrossRef]
  31. Egelkrout, E.; Rajan, V.; Howard, J.A. Overproduction of recombinant proteins in plants. Plant Sci. 2012, 184, 83–101. [Google Scholar] [CrossRef]
  32. Nerkar, G.; Suresha, G.; Ram, B.; Appunu, C. Key Challenges in Developing Products from Transgenic Plants. In Advances in Plant Transgenics: Methods and Applications; Sathishkumar, R., Kumar, S.R., Hema, J., Baskar, V., Eds.; Springer Nature Singapore Pte Ltd.: Singapore, 2019; pp. 315–331. [Google Scholar]
  33. Nikolov, Z.L.; Regan, J.T.; Dickey, L.F.; Woodard, S.L. Purification of monoclonal antibodies from plants. In Process Scale Purification of Antibodies, 2nd ed.; Gottschalk, U., Ed.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2017; pp. 631–653. [Google Scholar]
  34. Szeto, W.; Hamer, D.; Carlson, P.; Thomas, C., Jr. Cloning of cauliflower mosaic virus (CLMV) DNA in Escherichia coli. Science 1977, 196, 210–212. [Google Scholar] [CrossRef]
  35. Klein, T.M.; Wolf, E.D.; Wu, R.; Sanford, J.C. High-velocity microprojectiles for delivering nucleic acids into living cells. Nature 1987, 327, 70–73. [Google Scholar] [CrossRef]
  36. Marcotte, W.R., Jr.; Bayley, C.C.; Quatrano, R.S. Regulation of a wheat promoter by abscisic acid in rice protoplasts. Nature 1988, 335, 454–457. [Google Scholar] [CrossRef]
  37. Gronenborn, B.; Matzeit, V. Plant gene vectors and genetic transformation: Plant viruses as vectors. In Molecular Biology of Plant Nuclear Genes; Daniell, H., Streatfield, S.J., Eds.; Elsevier: Amsterdam, The Netherlands, 1989; pp. 69–100. [Google Scholar]
  38. 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] [PubMed]
  39. Joshi, R.L.; Joshi, V.; Ow, D. BSMV genome mediated expression of a foreign gene in dicot and monocot plant cells. EMBO J. 1990, 9, 2663–2669. [Google Scholar] [CrossRef] [PubMed]
  40. Donson, J.; Kearney, C.; Hilf, M.A.; Dawson, W. Systemic expression of a bacterial gene by a tobacco mosaic virus-based vector. Proc. Natl. Acad. Sci. USA 1991, 88, 7204–7208. [Google Scholar] [CrossRef]
  41. Tang, X.; Frederick, R.D.; Zhou, J.; Halterman, D.A.; Jia, Y.; Martin, G.B. Initiation of plant disease resistance by physical interaction of AvrPto and Pto kinase. Science 1996, 274, 2060–2063. [Google Scholar] [CrossRef]
  42. Schöb, H.; Kunz, C.; Meins Jr, F. Silencing of transgenes introduced into leaves by agroinfiltration: A simple, rapid method for investigating sequence requirements for gene silencing. Mol. Gen. Genet. 1997, 256, 581–585. [Google Scholar] [CrossRef]
  43. Giritch, A.; Marillonnet, S.; Engler, C.; van Eldik, G.; Botterman, J.; Klimyuk, V.; Gleba, Y. Rapid high-yield expression of full-size IgG antibodies in plants coinfected with noncompeting viral vectors. Proc. Natl. Acad. Sci. USA 2006, 103, 14701–14706. [Google Scholar] [CrossRef]
  44. 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]
  45. Erokhina, T.; Ryabukhina, E.V.; Lyapina, I.S.; Ryazantsev, D.Y.; Zavriev, S.K.; Morozov, S.Y. Promising Biotechnological Applications of the Artificial Derivatives Designed and Constructed from Plant microRNA Genes. Plants 2025, 14, 325. [Google Scholar] [CrossRef]
  46. Komarova, T.; Skulachev, M.; Zvereva, A.; Schwartz, A.; Dorokhov, Y.L.; Atabekov, J. New viral vector for efficient production of target proteins in plants. Biochemistry 2006, 71, 846–850. [Google Scholar] [CrossRef]
  47. Merwaiss, F.; Lozano-Sanchez, E.; Zulaica, J.; Rusu, L.; Vazquez-Vilar, M.; Orzáez, D.; Rodrigo, G.; Geller, R.; Daròs, J.A. Plant virus-derived nanoparticles decorated with genetically encoded SARS-CoV-2 nanobodies display enhanced neutralizing activity. Plant Biotechnol. J. 2024, 22, 876–891. [Google Scholar] [CrossRef] [PubMed]
  48. Sainsbury, F.; Lomonossoff, G.P. Extremely high-level and rapid transient protein production in plants without the use of viral replication. Plant Physiol. 2008, 148, 1212–1218. [Google Scholar] [CrossRef]
  49. Huang, Z.; Chen, Q.; Hjelm, B.; Arntzen, C.; Mason, H. A DNA replicon system for rapid high-level production of virus-like particles in plants. Biotechnol. Bioeng. 2009, 103, 706–714. [Google Scholar] [CrossRef]
  50. Mor, T.S.; Moon, Y.S.; Palmer, K.E.; Mason, H.S. Geminivirus vectors for high-level expression of foreign proteins in plant cells. Biotechnol. Bioeng. 2003, 81, 430–437. [Google Scholar] [CrossRef]
  51. García Pérez, E. Development of a Copper Sensor and Geminivirus-Based Processors for Engineering Synthetic Gene Circuits in Plants. Ph.D. Thesis, Universitat Politècnica de València, Valencia, Spain, December 2024. [Google Scholar]
  52. Marillonnet, S.; Thoeringer, C.; Kandzia, R.; Klimyuk, V.; Gleba, Y. Systemic Agrobacterium tumefaciens–mediated transfection of viral replicons for efficient transient expression in plants. Nat. Biotechnol. 2005, 23, 718–723. [Google Scholar] [CrossRef]
  53. Fox, J.L. First plant-made biologic approved. Nat. Biotechnol. 2012, 30, 472–473. [Google Scholar] [CrossRef]
  54. Zeitlin, L.; Pettitt, J.; Scully, C.; Bohorova, N.; Kim, D.; Pauly, M.; Hiatt, A.; Ngo, L.; Steinkellner, H.; Whaley, K.J. Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proc. Natl. Acad. Sci. USA 2011, 108, 20690–20694. [Google Scholar] [CrossRef] [PubMed]
  55. Phoolcharoen, W.; Dye, J.M.; Kilbourne, J.; Piensook, K.; Pratt, W.D.; Arntzen, C.J.; Chen, Q.; Mason, H.S.; Herbst-Kralovetz, M.M. A nonreplicating subunit vaccine protects mice against lethal Ebola virus challenge. Proc. Natl. Acad. Sci. USA 2011, 108, 20695–20700. [Google Scholar] [CrossRef]
  56. Arntzen, C. Plant-made pharmaceuticals: From ‘Edible Vaccines’ to Ebola therapeutics. Plant Biotechnol. J. 2015, 13, 1013. [Google Scholar] [CrossRef]
  57. Dhama, K.; Natesan, S.; Iqbal Yatoo, M.; Patel, S.K.; Tiwari, R.; Saxena, S.K.; Harapan, H. Plant-based vaccines and antibodies to combat COVID-19: Current status and prospects. Hum. Vaccines Immunother. 2020, 16, 2913–2920. [Google Scholar] [CrossRef]
  58. Hager, K.J.; Pérez Marc, G.; Gobeil, P.; Diaz, R.S.; Heizer, G.; Llapur, C.; Makarkov, A.I.; Vasconcellos, E.; Pillet, S.; Riera, F. Efficacy and safety of a recombinant plant-based adjuvanted COVID-19 vaccine. N. Engl. J. Med. 2022, 386, 2084–2096. [Google Scholar] [CrossRef] [PubMed]
  59. Benvenuto, E.; Broer, I.; D’Aoust, M.-A.; Hitzeroth, I.; Hundleby, P.; Menassa, R.; Oksman-Caldentey, K.-M.; Peyret, H.; Salgueiro, S.; Saxena, P. Plant molecular farming in the wake of the closure of Medicago Inc. Nat. Biotechnol. 2023, 41, 893–894. [Google Scholar] [CrossRef]
  60. Chung, Y.H.; Church, D.; Koellhoffer, E.C.; Osota, E.; Shukla, S.; Rybicki, E.P.; Pokorski, J.K.; Steinmetz, N.F. Integrating plant molecular farming and materials research for next-generation vaccines. Nat. Rev. Mater. 2022, 7, 372–388. [Google Scholar] [CrossRef] [PubMed]
  61. Nosaki, S.; Hoshikawa, K.; Ezura, H.; Miura, K. Transient protein expression systems in plants and their applications. Plant Biotechnol. 2021, 38, 297–304. [Google Scholar] [CrossRef]
  62. Castells-Graells, R.; Lomonossoff, G.P. Plant-based production can result in covalent cross-linking of proteins. Plant Biotechnol. J. 2021, 19, 1095. [Google Scholar] [CrossRef] [PubMed]
  63. Lindbo, J.A. TRBO: A high-efficiency tobacco mosaic virus RNA-based overexpression vector. Plant Physiol. 2007, 145, 1232–1240. [Google Scholar] [CrossRef]
  64. Diamos, A.G.; Hunter, J.G.; 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. 2020, 7, 472. [Google Scholar] [CrossRef] [PubMed]
  65. Pantazica, A.M.; van Eerde, A.; Dobrica, M.O.; Caras, I.; Ionescu, I.; Costache, A.; Tucureanu, C.; Steen, H.; Lazar, C.; Heldal, I. The “humanized” N-glycosylation pathway in CRISPR/Cas9-edited Nicotiana benthamiana significantly enhances the immunogenicity of a S/preS1 Hepatitis B Virus antigen and the virus-neutralizing antibody response in vaccinated mice. Plant Biotechnol. J. 2023, 21, 1176–1190. [Google Scholar] [CrossRef]
  66. Strasser, R. Engineering of human-type O-glycosylation in Nicotiana benthamiana plants. Bioengineered 2013, 4, 191–196. [Google Scholar] [CrossRef]
  67. Coates, R.J.; Scofield, S.; Young, M.T. Incorporation of regulatory DNA elements within a viral vector improves recombinant protein expression in plants. Sci. Rep. 2024, 14, 28865. [Google Scholar] [CrossRef]
  68. Hefferon, K. Plant virus expression vectors: A powerhouse for global health. Biomedicines 2017, 5, 44. [Google Scholar] [CrossRef]
  69. Fujiuchi, N.; Matoba, N.; Fujiwara, K.; Matsuda, R. Effects of lighting conditions on Agrobacterium-mediated transient expression of recombinant hemagglutinin in detached Nicotiana benthamiana leaves inoculated with a deconstructed viral vector. Plant Cell Tiss. Organ Cult. 2021, 145, 679–688. [Google Scholar] [CrossRef]
  70. Gleba, Y.; Klimyuk, V.; Marillonnet, S. Viral vectors for the expression of proteins in plants. Curr. Opin. Biotechnol. 2007, 18, 134–141. [Google Scholar] [CrossRef]
  71. Shcherbakov, D.N.; Isaeva, A.A.; Mustaev, E.A. Treatment of Ebola Virus Disease: From Serotherapy to the Use of Monoclonal Antibodies. Antibodies 2025, 14, 22. [Google Scholar] [CrossRef]
  72. Xie, D.; Cao, L.; Guo, M.; Wang, L.; Zhang, X.; Huang, S. Study on the Recombinant Human Interferon α1b, α2b, and Gamma Transient Expression and in Vitro Activities in Tobacco. J. Interferon Cytokine Res. 2024, 44, 467–475. [Google Scholar] [CrossRef] [PubMed]
  73. Keshvari, T.; Melnik, S.; Sun, L.; Niazi, A.; Aram, F.; Moghadam, A.; Kogelmann, B.; Wozniak-Knopp, G.; Kallolimath, S.; Ramezani, A. Efficient expression of functionally active aflibercept with designed N-glycans. Antibodies 2024, 13, 29. [Google Scholar] [CrossRef]
  74. Modarresi, M.; Javaran, M.J.; Shams-Bakhsh, M.; Zeinali, S.; Behdani, M.; Mirzaee, M. Transient expression of anti-VEFGR2 nanobody in Nicotiana tabacum and N. benthamiana. 3 Biotech 2018, 8, 484. [Google Scholar] [CrossRef] [PubMed]
  75. Kallolimath, S.; Palt, R.; Föderl-Höbenreich, E.; Sun, L.; Chen, Q.; Pruckner, F.; Eidenberger, L.; Strasser, R.; Zatloukal, K.; Steinkellner, H. Glyco engineered pentameric SARS-CoV-2 IgMs show superior activities compared to IgG1 orthologues. Front. Immunol. 2023, 14, 1147960. [Google Scholar] [CrossRef] [PubMed]
  76. Leroux-Roels, I.; Maes, C.; Joye, J.; Jacobs, B.; Jarczowski, F.; Diessner, A.; Janssens, Y.; Waerlop, G.; Tamminen, K.; Heinimäki, S. A randomized, double-blind, placebo-controlled, dose-escalating phase I trial to evaluate safety and immunogenicity of a plant-produced, bivalent, recombinant norovirus-like particle vaccine. Front. Immunol. 2022, 13, 1021500. [Google Scholar] [CrossRef]
  77. Tusé, D.; Malm, M.; Tamminen, K.; Diessner, A.; Thieme, F.; Jarczowski, F.; Blazevic, V.; Klimyuk, V. Safety and immunogenicity studies in animal models support clinical development of a bivalent norovirus-like particle vaccine produced in plants. Vaccine 2022, 40, 977–987. [Google Scholar] [CrossRef]
  78. 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] [PubMed]
  79. Yamamoto, T.; Hoshikawa, K.; Ezura, K.; Okazawa, R.; Fujita, S.; Takaoka, M.; Mason, H.S.; Ezura, H.; Miura, K. Improvement of the transient expression system for production of recombinant proteins in plants. Sci. Rep. 2018, 8, 4755. [Google Scholar] [CrossRef]
  80. Ceballo, Y.; López, A.; González, C.E.; Ramos, O.; Andújar, I.; Martínez, R.U.; Hernández, A. Transient production of receptor-binding domain of SARS-CoV-2 in Nicotiana benthamiana plants induces specific antibodies in immunized mice. Mol. Biol. Rep. 2022, 49, 6113–6123. [Google Scholar] [CrossRef]
  81. Bulaon, C.J.I.; Khorattanakulchai, N.; Rattanapisit, K.; Sun, H.; Pisuttinusart, N.; Strasser, R.; Tanaka, S.; Soon-Shiong, P.; Phoolcharoen, W. Antitumor effect of plant-produced anti-CTLA-4 monoclonal antibody in a murine model of colon cancer. Front. Plant Sci. 2023, 14, 1149455. [Google Scholar] [CrossRef]
  82. Rattanapisit, K.; Bulaon, C.J.I.; Strasser, R.; Sun, H.; Phoolcharoen, W. In vitro and in vivo studies of plant-produced Atezolizumab as a potential immunotherapeutic antibody. Sci. Rep. 2023, 13, 14146. [Google Scholar] [CrossRef]
  83. Krittanai, S.; Rattanapisit, K.; Bulaon, C.J.I.; Pitaksajjakul, P.; Keadsanti, S.; Ramasoota, P.; Strasser, R.; Phoolcharoen, W. Nicotiana benthamiana as a potential source for producing anti-dengue virus D54 neutralizing therapeutic antibody. Biotechnol. Rep. 2024, 42, e00844. [Google Scholar] [CrossRef]
  84. 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; Gleba, Y., Klimyuk, V., Eds.; Springer: Berlin/Heidelberg, Germany, 2012; Volume 375, pp. 127–154. [Google Scholar]
  85. Hoshikawa, K.; Fujita, S.; Renhu, N.; Ezura, K.; Yamamoto, T.; Nonaka, S.; Ezura, H.; Miura, K. Efficient transient protein expression in tomato cultivars and wild species using agroinfiltration-mediated high expression system. Plant Cell Rep. 2019, 38, 75–84. [Google Scholar] [CrossRef] [PubMed]
  86. Lai, H.; He, J.; Engle, M.; Diamond, M.S.; Chen, Q. Robust production of virus-like particles and monoclonal antibodies with geminiviral replicon vectors in lettuce. Plant Biotechnol. J. 2012, 10, 95–104. [Google Scholar] [CrossRef] [PubMed]
  87. Diamos, A.G.; Mason, H.S. High-level expression and enrichment of norovirus virus-like particles in plants using modified geminiviral vectors. Protein Expr. Purif. 2018, 151, 86–92. [Google Scholar] [CrossRef]
  88. 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, 1, 38–46. [Google Scholar] [CrossRef]
  89. 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, 77, 50521. [Google Scholar]
  90. Nandi, S.; Kwong, A.T.; Holtz, B.R.; Erwin, R.L.; Marcel, S.; McDonald, K.A. Techno-economic analysis of a transient plant-based platform for monoclonal antibody production. mAbs 2016, 8, 1456–1466. [Google Scholar] [CrossRef]
  91. Fernández, F.J.; Gómez, S.; Vega, M.C. High-throughput protein production in yeast. In High-Throughput Protein Production and Purification: Methods and Protocols; Chen, Y.W., Ed.; Springer: New York, NY, USA, 2019; pp. 69–91. [Google Scholar]
  92. Tominaga, M.; Shima, Y.; Nozaki, K.; Ito, Y.; Someda, M.; Shoya, Y.; Hashii, N.; Obata, C.; Matsumoto-Kitano, M.; Suematsu, K. Designing strong inducible synthetic promoters in yeasts. Nat. Commun. 2024, 15, 10653. [Google Scholar] [CrossRef]
  93. Moon, K.-B.; Park, J.-S.; Park, Y.-I.; Song, I.-J.; Lee, H.-J.; Cho, H.S.; Jeon, J.-H.; Kim, H.S. Development of systems for the production of plant-derived biopharmaceuticals. Plants 2019, 9, 30. [Google Scholar] [CrossRef]
  94. Pogue, G.P.; Vojdani, F.; Palmer, K.E.; Hiatt, E.; Hume, S.; Phelps, J.; Long, L.; Bohorova, N.; Kim, D.; Pauly, M.; et al. Production of pharmaceutical-grade recombinant aprotinin and a monoclonal antibody product using plant-based transient expression systems. Plant Biotechnol. J. 2010, 8, 638–654. [Google Scholar] [CrossRef] [PubMed]
  95. De Marco, A. Recent advances in recombinant production of soluble proteins in E. coli. Microb. Cell Factories 2025, 24, 21. [Google Scholar] [CrossRef]
  96. dos Santos, P.B.; Léo, P.; de Souza Oliveira, R.P.; Stephano, M.A. Mammalian cell culture technology. In Pharmaceutical Biotechnology; Galdino, A.S., Meneguello, L., Eds.; CRC Press: Boca Raton, FL, USA, 2021; pp. 195–223. [Google Scholar]
  97. Mukherjee, S.; Malik, P.; Mukherjee, T.K. Mammalian Cells, Tissues and Organ Culture: Applications. In Practical Approach to Mammalian Cell and Organ Culture; Lim, Y.B., Ed.; Springer Nature: Singapore, 2023; pp. 837–915. [Google Scholar]
  98. Ceaglio, N.; Oggero, M. Development and Production of Protein-Based Biotherapeutics in Mammalian Cells. In Topics in Medicinal Chemistry; Springer: Cham, Switzerland, 2024; Volume 42, pp. 1–58. [Google Scholar]
  99. Joshi, M.; Verma, P.; Mago, P.; Agrawal, Y.; Gunwal, I.; Khurana, S.; Tanwar, J.; Yadav, U.; Yadav, A. Recombinant protein expression in mammalian cells. In Fundamentals of Recombinant Protein Production, Purification and Characterization; Yadav, D., Guldhe, A., Kudanga, T., Eds.; Academic Press: Cambridge, MA, USA, 2025; pp. 79–102. [Google Scholar]
  100. Zitzmann, J.; Sprick, G.; Weidner, T.; Schreiber, C.; Czermak, P. Process optimization for recombinant protein expression in insect cells. In New Insights into Cell Culture Technology, 1st ed.; Gowder, S.J.T., Ed.; InTech: Rijeka, Croatia, 2017; pp. 43–97. [Google Scholar]
  101. Cox, M.M. Innovations in the insect cell expression system for industrial recombinant vaccine antigen production. Vaccines 2021, 9, 1504. [Google Scholar] [CrossRef]
  102. Ma, K.; Deng, L.; Wu, H.; Fan, J. Towards green biomanufacturing of high-value recombinant proteins using promising cell factory: Chlamydomonas reinhardtii chloroplast. Bioresour. Bioprocess. 2022, 9, 83. [Google Scholar] [CrossRef] [PubMed]
  103. Malla, A.; Rosales-Mendoza, S.; Phoolcharoen, W.; Vimolmangkang, S. Efficient transient expression of recombinant proteins using DNA viral vectors in freshwater microalgal species. Front. Plant Sci. 2021, 12, 650820. [Google Scholar] [CrossRef]
  104. Kim, J.Y.; Kim, Y.-G.; Lee, G.M. CHO cells in biotechnology for production of recombinant proteins: Current state and further potential. Appl. Microbiol. Biotechnol. 2012, 93, 917–930. [Google Scholar] [CrossRef]
  105. O’Flaherty, R.; Bergin, A.; Flampouri, E.; Mota, L.M.; Obaidi, I.; Quigley, A.; Xie, Y.; Butler, M. Mammalian cell culture for production of recombinant proteins: A review of the critical steps in their biomanufacturing. Biotechnol. Adv. 2020, 43, 107552. [Google Scholar] [CrossRef] [PubMed]
  106. Niazi, S.K.; Magoola, M. Advances in Escherichia coli-based therapeutic protein expression: Mammalian conversion, continuous manufacturing, and cell-free production. Biologics 2023, 3, 380–401. [Google Scholar] [CrossRef]
  107. Fiebig, D.; Bogen, J.P.; Carrara, S.C.; Deweid, L.; Zielonka, S.; Grzeschik, J.; Hock, B.; Kolmar, H. Streamlining the transition from yeast surface display of antibody fragment immune libraries to the production as IgG format in mammalian cells. Front. Bioeng. Biotechnol. 2022, 10, 794389. [Google Scholar] [CrossRef] [PubMed]
  108. Jarvis, D.L. Baculovirus–insect cell expression systems. Methods Enzymol. 2009, 463, 191–222. [Google Scholar]
  109. Baldi, L.; Hacker, D.L.; Adam, M.; Wurm, F.M. Recombinant protein production by large-scale transient gene expression in mammalian cells: State of the art and future perspectives. Biotechnol. Lett. 2007, 29, 677–684. [Google Scholar] [CrossRef]
  110. Sahdev, S.; Khattar, S.K.; Saini, K.S. Production of active eukaryotic proteins through bacterial expression systems: A review of the existing biotechnology strategies. Mol. Cell. Biochem. 2008, 307, 249–264. [Google Scholar] [CrossRef]
  111. Cregg, J.M.; Tolstorukov, I.; Kusari, A.; Sunga, J.; Madden, K.; Chappell, T. Expression in the yeast Pichia pastoris. Methods Enzymol. 2009, 463, 169–189. [Google Scholar]
  112. Shen, X.; Pitol, A.K.; Bachmann, V.; Hacker, D.L.; Baldi, L.; Wurm, F.M. A simple plasmid-based transient gene expression method using High Five cells. J. Biotechnol. 2015, 216, 67–75. [Google Scholar] [CrossRef]
  113. Siddiqui, A.; Wei, Z.; Boehm, M.; Ahmad, N. Engineering microalgae through chloroplast transformation to produce high-value industrial products. Biotechnol. Appl. Biochem. 2020, 67, 30–40. [Google Scholar] [CrossRef]
  114. Gecchele, E.; Merlin, M.; Brozzetti, A.; Falorni, A.; Pezzotti, M.; Avesani, L. A comparative analysis of recombinant protein expression in different biofactories: Bacteria, insect cells and plant systems. J. Vis. Exp. 2015, 97, 52459. [Google Scholar] [CrossRef]
  115. Karbalaei, M.; Rezaee, S.A.; Farsiani, H. Pichia pastoris: A highly successful expression system for optimal synthesis of heterologous proteins. J. Cell. Physiol. 2020, 235, 5867–5881. [Google Scholar] [CrossRef] [PubMed]
  116. de Grahl, I.; Rout, S.S.; Maple-Grødem, J.; Reumann, S. Development of a constitutive and an auto-inducible high-yield expression system for recombinant protein production in the microalga Nannochloropsis oceanica. Appl. Microbiol. Biotechnol. 2020, 104, 8747–8760. [Google Scholar] [CrossRef]
  117. Yan, N.; Fan, C.; Chen, Y.; Hu, Z. The potential for microalgae as bioreactors to produce pharmaceuticals. Int. J. Mol. Sci. 2016, 17, 962. [Google Scholar] [CrossRef]
  118. Ahmad, N.; Mukhtar, Z. Green factories: Plastids for the production of foreign proteins at high levels. Gene Ther. Mol. Biol. 2013, 15, 14–29. [Google Scholar]
  119. Daskalova, S.M.; Radder, J.E.; Cichacz, Z.A.; Olsen, S.H.; Tsaprailis, G.; Mason, H.; Lopez, L.C. Engineering of N. benthamiana L. plants for production of N-acetylgalactosamine-glycosylated proteins-towards development of a plant-based platform for production of protein therapeutics with mucin type O-glycosylation. BMC Biotechnol. 2010, 10, 62. [Google Scholar]
  120. Tokmakov, A.A.; Kurotani, A.; Takagi, T.; Toyama, M.; Shirouzu, M.; Fukami, Y.; Yokoyama, S. Multiple post-translational modifications affect heterologous protein synthesis. J. Biol. Chem. 2012, 287, 27106–27116. [Google Scholar] [CrossRef] [PubMed]
  121. De Pourcq, K.; De Schutter, K.; Callewaert, N. Engineering of glycosylation in yeast and other fungi: Current state and perspectives. Appl. Microbiol. Biotechnol. 2010, 87, 1617–1631. [Google Scholar] [CrossRef]
  122. Geisler, C.; Mabashi-Asazuma, H.; Jarvis, D.L. An overview and history of glyco-engineering in insect expression systems. In Glyco-Engineering: Methods and Protocols; Castilho, A., Ed.; Springer: Berlin/Heidelberg, Germany, 2015; Volume 1321, pp. 131–152. [Google Scholar]
  123. Banerjee, A.; Ward, V. Production of recombinant and therapeutic proteins in microalgae. Curr. Opin. Biotechnol. 2022, 78, 102784. [Google Scholar] [CrossRef]
  124. Kaur, M.; Manchanda, P.; Kalia, A.; Ahmed, F.K.; Nepovimova, E.; Kuca, K.; Abd-Elsalam, K.A. Agroinfiltration mediated scalable transient gene expression in genome edited crop plants. Int. J. Mol. Sci. 2021, 22, 10882. [Google Scholar] [CrossRef]
  125. Gutiérrez-Granados, S.; Cervera, L.; Kamen, A.A.; Gòdia, F. Advancements in mammalian cell transient gene expression (TGE) technology for accelerated production of biologics. Crit. Rev. Biotechnol. 2018, 38, 918–940. [Google Scholar] [CrossRef]
  126. Khow, O.; Suntrarachun, S. Strategies for production of active eukaryotic proteins in bacterial expression system. Asian Pac. J. Trop. Biomed. 2012, 2, 159–162. [Google Scholar] [CrossRef] [PubMed]
  127. Vandermies, M.; Fickers, P. Bioreactor-scale strategies for the production of recombinant protein in the yeast Yarrowia lipolytica. Microorganisms 2019, 7, 40. [Google Scholar] [CrossRef] [PubMed]
  128. Scholz, J.; Suppmann, S. A new single-step protocol for rapid baculovirus-driven protein production in insect cells. BMC Biotechnol. 2017, 17, 83. [Google Scholar] [CrossRef]
  129. Hammel, A.; Neupert, J.; Bock, R. Optimized transgene expression in the red alga Porphyridium purpureum and efficient recombinant protein secretion into the culture medium. Plant Mol. Biol. 2024, 114, 18. [Google Scholar] [CrossRef] [PubMed]
  130. Kurotani, K.-i.; Hirakawa, H.; Shirasawa, K.; Tanizawa, Y.; Nakamura, Y.; Isobe, S.; Notaguchi, M. Genome sequence and analysis of Nicotiana benthamiana, the model plant for interactions between organisms. Plant Cell Physiol. 2023, 64, 248–257. [Google Scholar] [CrossRef]
  131. Wang, J.; Zhang, Q.; Tung, J.; Zhang, X.; Liu, D.; Deng, Y.; Tian, Z.; Chen, H.; Wang, T.; Yin, W. High-quality assembled and annotated genomes of Nicotiana tabacum and Nicotiana benthamiana reveal chromosome evolution and changes in defense arsenals. Mol. Plant. 2024, 17, 423–437. [Google Scholar] [CrossRef]
  132. Nausch, H.; Mikschofsky, H.; Koslowski, R.; Meyer, U.; Broer, I.; Huckauf, J. High-level transient expression of ER-targeted human interleukin 6 in Nicotiana benthamiana. PLoS ONE 2012, 7, e48938. [Google Scholar] [CrossRef]
  133. Park, S.H.; Ji, K.-Y.; Kim, H.M.; Ma, S.H.; Park, S.Y.; Do, J.H.; Oh, D.-B.; Kang, H.S.; Shim, J.S.; Joung, Y.H. Optimization of the human colorectal carcinoma antigen GA733-2 production in tobacco plants. Plant Biotechnol. Rep. 2021, 15, 55–67. [Google Scholar] [CrossRef]
  134. Golubova, D.; Tansley, C.; Su, H.; Patron, N. Engineering Nicotiana benthamiana as a platform for natural product biosynthesis. Curr. Opin. Plant Biol. 2024, 81, 102611. [Google Scholar] [CrossRef]
  135. Ma, X.; Li, X.; Li, Z. Transgene-free genome editing in Nicotiana benthamiana with CRISPR/Cas9 delivered by a rhabdovirus vector. In Plant Genome Engineering: Methods and Protocols; Yang, B., Harwood, W., Que, Q., Eds.; Springer: New York, NY, USA, 2023; pp. 173–185. [Google Scholar]
  136. Shaaltiel, Y.; Tekoah, Y. Plant specific N-glycans do not have proven adverse effects in humans. Nat. Biotechnol. 2016, 34, 706–708. [Google Scholar] [CrossRef]
  137. Jansing, J.; Sack, M.; Augustine, S.M.; Fischer, R.; Bortesi, L. CRISPR/Cas9-mediated knockout of six glycosyltransferase genes in Nicotiana benthamiana for the production of recombinant proteins lacking β-1, 2-xylose and core α-1, 3-fucose. Plant Biotechnol. J. 2019, 17, 350–361. [Google Scholar] [CrossRef]
  138. Larsen, J.S.; Karlsson, R.T.G.; Tian, W.; Schulz, M.A.; Matthes, A.; Clausen, H.; Petersen, B.L.; Yang, Z. Engineering mammalian cells to produce plant-specific N-glycosylation on proteins. Glycobiology 2020, 30, 528–538. [Google Scholar] [CrossRef]
  139. Kogelmann, B.; Melnik, S.; Bogner, M.; Kallolimath, S.; Stöger, E.; Sun, L.; Strasser, R.; D’Aoust, M.A.; Lavoie, P.O.; Saxena, P.; et al. A genome-edited N. benthamiana line for industrial-scale production of recombinant glycoproteins with targeted N-glycosylation. Biotechnol. J. 2024, 19, 2300323. [Google Scholar]
  140. Eidenberger, L.; Eminger, F.; Castilho, A.; Steinkellner, H. Comparative analysis of plant transient expression vectors for targeted N-glycosylation. Front. Bioeng. Biotechnol. 2022, 10, 1073455. [Google Scholar] [CrossRef] [PubMed]
  141. Knödler, M.; Buyel, J.F. Extraction and purification of malaria vaccine candidate CLCT produced by transient expression in Nicotiana benthamiana plants. Discov. Chem. Eng. 2023, 3, 14. [Google Scholar] [CrossRef]
  142. Zhou, N.; Thilakarathna, W.W.; He, Q.S.; Rupasinghe, H.V. A review: Depolymerization of lignin to generate high-value bio-products: Opportunities, challenges, and prospects. Front. Energy Res. 2022, 9, 758744. [Google Scholar] [CrossRef]
  143. Gimenes, N.C.; Silveira, E.; Tambourgi, E.B. An overview of proteases: Production, downstream processes and industrial applications. Sep. Purif. Rev. 2021, 50, 223–243. [Google Scholar] [CrossRef]
  144. Weiss, J.; Mannweiler, S.; Salminen, H. Precision processing for value-added fats and oils. Annu. Rev. Food Sci. Technol. 2025, 16, 39–61. [Google Scholar] [CrossRef]
  145. Menzel, S.; Holland, T.; Boes, A.; Spiegel, H.; Fischer, R.; Buyel, J.F. Downstream processing of a plant-derived malaria transmission-blocking vaccine candidate. Protein Expr. Purif. 2018, 152, 122–130. [Google Scholar] [CrossRef]
  146. Martin, R.; Liu, F.; Staskawicz, B. Isolation of Protein Complexes from Tobacco Leaves by a Two-Step Tandem Affinity Purification. Curr. Protoc. 2022, 2, e572. [Google Scholar] [CrossRef]
  147. Opdensteinen, P.; Lobanov, A.; Buyel, J.F. A combined pH and temperature precipitation step facilitates the purification of tobacco-derived recombinant proteins that are sensitive to extremes of either parameter. Biotechnol. J. 2021, 16, 2000340. [Google Scholar] [CrossRef] [PubMed]
  148. Stephan, A.; Hahn-Löbmann, S.; Rosche, F.; Buchholz, M.; Giritch, A.; Gleba, Y. Simple purification of Nicotiana benthamiana-produced recombinant colicins: High-yield recovery of purified proteins with minimum alkaloid content supports the suitability of the host for manufacturing food additives. Int. J. Mol. Sci. 2017, 19, 95. [Google Scholar] [CrossRef] [PubMed]
  149. Opdensteinen, P.; Clodt, J.I.; Müschen, C.R.; Filiz, V.; Buyel, J.F. A combined ultrafiltration/diafiltration step facilitates the purification of cyanovirin-N from transgenic tobacco extracts. Front. Bioeng. Biotechnol. 2019, 6, 206. [Google Scholar] [CrossRef]
  150. Faye, L.; Grünwald-Gruber, C.; Vezina, L.-P.; Gomord, V.; Morel, B. A fast and easy one-step purification strategy for plant-made antibodies using Protein A magnetic beads. Front. Plant Sci. 2024, 14, 1276148. [Google Scholar] [CrossRef]
  151. Su, H.; Van Eerde, A.; Rimstad, E.; Bock, R.; Branza-Nichita, N.; Yakovlev, I.A.; Clarke, J.L. Plant-made vaccines against viral diseases in humans and farm animals. Front. Plant Sci. 2023, 14, 1170815. [Google Scholar] [CrossRef] [PubMed]
  152. Holtz, B.R.; Berquist, B.R.; Bennett, L.D.; Kommineni, V.J.; Munigunti, R.K.; White, E.L.; Wilkerson, D.C.; Wong, K.Y.I.; Ly, L.H.; Marcel, S. Commercial-scale biotherapeutics manufacturing facility for plant-made pharmaceuticals. Plant Biotechnol. J. 2015, 13, 1180–1190. [Google Scholar] [CrossRef]
  153. Ojo, M.O.; Zahid, A. Deep learning in controlled environment agriculture: A review of recent advancements, challenges and prospects. Sensors 2022, 22, 7965. [Google Scholar] [CrossRef]
  154. Rathor, A.S.; Choudhury, S.; Sharma, A.; Nautiyal, P.; Shah, G. Empowering vertical farming through IoT and AI-Driven technologies: A comprehensive review. Heliyon 2024, 10, e34998. [Google Scholar] [CrossRef]
  155. Rademacher, T.; Sack, M.; Blessing, D.; Fischer, R.; Holland, T.; Buyel, J. Plant cell packs: A scalable platform for recombinant protein production and metabolic engineering. Plant Biotechnol. J. 2019, 17, 1560–1566. [Google Scholar] [CrossRef]
  156. Gengenbach, B.B.; Opdensteinen, P.; Buyel, J.F. Robot cookies–plant cell packs as an automated high-throughput screening platform based on transient expression. Front. Bioeng. Biotechnol. 2020, 8, 393. [Google Scholar] [CrossRef]
  157. Poborilova, Z.; Plchova, H.; Cerovska, N.; Gunter, C.J.; Hitzeroth, I.I.; Rybicki, E.P.; Moravec, T. Transient protein expression in tobacco BY-2 plant cell packs using single and multi-cassette replicating vectors. Plant Cell Rep. 2020, 39, 1115–1127. [Google Scholar] [CrossRef]
  158. Skarjinskaia, M.; Ruby, K.; Araujo, A.; Taylor, K.; Gopalasamy-Raju, V.; Musiychuk, K.; Chichester, J.A.; Palmer, G.A.; de la Rosa, P.; Mett, V. Hairy roots as a vaccine production and delivery system. In Biotechnology of Hairy Root Systems; Doran, P.M., Ed.; Springer: Berlin/Heidelberg, Germany, 2013; Volume 134, pp. 115–134. [Google Scholar]
  159. Ron, M.; Kajala, K.; Pauluzzi, G.; Wang, D.; Reynoso, M.A.; Zumstein, K.; Garcha, J.; Winte, S.; Masson, H.; Inagaki, S. Hairy root transformation using Agrobacterium rhizogenes as a tool for exploring cell type-specific gene expression and function using tomato as a model. Plant Physiol. 2014, 166, 455–469. [Google Scholar] [CrossRef] [PubMed]
  160. Varma, A.; Gemeda, H.B.; McNulty, M.J.; McDonald, K.A.; Nandi, S.; Knipe, J.M. Immobilization of transgenic plant cells towards bioprinting for production of a recombinant biodefense agent. Biotechnol. J. 2021, 16, 2100133. [Google Scholar] [CrossRef] [PubMed]
  161. Beyer, H.M.; Ramírez, V. Integrating bioprinting and optogenetic technologies for precision plant tissue engineering. Curr. Opin. Biotechnol. 2024, 89, 103193. [Google Scholar] [CrossRef]
  162. Ortega, C.; Prieto, D.; Abreu, C.; Oppezzo, P.; Correa, A. Multi-compartment and multi-host vector suite for recombinant protein expression and purification. Front. Microbiol. 2018, 9, 1384. [Google Scholar] [CrossRef] [PubMed]
  163. Bleckmann, M.; Schürig, M.; Endres, M.; Samuels, A.; Gebauer, D.; Konisch, N.; van den Heuvel, J. Identifying parameters to improve the reproducibility of transient gene expression in High Five cells. PLoS ONE 2019, 14, e0217878. [Google Scholar] [CrossRef]
  164. Grosse-Holz, F.; Madeira, L.; Zahid, M.A.; Songer, M.; Kourelis, J.; Fesenko, M.; Ninck, S.; Kaschani, F.; Kaiser, M.; van der Hoorn, R.A. Three unrelated protease inhibitors enhance accumulation of pharmaceutical recombinant proteins in Nicotiana benthamiana. Plant Biotechnol. J. 2018, 16, 1797–1810. [Google Scholar] [CrossRef]
  165. Beritza, K.; Watts, E.C.; van der Hoorn, R.A. Improving transient protein expression in agroinfiltrated Nicotiana benthamiana. New Phytol. 2024, 243, 846–850. [Google Scholar] [CrossRef]
  166. Strasser, R. Plant glycoengineering for designing next-generation vaccines and therapeutic proteins. Biotechnol. Adv. 2023, 67, 108197. [Google Scholar] [CrossRef]
  167. Buyel, J.F.; Twyman, R.M.; Fischer, R. Very-large-scale production of antibodies in plants: The biologization of manufacturing. Biotechnol. Adv. 2017, 35, 458–465. [Google Scholar] [CrossRef]
  168. Buyel, J.F. Plant molecular farming–integration and exploitation of side streams to achieve sustainable biomanufacturing. Front. Plant Sci. 2019, 9, 1893. [Google Scholar] [CrossRef] [PubMed]
  169. Sainsbury, F.; Lomonossoff, G.P. Transient expressions of synthetic biology in plants. Curr. Opin. Plant Biol. 2014, 19, 1–7. [Google Scholar] [CrossRef] [PubMed]
  170. Bibik, J.D.; Bryson, A.E.; Hamberger, B. Compartmentalized terpenoid production in plants using Agrobacterium-mediated transient expression. In Synthetic Biology: Methods and Protocols; Braman, J.C., Ed.; Springer: New York, NY, USA, 2024; Volume 2760, pp. 21–34. [Google Scholar]
  171. Singh, A.A.; Pillay, P.; Naicker, P.; Alexandre, K.; Malatji, K.; Mach, L.; Steinkellner, H.; Vorster, J.; Chikwamba, R.; Tsekoa, T.L. Transient proteolysis reduction of Nicotiana benthamiana-produced CAP256 broadly neutralizing antibodies using CRISPR/Cas9. Front. Plant Sci. 2022, 13, 953654. [Google Scholar] [CrossRef]
Figure 1. Milestones in plant transient expression technology.
Figure 1. Milestones in plant transient expression technology.
Ijms 26 05510 g001
Figure 2. Diagrammatic illustration of the process of plant transient expression in biopharmaceuticals (PMF) and expansion.
Figure 2. Diagrammatic illustration of the process of plant transient expression in biopharmaceuticals (PMF) and expansion.
Ijms 26 05510 g002
Figure 3. Schematic diagram of plant virus-based expression systems.
Figure 3. Schematic diagram of plant virus-based expression systems.
Ijms 26 05510 g003
Figure 4. Hydroponics and Controlled Environment Agriculture (CEA) are pivotal advancements in Plant Molecular Farming (PMF).
Figure 4. Hydroponics and Controlled Environment Agriculture (CEA) are pivotal advancements in Plant Molecular Farming (PMF).
Ijms 26 05510 g004
Table 3. List of companies using N. benthamiana for plant transient expression products.
Table 3. List of companies using N. benthamiana for plant transient expression products.
Company NameCountryProductVector System UsedWebsite/Reference
Icon Genetics (Denka)GermanyVarious biologicsmagnICON® systemhttps://www.icongenetics.com/
Kentucky BioProcessing (KBP)USAVaccines, antibodiesTMV, other vectors, magnICON® systemhttps://kbio.com/
Leaf Expression SystemsUKProteins, antibodies, enzymesHypertrans® Expression Systemhttps://www.leafexpressionsystems.com/
PlantForm CorporationCanadaAntibody drugs, biosimilarsvivoXPRESS®https://www.plantformcorp.com/
Cape BiologixSouth AfricaTherapeutic proteinsPtX™ viral systemhttps://capebiologix.com/
Baiya PhytopharmThailandVaccines, antibodies,
growth factors
BaiyaPharming™https://baiyaphytopharm.com/
Nomad BioSciencesGermanyAntibacterial and antiviral biologicsmagnICON® and NOMADIC™ platformshttps://www.nomadbioscience.com/
BioapplicationsSouth KoreaVaccinesmagnICON® systemhttps://www.bioapplications.global/ (accessed on 18 May 2025)
Cirsium BiosciencesUSAAAV vectorsTMV, other vectors, magnICON® systemhttps://cirsiumbio.com/
* iBio Inc.USAVaccines, therapeutic proteinsHypertrans® Expression Systemhttps://ir.ibioinc.com/
* MedicagoCanadaVaccines (e.g., COVID-19 vaccine)vivoXPRESS®[59]
Note: * Medicago has ceased operations. iBio is accelerating its transformation into AI-powered biotech company.
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

Akher, S.A.; Wang, K.Y.; Hall, K.; Hunpatin, O.S.; Shan, M.; Zhang, Z.; Guo, Y. Harnessing Transient Expression Systems with Plant Viral Vectors for the Production of Biopharmaceuticals in Nicotiana benthamiana. Int. J. Mol. Sci. 2025, 26, 5510. https://doi.org/10.3390/ijms26125510

AMA Style

Akher SA, Wang KY, Hall K, Hunpatin OS, Shan M, Zhang Z, Guo Y. Harnessing Transient Expression Systems with Plant Viral Vectors for the Production of Biopharmaceuticals in Nicotiana benthamiana. International Journal of Molecular Sciences. 2025; 26(12):5510. https://doi.org/10.3390/ijms26125510

Chicago/Turabian Style

Akher, Sayed Abdul, Kevin Yueju Wang, Kylie Hall, Oluwaseyi Setonji Hunpatin, Muhammad Shan, Zenglin Zhang, and Yongfeng Guo. 2025. "Harnessing Transient Expression Systems with Plant Viral Vectors for the Production of Biopharmaceuticals in Nicotiana benthamiana" International Journal of Molecular Sciences 26, no. 12: 5510. https://doi.org/10.3390/ijms26125510

APA Style

Akher, S. A., Wang, K. Y., Hall, K., Hunpatin, O. S., Shan, M., Zhang, Z., & Guo, Y. (2025). Harnessing Transient Expression Systems with Plant Viral Vectors for the Production of Biopharmaceuticals in Nicotiana benthamiana. International Journal of Molecular Sciences, 26(12), 5510. https://doi.org/10.3390/ijms26125510

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