Next Article in Journal
Vaccine Strategies Against RNA Viruses: Current Advances and Future Directions
Next Article in Special Issue
The Salmonella Paratyphi A O-Antigen Glycoconjugate Vaccine Is Able to Induce Antibodies with Bactericidal Activity Against a Panel of Clinical Isolates
Previous Article in Journal
Microfluidic-Chip-Based Formulation and In Vivo Evaluations of Squalene Oil Emulsion Adjuvants for Subunit Vaccines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Protein Expression Platforms and the Challenges of Viral Antigen Production

1
Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3R2, Canada
2
Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
3
Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
4
Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
5
Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
6
Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
*
Author to whom correspondence should be addressed.
Vaccines 2024, 12(12), 1344; https://doi.org/10.3390/vaccines12121344
Submission received: 20 September 2024 / Revised: 19 November 2024 / Accepted: 19 November 2024 / Published: 28 November 2024
(This article belongs to the Special Issue Recombinant Vaccine for Human and Animal Diseases)

Abstract

:
Several protein expression platforms exist for a wide variety of biopharmaceutical needs. A substantial proportion of research and development into protein expression platforms and their optimization since the mid-1900s is a result of the production of viral antigens for use in subunit vaccine research. This review discusses the seven most popular forms of expression systems used in the past decade—bacterial, insect, mammalian, yeast, algal, plant and cell-free systems—in terms of advantages, uses and limitations for viral antigen production in the context of subunit vaccine research. Post-translational modifications, immunogenicity, efficacy, complexity, scalability and the cost of production are major points discussed. Examples of licenced and experimental vaccines are included along with images which summarize the processes involved.

1. Introduction

Vaccines have been the most successful intervention to respond to infectious disease. There are several types of vaccines including live-attenuated, vectored, DNA or mRNA, killed and subunit. Each vaccine type has its strengths and weaknesses. Although live-attenuated and vectored vaccines can induce strong protective immunity, it is more difficult to demonstrate the safety of these vaccine types, impeding their development as a rapid response to emerging disease outbreaks. DNA or mRNA vaccines use the host cells to produce the antigen offering improved safety over live and vectored vaccines. Killed and subunit vaccines are safer compared to live-attenuated and vectored vaccines, with subunit vaccines considered the most safe [1,2,3,4]. Unfortunately, both killed and subunit vaccines require adjuvants to induce protective immune responses. The immunogenicity of subunit vaccines can be increased by producing virus-like particles (VLPs), which are generated using the component proteins of a virus which spontaneously self-assemble into particles in the absence of a viral genome [5]. The repetitive structure of VLPs stimulates the innate and adaptive immune responses, resulting in improved antibody responses [5,6]. An advantage of subunit vaccines, compared to conventional live-attenuated and most killed vaccines, is their potential for differentiating from infected and vaccinated animals (DIVA) when combined with companion diagnostics [7,8].
The biotechnology industry has rapidly advanced in the past half-century and the expression of recombinant proteins in different systems for a wide variety of applications is a major use of this technology. This has resulted in a range of proteins expressed for therapeutics and diagnostic kits which are discussed elsewhere [2,9,10]. These major advances in genetic engineering have also allowed for the development of recombinant subunit vaccines consisting of protective viral antigens which is the focus of this review. Successful licenced recombinant subunit vaccines have been produced in different protein expression systems such as the Hepatitis B surface antigen (HBsAg) in yeast [3], the human papilloma virus-like particle (VLP) vaccine in baculovirus [4] and several systems for influenza [11,12] and the dengue [13] in mammalian expression systems.
Advances in genetic engineering, including gene synthesis combined with the biotechnology of cell culture systems, has led to the availability of multiple different protein expression systems used to produce recombinant subunit vaccines [10]. After decades of research into the utilities of various protein expression systems, including bacterial, insect, mammalian, plant and cell-free systems, an empirical approach is still required for identifying an appropriate expression system for a specific viral glycoprotein [2,14]. Viral proteins as vaccine antigens often contain post-translational modifications including disulphide bonds and the unique glycosylation patterns that are required for proper folding and/or biological activity, preventing their expression in prokaryotes. The immunogenicity of a recombinant glycoprotein antigen can be highly dependent on its glycosylation and is influenced by the biology of the cell used and the protein being expressed [2,15,16]. Eukaryotic expression systems can be used to express these proteins, including yeast, e.g., Pichia pastoris and Saccharomyces cerevisiae; baculovirus expression vector systems (BEVSs) using Autographa californica multiple nuclear polyhedrosis virus (AcMNPV) and insect cell hosts Spodoptera frugiperda (Sf) or Trichoplusia ni (Tni); and mammalian cell systems including a variety of transformed cell lines, such as the Chinese hamster ovary (CHO) and human embryonic kidney (HEK)293 [17]. Although all these expression systems can express protein antigens, the glycosylation patterns will differ between the antigen expression systems. The effect of the glycosylation patterns from the different expression systems can be important or not depending on the specific protein use of the protein; this makes the selection of the optimal protein production system difficult without prior knowledge of expression in different systems.
Variations in glycan types and the glycosylation of proteins is a major limiting factor in recombinant antigen production as glycosylation can dramatically impact efficacy. Several glycoproteins carry glycans derived from the initiation of N-Acetylgalactosamine (GalNAc) attached to Ser or Thr residues. Mucins are a class of glycoproteins that have the highest amount of O-GalNAc glycans. These have four core structures which can be extended by sugar residues to produce either linear or branched chains, similar to what is seen on N-glycans. N-glycans are attached by covalent bonds to asparagine residues by N-glycosidic bonds. The synthesis of N-glycans is highly complex in mammalian glycoproteins. Eukaryotic N-glycans have a common core sequence and are then classified into three types: (1) oligomannose, where only mannose residues extend the core; (2) complex types, where ‘antennae’ structures derived from N-acetylglucosamine extend the core; and (3) hybrid types, where mannose extends an arm of the core and one or two N-acetylglucosamine antennae extend another arm [18].
Most viral vaccines produced by recombinant protein expression systems are viral proteins expressed on the surface [19,20,21,22], either capsid proteins in non-enveloped viruses or membrane proteins in enveloped viruses [23,24,25,26,27,28,29,30]. In terms of viral subunit vaccines, Escherichia coli (E. coli) expression systems have been attempted but have not demonstrated significant enough effectiveness to be commercially viable, though they have been highly effective for bacterial subunit vaccines [27]. Prokaryotic systems have limitations when expressing mammalian (viral) membrane proteins as post-translational modifications are required for their correct folding into a fully functional protein.
This review will discuss the different protein expression systems and their applications for viral antigen expression. In addition, future areas of research for improving protein expression systems focussing on cell-free systems and transgenic animals will be discussed.

2. Bacterial Expression Systems

E. coli is the most commonly used bacterial system for protein expression due to its simplicity in being able to use a plasmid encoding the gene of interest to transform the bacteria and the simple production and efficiency of producing protein (Figure 1 and Figure 2) [31,32,33,34,35]. The majority of viral glycoproteins are large proteins (greater than 60 kDa) that are difficult to express using E. coli. Though it has been shown that E. coli strains such as SHuffle [32] and other strains [33] can produce disulphide bonds in expressed proteins, a lack of post-translational machinery and protein expressed in inclusion bodies make E. coli expression not suitable for many viral glycoproteins [34].
Additional issues including the presence of endotoxins such as lipopolysaccharides [35] require additional bioprocessing steps [36] though some species of E. coli such as BL21 (DE3) are endotoxin-free [37]. Furthermore, even following endotoxin removal, many proteins still contain level of endotoxin able to activate dendritic cells [38], indicating the difficulty in removing endotoxin from recombinant proteins. In addition, protein expression in bacterial expression systems produces proteins which aggregate in insoluble inclusion bodies, which requires further protein purification procedures to solubilize the protein from the inclusion bodies [39]. Since certain expressed proteins may be insoluble, unstable or inactive without post-translational modifications, E. coli may only be the choice expression system for the production of small recombinant proteins without post-translational modification needs [40,41].

Applications of Bacterial Expression Systems

Several E. coli expressed viral antigen vaccine candidates have been developed and are at different stages of evaluation, as summarized in Table 1. The only licenced vaccine was developed by Xiamen Innovax Biotech Co., Ltd. (Xiamen, China) namely the hepatitis E virus (HEV) capsid polypeptide, HEV 239, from E. coli, and was the first VLP-based vaccine (Hecolin) against HEV licenced by the FDA of China in 2011 [42]. Antigen production and product consistency was demonstrated at different scales indicating the manufacturing process was robust and scalable [43].
Foot-and-mouth disease virus (FMDV) VLPs have also been produced in this system, providing protection in multiple species [44,45]. VLPs mimicking viral capsids or chimeric VLPs by grafting epitopes of interest onto a VLP display vector are possible platforms for future vaccines via structure-based molecular design. One study used calcium phosphate-mineralized VLPs (CaP-VLPs) and tested three commercial mucosal adjuvants and found that the induction of local mucosal and systemic immune responses via intranasal immunization was specifically enhanced by the adjuvant: STR products [46]. The anti-influenza A M2e-Hbc vaccine candidate, ACAM-FLU-A was produced by E. coli using the recombinant hepatitis B core antigen as a carrier VLP, which is an approach that synergizes the benefits of broadly cross-protecting epitopes with rapid scale-up vaccine manufacture using bacterial expression systems [48]. This vaccine resulted in high seroconversion rates in Phase 1 trials but Phase II efficacy trials were cancelled due to a rapid decline in antibody titres [49,50]. Recombinant VLPs for porcine circovirus type 2 capsid protein and porcine parvovirus (PPV) VP2 protein have been expressed in E. coli and have demonstrated immunogenicity in piglets and improved growth indices [51].
Since the start of the COVID-19 pandemic, several laboratories have produced SARS-CoV-2 spike and RBD fragments in E. coli expression systems [52,53,54,55,56,57]. In these systems, as described above, the proteins are produced insoluble in inclusion bodies which then require extra steps of centrifugation, ultrasonication, redissolving, refolding and then purification. In these studies, only fragments of the SARS-CoV-2 surface glycoproteins approximately 300 bp could be successfully produced. One study [55] was able to produce soluble spike and nucleocapsid protein fragments by fusing the fragments to carrier family 9 carbohydrate-binding module (CBM9) through a flexible linker. Studies using E. coli have been successful in producing the RBD of the spike protein [57]. As this system produces little glycosylation, one study [52] showed that non-glycosylated RBD adjuvanted with both 100 μg of Al(OH)3 and 50 μg of CpG elicited a robust neutralizing antibody response in mice. While the E. coli expression system is a cost-effective system to produce many useful proteins, the requirements for disulphide bond formation and glycosylation for proper expression and folding for viral proteins make this a less desirable system.

3. Insect Cell (Baculovirus) Expression Systems

Baculoviruses are large, double-stranded DNA viruses which infect insects and have been developed as a recombinant protein expression system using insect cells [58]. To produce recombinant proteins, the baculovirus, Autographa californica nuclear polyhedrosis virus (AcNPV), is genetically engineered to encode the gene of the protein of interest. These baculoviruses are then used to infect susceptible insect cells either Spodoptera frugiperda (Sf) or Trichoplusia ni (Tni) to transiently express the protein (Figure 3).
Homologous recombination is used to generate recombinant baculoviruses in insect cells by the co-transfection of viral DNA and a plasmid containing the gene of interest under the control of the strong polyhedrin promoter of the baculovirus and is flanked by sequences from the polyhedron region [59,60]. The selection of recombinant baculovirus is based on the removal of the polyhedrin gene and the detection of occlusion-negative plaques in insect cell monolayers. Several improvements have been made for the selection of recombinant baculovirus, including the linearization of the virus at the polyhedrin locus [61,62]. Recombination recircularizes the viral genome, which allows for the amplification of the genome [63]. Combinations of this approach were also used, such as the incorporation of a lacZ gene at the polyhedrin locus and the subsequent linearization or engineering of unique restriction enzyme sites into an essential gene (ORF1629) which flanks the polyhedrin locus [64]. The linearization of these sites results in defective gene expression, and the part of the ORF1629 gene that is deleted from the baculovirus genome is incorporated into the transfer plasmid (bacmid) so that ORF1629 function is restored only in the recombinant viruses [65]. The recombinant bacmid DNA is then used to transfect insect cells using a transfection reagent in small cell cultures. Infected cells then give rise to live baculovirions used to infect larger cell cultures to produce and purify the protein of interest expressed in the insect cells [66].
Post-translational modifications are similar, although not identical, to mammalian cells. Glycoproteins produced in insect systems have O- and N-glycans with structures similar to those produced by other eukaryotes, although usually insect expressed proteins do not have antennae of peripheral sugars, but are mainly composed of sialic acids, which are often found on native mammalian glycans [67]. Sialic acids are often found as terminal residues on cell surface glycoproteins and are involved in several immunological interactions in higher eukaryotes. The absence of sialic acids in proteins expressed in insect cells is a significant and somewhat controversial issue in terms of recombinant glycoprotein production. Understanding the important roles that glycan moieties play in protein expression and the need for systems that can produce mammalian-type glycoproteins has led to an increased interest in protein glycosylation pathways and host cells that can be used for functional recombinant glycoprotein expression [68].
Glycoforms of recombinant proteins produced in the BEVS are sometimes different from those produced in mammalian expression systems [69]. Insect-expressed proteins mainly display paucimannose and hybrid-type N-glycans with, at most, minor fractions of complex-type and oligomannose-type N-glycan. Tni cells compared to Sf9 cells can also produce core α-1,3-fucose-linked glycans [70], which may cause hypersensitivity reactions when applied to patients with allergy and therefore should be avoided in cell line engineering [71]. This limitation has been addressed by genetically transforming insect cell lines with constitutively expressible mammalian genes which yield transgenic insect cell lines that can produce humanized recombinant glycoproteins [72]. Genetically modified insect cells include Mimic SF9, SfSWT-1, SfSWT-3, SfSWT-4, SfSWT-5, SfB4GalT, SfRVNlec1, DpN1 and A7S [68].
The baculovirus expression system is able to produce complex antigens unable to be effectively produced in prokaryotic systems [73]. However, their use in expressing secreted mammalian proteins is limited for several reasons, the simplest being that insect cell culture media is often just as costly as mammalian cell media. Both insect and mammalian cell lines can be cultured in classical media containing serum, chemically defined media or serum-free media and the overall costs of these are nearly identical for both cell types. Mammalian cells produce glycosylation patterns on proteins that are more representative of those seen in native organisms because insect cells do not produce sialylated complex glycans; however, protein yields in both are similar [73,74]. The construction of the plasmid, amplification, titration and optimization are all time-consuming steps requiring approximately one month or more. Upon infection, the baculovirus genome including the trans-expression cassette is significantly amplified while host synthesis stops. Despite having to compete with viral genes, the expression cassette under the control of the strong viral polyhedrin promoter is itself transcribed and translated at exceptionally high levels. Complex secreted proteins such as viral glycoproteins require several host factors, including glycosylation machinery, chaperones for accurate folding and disulphide isomerases. Under these conditions, the insect cell host machinery is often overwhelmed and so secreted proteins may fail to substantially fold and are instead sometimes found trapped inside the cell in aggregates [75]. Extreme over-expression in the BEVS can result in lower yields of properly folded and secreted protein. This issue has been significantly improved, however, by the inclusion of honeybee melittin, silkworm SP1 and Drosophila Bip signal peptides in the trans-expression plasmid, which have allowed for improved and efficient recombinant protein secretion in the BEVS [76,77].
There are several studies which have produced protein antigens in insect cells without the use of the BEVS [77,78,79,80,81,82,83,84,85]. This involves the use of polyethylenimine (PEI)-based transient gene expression and has been shown in both Tni and Sf9 cells. PEI transfection is a high-efficiency nucleic acid delivery system based on transferrin receptor-mediated endocytosis to carry DNA into cells [84]. The expression vector is efficiently delivered by PEI using a variety of different expression vectors and protocols. A higher secreted protein yield has been demonstrated compared to BEVS [79,80].

Applications of Baculovirus Expression System

There are currently seven licenced BEVS-expressed subunits or VLP vaccines against human and animal viruses (Table 2) [86,87], and several other BEVS viral vaccines in development are described in Table 3. Examples of Drosophila S2 cell line-expressed vaccines are also included [88,89].
Two separate studies indicated the effectiveness of BEVS expressed VLPs produced against chikungunya virus (CHKV). One study showed that a single immunization with 1µg of non-adjuvanted CHIKV-S27 structural polyprotein (C, E3, E2, 6K, E1) induced high-titre neutralizing antibody responses and provided complete protection against viremia and joint inflammation upon challenge using an adult wild-type mouse model of CHIKV disease with the Réunion Island CHIKV strain [96]. The other compared BEVS expressed CHKV VLPs to BEVS expressed CHKV glycoproteins E1 and E2 subunits and showed that the VLPs provided a superior immune response and protection against CHKV-induced disease in mice [97].
A classical swine fever (CSF) BEVS subunit vaccine developed in Vietnam was shown to be safe to administer and effectively stimulated protective immunity against CSFV with a two-dose vaccination [98]. The subunit VN91-E2 recombinant vaccine developed from the E2 CSFV envelope glycoprotein plus complete/incomplete adjuvant was shown to protect experimental pigs against high virulence CSFV’s circulating in Vietnam, including genotypes 1.1, 1.2 and 2.2. E2-specific neutralizing antibodies appeared at day 24, peaked at day 35 and continued until day 63, but transplacental transmission was not described.
Intradermal immunization with VLPs of Ebolavirus (EBOV) GP and VP40 in guinea pigs were shown to induce broad antibody responses and all vaccinated guinea pigs survived challenge with a high dose of guinea pig-adapted EBOV, while all control animals succumbed to the challenge. Both IgG1 and IgG2 antibody responses were elicited by intradermal vaccination with BEVS-expressed EBOV VLPs representing a successful and possibly advantageous route of vaccine against EBOV infection [99].
BEVS recombinant VP2 proteins of epizootic hemorrhagic disease virus (EHDV)-2 and EHDV-6 were used as subunit vaccines in white-tailed deer against the disease. Following vaccination, no deer developed clinical disease or EHDV-induced lesions and in addition, no viral RNA was detected in blood or tissues [100].
Pandemic influenza H1N1 proteins HA, NA and M1 were produced as VLPs using the BEVS and were demonstrated to induce immunity and protect pigs from pandemic influenza challenge. The vaccinations induced robust levels of serum IgG, mucosal IgA and viral neutralizing antibodies. Following challenge with pandemic H1N1 2009, pigs showed reduced lung lesions, viral shedding and the inhibition of viral replication in the lungs compared to unvaccinated pigs [101].
Drosophila S2 cells were used to express recombinant Lassa virus (LASV) glycoprotein (GP) elicited high antibody titres when formulated with a suitable adjuvant after two doses [88]. The BEVS has been demonstrated to express Rift Valley fever virus (RVFV) Gn and Gc glycoproteins, which elicited strong neutralizing antibody responses in sheep and conferred complete protection in all vaccinated sheep demonstrated by the prevention of viremia and fever and the absence of RVFV-associated histopathological lesions [104,105].
Recently, insect cell lines and the baculovirus expression vector system have been used to produce SARS-CoV-2 proteins and VLPs with success [105,106,107,108,109,110,111,112]. SARS-CoV-2 RBD proteins have been expressed and purified in Tni insect cells in a baculovirus mediated Bac-to-Bac expression system in three separate studies, which showed high neutralizing titres [112] and protective immunity in NHPs [105,106].
The E80 and EDIII domains of Zika virus (ZIKV) were produced using the Drosophila S2 cell line and were shown to efficiently elicit protective immunity in a mouse model. Mice were protected against lethal ZIKV challenge by the passive transfer of either anti-E80 or anti-EDIII sera [89].

4. Mammalian Expression Systems

Mammalian protein expression systems have been modified and optimized over the past twenty years with Chinese hamster ovary (CHO), human embryonic kidney (HEK) and NS0 murine myeloma being the most preferred cell lines (Figure 4) [113]. The advantage to using mammalian cell lines is the requirement for post-translational modifications and the biosynthetic complexity of the proteins of interest [114]. Mammalian cells are more likely to express properly folded proteins with native post-translational modifications. Glycosylation patterns of mammalian-expressed secreted proteins are similar with those observed in vivo with minor differences between species of cell hosts [115,116]. For the expression of the protein of interest, this can be achieved by either transient expression using a plasmid or the generation of a stable cell line using lentiviral vectors. Unfortunately, most mammalian expression systems require using serum to support the growth of the cells, which is costly and requires additional safety testing of the serum used.
Lentiviral vectors play a major role in transferring the gene of interest into the mammalian cell line [117] and their broad tropism allows them to infect most mammalian cell types. Lentiviruses allow for stable integration into the host cell genome ensuring long term expression for stable cell line generation. They can infect both dividing and non-dividing cells such as hepatocytes and neurons, which are known to be difficult to infect. They can deliver transgene fragments as large as 10 kb (~350 kDa), which is well within the range of viral vaccine candidates of 100–200 kDa, and great effort has been put into developing recombinant lentiviruses for clinical and research purposes with viral vaccine development being one major use. Precautions are needed to ensure that replication-competent lentiviruses are not accidentally generated via recombination between the delivered and endogenous viral elements in the producer cells [118,119].
Lentiviral delivery systems have evolved over three generations to reduce biosafety risk. The first-generation system is no longer in use due to biosafety risks. The second-generation system splits essential components of the lentiviral system across three plasmids—transfer, packaging and envelope—that are delivered separately for safety. The transfer plasmid encodes for the transgene and contains cis-acting elements essential for the transcription of viral mRNA and genome packaging. The packaging plasmid is provided in trans and only encodes the essential trans-acting genes required for the entry and integration of the viral genome, and the envelope plasmid contains genes encoding for envelope proteins. The third generation of lentiviral vectors almost completely eliminates dangerous lentiviral recombination events by splitting the viral genome into four plasmids: the packaging plasmid containing only the packaging genes (gag and pol), a regulator plasmid containing only the regulatory gene (rev), a plasmid carrying only the envelope gene (env) and a transgene plasmid containing the gene of interest for producing the vaccine subunit protein [119,120]. Although the third generation is significantly safer, its viral yield is typically lower than that of the second-generation system.
As tropism is determined by the glycoproteins on the surface of the virus, the tropism of a lentivirus can be further broadened by replacing the HIV-1 env glycoprotein with the envelope glycoprotein (G) from the vesicular stomatitis virus (VSV-G). This widens the range of cell types the virus can bind to [116,117,118,119]. Lentiviral vectors have a comparatively high capacity for encoding transgenes and can be engineered with improved safety and efficiency parameters for high transduction efficiency, low anti-vector host immunity, low genotoxicity and persistent gene expression [121,122].
Transient gene expression is the temporary expression of genes following transfection with a plasmid using transfection reagents such as polyethylenimine (PEI) [123,124]. The use of suspension-adapted CHO and HEK cell lines, together with optimized commercial media and inexpensive transfection agents, has allowed transient transfection to be used at production scale to hundreds of litres in bioreactors [125]. This approach requires several milligrams of high-quality, purified plasmid DNA produced by E. coli, free of endotoxin and other contaminants [123,126].
Regulated, inducible expression in mammalian cells is often desirable, especially when the product is cytotoxic or cytostatic. Inducible expression is achieved using bacterial gene control elements and transactivator proteins, for example, combinations of hybrid viral transactivators, bacterial repressor proteins and simple chemical inducers provide induction ratios of over 1000-fold [127].
Stable mammalian protein expression can be induced when the transgene expression cassette is replicated extra-chromosomally using viral proteins and cis-acting elements, or when integrated into the host genome. This is usually expected for large-scale production and control over protein quality and homogeneity. Transient expression experiments require large quantities of consumable reagents such as transfection reagents and plasmid DNA. Protein yield is often dependent on the transfection efficiency, which varies considerably from one experiment or technician to the next. Protein transiently produced from older cells may not be qualitatively equivalent to protein produced in cells from an earlier passage due to the transformed nature of the cell lines used which increases protein heterogeneity and decreasing protein quality [75,128]. Transient expression requires much less time compared to establishing a stable clonal cell line. However, if several transient transfections are required, the cost of repeated transfections will make the time commitment required for the stable cell line production worthwhile [17].
Stable transgene expression can be maintained using either of two strategies: episomal expression or chromosomal integration and expression. Episomal vectors based on viral elements promote autonomous replication and retention in the nucleus and rely on a cis-acting viral origin of replication and trans-acting virally encoding protein [129]. Transgene amplification copy numbers can vary drastically, which is a factor that makes it less desirable than stable chromosomal integration [129]. Stable chromosomal integration and expression is reliant on the presence of a suitable selection marker for isolating and screening clonal cell lines. Several selection systems have been developed, such as methotrexate/DHFR and glutamine synthetase protein expression systems, as well as conventional selectable drug markers [130,131]. Stable expression is maintained by physically coupling the transgene expression cassette with a dominant selection marker. One issue with this method is that chromosomal integration is usually a random event, which in some cases results in gene silencing. These integration “position effects” are significant therefore clonal screening is necessary to achieve desirable levels of protein expression [17].
The CHO cell line is widely used due to its ability to grow at high densities in suspension cultures and its ease of adaptation to serum free conditions. This adaptability also has drawbacks as each production target requires the selection of clones that exhibit the necessary phenotypic properties. These include cell doubling time and long-term viability under bioprocess conditions to ensure product quality/uniformity. Even when a appropriate CHO clone is identified, phenotypic drift can occur, making working with CHO clones challenging [132]. For protein expression optimization in CHO cells, glycosylation has been shown to be important, since variation in glycosylation patterns affects protein function and stability, and in addition, non-natural glycoforms can be immunogenic [131,132,133,134,135]. This applies strongly to the terminal α-Gal epitope, which has been added to proteins produced in murine cell lines and is capable of inducing unwanted immune responses in humans [136]. It is therefore essential that CHO production clones are monitored to ensure proper glycosylation. The overexpression of appropriate glycosyltransferases can be used to enhance glycan quality by modifying oligosaccharide structures on a recombinant protein. Improvements are either made by increasing the homogeneity of native structures or introducing non-host cell residues to specialize glycan quality and function [2]. Medium supplementation with glucosamine and N-acetylmannosamine allowed for the glycosylation control of a recombinant glycoprotein produced in both NS0 and CHO cells [137,138].

Applications of Mammalian Expression System

Several licenced (Table 4) and experimental (Table 5) vaccines have been developed using the mammalian expression system.
The CHO cell line been used to produce a novel subunit vaccine of the CSFV-E2 recombinant fusion protein as a mucosal vaccine. A major issue with most subunit vaccines is the inability to stimulate a significant cell-mediated response, but this study was able to show that the CHO-expressed CSFV-E2-FC protein enhanced IgA levels and induced a CSFV-specific T cell immune response and Th1-biased cellular immune response in pigs. Notably, the efficacy of this mucosal subunit vaccine was found to be comparable to the commercial live-attenuated CSFV C-strain vaccine [152].
A Hendra virus (HeV) G glycoprotein subunit vaccine produced using a soluble form of the HeV attachment (G) envelope glycoprotein (sGHeV) made in human HeLa [155,161] and human 293F [156] cells demonstrated protective efficacy in African Green Monkeys [154], cats [155] and ferrets [156] and is currently used to protect horses against Hendra virus in Australia [157]. All studies used CpG oligodeoxynucleotide and AlhydrogelTM as adjuvants. Nipah virus (NiV) and HeV are both closely related, zoonotic paramyxoviruses. In African Green Monkeys, vaccination provided complete protection against subsequent NiV infection with no evidence of clinical disease, virus replication or pathology observed in any challenged subjects [154]. Mucosal immunity was generated in cats vaccinated with recombinant sGHeV as all vaccinated cats possessed antigen-specific IgA on the mucosa. Upon oronasal challenge with NiV, all vaccinated cats were protected from disease even though virus was detected on day 21 post challenge in one cat [155].
Since the start of the COVID-19 pandemic, several studies have been conducted using mammalian expression systems to produce SARS-CoV-2 proteins for serological and vaccine research [159,160,161,162,163,164,165,166,167]. The HPLC analysis of RBD produced in HEK-293T cells was highly homogenous with a sharp elution peak in a reverse phase C18 column and was shown to produce soluble and properly folded polypeptides [163]. Another study, also using HEK-293 cells produced S1 and N proteins that were effective to identify sera from patients positive for COVID-19 by ELISA and immunofluorescence assays [168]. The use of engineered ferritin fused to viral immunogens has also been used to produce RBD vaccines against SARS-CoV-2 [159]. Here, the human codon-optimized RBD was fused to the IL-2 signal peptide at the amino terminus and ferritin at the C terminus to generate an RBD-ferritin fusion. These fusion proteins were readily purified from supernatants of transfected HEK-293 cells. Ferrets were then immunized with the RBD-ferritin fusion plus AddaVax adjuvant and challenged with two different doses of SARS-CoV-2. At challenge doses, the RBD-nanoparticle immunized animals showed strong neutralizing antibody responses and rapid viral clearance in nasal washes and lung tissue vs. control animals. Another study involved the use of an HEK-generated, pan-HLA-DR monoclonal antibody fused to SARS-CoV-2 RBD used as a vaccine in ferrets, which elicited robust protection, strong neutralizing antibody responses and viral clearance in ferret nasal washes vs. control animals [160].

5. Yeast Expression Systems

Yeast is the simplest unicellular eukaryotic organism, and the species Saccharomyces cerevisiae is just as well studied and characterized as E. coli. Hepatitis B virus and HPV recombinant subunit vaccines produced in yeast are the first approved recombinant vaccines for human use [169,170]. The two major systems used for yeast protein expression are S. cerevisiae and Pichia pastoris (Figure 5) [2]. Yeast systems have been able to produce high yields of secreted recombinant protein while also being fast and relatively inexpensive [171,172]. Culture media for yeast systems can be much less expensive than media for insect or mammalian cell culture, and yeast can easily be cultured to high densities in reusable shaker flasks or at higher densities in oxygen-sparged fermenters. The time commitment for developing a yeast expression cell line is relatively low, cell doubling is rapid and expression experiments are relatively straightforward [173,174]. One well-documented setback is that yeast express several proteases that can lead to significant, but often protein-dependent, protein degradation, though protease deficient cell lines do exist [174,175].
Beyond S. cerevisiae, there are other yeast species (P. pastoris, Yarrowia lipolytica and Hansenula polymorpha) that are ‘generally regarded as safe’ (GRAS) for the production of therapeutics and heterologous proteins [176]. Their lower hyper-mannosylation compared to S. cerevisiae makes them good candidates for the secretion of human glycoproteins, especially by engineering their glycosylation patterns to resemble human post-translational modification pathways [177]. Both Pichia and Saccharomyces construct high-mannose-type N-glycosylations on proteins and are incapable of producing more complex glycosylation patterns often needed for efficient bioactivity in mammalian proteins [178]. This may be important, depending on the specific protein being produced. Proteins produced in yeast have a large number of O-glycans and it is usually difficult to predict glycosylation sites. N-glycosylation has been widely studied in yeast, and the signalling pathway has been found to be similar to that in mammalian cells. Glycosyltransferases in mammalian cells are significantly higher than in yeast such as S. cerevisiae. Glycosylated proteins in yeast species are also significantly longer than in mammalian cells but the components in mammalian cells are more complex. The engineering of N-glycosylation pathways in several yeast species has allowed for the production of proteins with hybrid N-glycosylation, galactosylation and sialyation complex glycans [179].
Though yeast can provide a robust expression system for many secreted proteins, their divergence from native mammalian post-translational modifications and variability in levels of expression renders them problematic for viral vaccine development, which usually requires the expression of complex mammalian viral surface glycoproteins. Sometimes more than 100 mannose residues are attached, which is called hyper-glycosylation. The artificial in vivo glycoengineering of P. pastoris strains has allowed for the development of practical tools for designer glycosylation pathways. This would allow yeast to produce different kinds of glycosylated proteins [180] valuable for recombinant viral vaccine production. Large amounts of protein secreted into an almost protein-free medium makes scaling-up, as well as downstream purification, a much easier task. Pichia also does not produce endotoxin, which is a major problem in bacterial expression, and since proteins produced in this yeast system are usually folded correctly and secreted into the medium, the fermentation of genetically engineered P. pastoris is an alternative to the E.coli system [181]. Yeast cells are also known to be able to enhance expression of MHC and costimulatory molecules on the surface of dendritic cells or antigen presenting cells, which allows for the efficient activation of T lymphocytes promoting cell-mediated immunity [178,179,180]. Though most yeast expression systems produce oligomannose glycans, the polymannose forms of these can be produced and result in strong inflammatory responses in humans. These are sensed by pattern recognition receptors on innate immune cells [182,183,184]. It has been seen that mannans lacking β-1,2-mannoside residues tend to induce the production of higher levels of inflammatory cytokines, such as IL-6 and TNF-α, in mouse DCs [185].
There is an emerging trend toward using P. pastoris over S. cerevisiae as the choice yeast host species for biopharmaceutical production made evident and as a result of the increased number of patents and publications using P. pastoris (see Applications). Host improvements via synthetic biology applications for improved promoters and secretion signals, the development of protease-deficient strains and the humanization of glycosylation patterns can further enhance the recombinant vaccine production potential of several yeast species and has been well-documented in P. pastoris. A further advantage includes an increasing toolkit, including the in silico mathematical models of metabolism and genomic approaches for the identification of systems regulating protein production that support the use of P.pastoris for biopharmaceutical production [186].

Applications of Yeast Expression System

The best-known yeast-expressed vaccine is the licenced quadrivalent HPV vaccine Gardasil, which contains VLPs of hrHPV16 and 18 and lrHPV6 and 11, produced in yeast with aluminum hydroxyphosphate sulphate as an adjuvant [187]. Some of the more recent antigens expressed by various species of yeast are shown in Table 6.
Hepatitis C virus envelope glycoproteins E1E2, in both S. cerevisiae and H. polymorpha as vaccine candidates, have also been included in pending patents. The H. polymorpha system produced E1E2 proteins, which were not hyperglycosylated (as the S. cerevisiae was) and had sugar chains with a length similar to those produced in mammalian cells. A H. polymorpha-derived HCV-envelope protein vaccine were able to clear HCV infection in chimpanzees following challenge [220].
The P. pastoris system has been used to produce chikungunya VLPs (CHIK-VLPs), which were evaluated both in vivo and in vitro for efficacy as vaccine candidates. The VLPs elicited high titres of antibodies that recognized native CHIKV and neutralized different strains of CHIKV. P. pastoris is in multiple patent filings for expression of flavivirus VLPs proteins as vaccine candidates [221]. The RBD of SARS-CoV-2 has been produced in P. pastoris in several studies with appropriate N- and O-glycosylation [163,207,222,223,224,225]. The RBD of SARS coronavirus was expressed in P.pastoris X-33. When adjuvanted with aluminum hydroxide, it elicited high neutralizing antibody titres and high RBD-specific antibody titres [226]. RBD produced in the P. pastoris X-33 system and adjuvanted with 3M-052 alum was shown to induce robust humoural immunity, strong and durable RBD-specific CD8+ and TH1-biased CD4+ T cell responses and significantly reduced viral load in LRT and URT in rhesus macaques after SARS-CoV-2 challenge [223]. With the recombinant protein fused to Saccharomyces cerevisiae α-factor secretion signal, P. pastoris expression yielded 10–13 mg/L in cell culture vs. 5 mg/L in an HEK293 cell line [163]. Yeast-derived SARS-CoV-2 RBD is highly glycosylated and, compared to mammalian and insect cell-derived RBD [105,227], yeast-derived RBD has a very distinct glycosylation pattern. In P. pastoris-derived proteins, N-glycans are high-mannose type without core fucose [228] and O-glycans are linear chains of four to five α-linked mannose residues [229]. RBD protein contains complex N-glycans and mainly Core-1 O-glycans when produced in CHO cells [230]. From several studies among expression systems discussed so far, the difference in glycosylation between yeast, CHO and insect-produced RBDs seems to not influence their immunogenicity. PichiaPink™ Strain 1 (Invitrogen, Waltham, MA, USA) cells were also used to produce SARS-CoV-2 RBD as both monomers and dimers, which were shown to elicit broadly neutralizing antibodies and long-lasting protective immunity against SARS-CoV-2 in mice [207]. However, these authors deglycosylated their purified RBD proteins before use in their experiments. Deglycosylation was performed under reducing conditions where purified RBD was denatured then digested with peptide-N-glycosidase F or endo-N-acetylglucosaminidase.
Patent filings have been disclosed for S. cerevisiae expressed VP1-4 proteins of enterovirus 71 (EV71) VLPs and for Coxsackievirus A16 (CA16) VLPs, both of which cause HFMD [187]. The VLPs of both viruses showed structural similarities to VLPs produced in EV71-or CA16-infected mammalian cells and mice immunized with the respective VLPs elicited strong and specific neutralizing antibodies and cellular responses that protected neonatal mice against lethal EV71 [231] and CA16 [232] challenge. Yeasts have been used for the production of other viral subunit vaccine antigens as well such as hantavirus, HIV1, poliovirus, rabies and influenza, which have been reviewed previously [233].

6. Algal Expression Systems

Green algae are highly diverse and consist of unicellular and multicellular species. The best characterized and the alga of most interest in vaccine production is the unicellular Chlamydomonas reinhardtii. The nucleus, chloroplast and mitochondria of this algal species are easily transformed, and there is a short culture period between generating initial transformants and scale-up to production volumes. Gametogenesis can be induced that allows for genetic crossing; it can be grown either heterotrophically or phototropically, and a wide range of promoters are available and secreted, and glycosylated proteins can be produced and grown in cultures up to 500,000 L in a contained, cost-effective manner (Figure 6) [234,235]. DNA can be delivered via mechanical agitation, surfactant permeabilization, electroporation, particle bombardment or bacterial DNA transfer using conjugation or Agrobacterium-mediated [236,237]. Emerging techniques also exist, such as cell-penetrating peptides, polymers, metal–organic frameworks, nanoparticles and liposomes [238,239,240]. Algae exhibit three times greater photosynthetic efficiency than plants and contain large amounts of protein ranging from 50 to 70% of their fresh weight, which is a much higher percentage than the edible and harvestable parts of any higher plant or animal system [234]. Algae can be lyophilized, and studies have been performed on algal-produced vaccines where dried algae was stored at room temperature for six months [240] and up to twenty months [241]. Protein purified from these stored algae was able to demonstrate antigen effectiveness similar to fresh algae, showing that algae containing unpurified antigen can be stored and transported without the need for a cold chain.
The accumulation of biomass in algae is rapid and the entire biomass can be used for vaccine production, as opposed to plants that expend energy-producing tissues that are not used in the vaccine or cannot be easily harvested. Algae are also not restricted by seasons or soil fertility and media can be recycled to minimize water and nutrient loss. There is no concern regarding the cross-contamination of nearby food crops and enclosed bioreactors can be used to increase biomass yields and reduce concerns of environmental escape [240,242]. While it is evident that algae can produce complex vaccine antigens that are able to elicit immunogenic responses, identifying adjuvants to complement the antigens is critical. Viral antigens in particular require proper post-translational modification, such as glycosylation to be recognized properly by the immune system, but glycosylation does not occur in the chloroplast [243,244,245], so would not be considered suitable for viral vaccine antigen production.
Dunaliella salina is another species of algae that has been used to produce recombinant protein. It has a strong adaptability to the environment and can live in various salt concentrations from 0.2% to 35% salt saturation, which prevents contamination from other microorganisms [246]. Like C. reinhardtii, D. salina is photosynthetic and can be easily, rapidly and inexpensively cultured by large-scale factory farming. As a eukaryotic organism, it can produce and modify recombinant proteins at the levels of transcription and translation. D. salina lacks a rigid cell wall and can be easily transformed with exogenous genes [247].

Applications of Algal Expression System

In recent years, several other algal species have been investigated for their protein expression uses, such as other green algae, diatoms and cyanobacteria, in order to increase host range compatibility [243,244,245]. Over twenty species of algae have been transformed and several promoters and selectable markers have been characterized for many of these species [248,249,250,251].
The RBD of the SARS-CoV-2 spike protein has been produced in algal systems using Chlamydomonas reinhardtii [252,253], Chlorella vulgaris [254] and Phaeodactylum tricornutum [255]. In one study, Nicotiana benthamiana agrobacterium-mediated transformation vectors were used to transiently express SARS-CoV-2 RBD in both Chlamydomonas reinhardtii and Chlorella vulgaris [254] but the biological characterization of the recombinant RBD to the ACE2 receptor as well as the long term expression levels beyond 72 h post-transformation was not investigated. Another study used nuclear transformation in C. reinhardtii [252], where stable nuclear genome integration vectors were able to generate recombinant RBD expression levels that were an order of magnitude higher than when using agrobacterium-mediated transformation [254] and maintained this expression for at least one year post transformation. This approach consisted of a nuclear-stable transformation, where a linear vector is randomly integrated in the genome to code a fusion protein RBD::mClover (RBD + a fluorescent protein for monitoring expression). Constructs aimed at inducing expression in the chloroplast and ER, and it was shown that the ER approach was deemed better allowing for yields up to 31 μg/g of fresh algae biomass. The purified RBD from ER-Golgi was also able to bind ACE2 at a similar affinity as mammalian-expressed RBD. It was discussed that higher expression levels using nuclear transformation stems from the use of endogenous promoters, UTRs and a codon-optimized RBD sequence. Using stronger endogenous or synthetic promoters in new transgene designs and the addition of more introns into the RBD coding sequence could be used to boost expression further. However, it is known that several chloroplast genome engineering approaches undertaken in algae that allow for the highest transgene expression also negatively affect the photosynthetic ability of the organism, which removes the option of low-cost photosynthetic cultivation [256]. Another method used is transient expression [253], which implies a short production time and has no need for clone selection and characterization or fusing the antigen to a protein partner, but still yields up to 4 µg/g. Where RBD has also been transiently expressed in N. benthamiana [257], yields of up to 8 μg of pure antigen/g leaf mass was recorded. The diatom Phaeodactylum tricornutum has an N-linked glycosylation pathway that is like that found in mammals and may be a reason that the RBD made in P. tricornutum is bioequivalent to the RBD made in mammalian cells. Expressed RBD in this system was similar to that in Chlamydomonas [252]. The RBD purified from whole cell extracts was essentially homogenous and possessed N-linked glycosylation, which suggests that a portion of the expressed RBD is transited through the ER/Golgi apparatus and may be secreted at too low levels to be detected or is not secreted at all. It is possible that other secretory peptides are better optimized for RBD expression and secretion. In purification attempts from P. tricornutum, 90–95% of expressed RBD did not bind the Ni-affinity column in their first chromatography step and the majority of RBD was lacking all or some of the C-terminal 6x-His tag, which is possibly because this alga encodes a protease which cleaves the C-terminal 6x-His tag or was due to premature transcription termination, post-translational processing or premature translation termination. Different tags may be better suited for the expression and purification of protein by this algal species.

7. Plant Expression Systems

Plant-based expression systems are cost-effective, have high scalability and greater safety as plants do not harbour mammalian pathogens (Figure 7). Plants are also able to provide post-translational modifications and appropriate folding resulting in functional, bioactive proteins [258]. Protein production in plants can used either stable or transient expression of transgenes. For stable expression, transgenes can be in the nuclear genome of transgenic plants or plant cell lines or in the chloroplast genome of transplastomic plants. For the transient expression of transgenes in plants an agrobacterial or agroviral vector can be used [258]. Virtually any genetically transformable plant can be used to produce recombinant proteins. Alfalfa, cereal crops like corn, rice and barley, as well as tobacco, are the most widely used as biofactories for biopharmaceuticals [259].
A plant-expressed vaccine should be stable and allow for high expression levels and low production and storage costs. Maize grain has evolved to maintain its proteins in a stable environment to allow germination after years of dormancy, which is in contrast to vegetative matter and fruits which degrade soon after harvesting [260]. This long-term stability has been shown in recombinant proteins where their activity is retained for years stored in the maize grain, which in fact allows for long-term storage and transportation at ambient temperatures and the grain can be processed at will, as opposed to needing to process large batches immediately upon harvest [261]. The removal of oils by supercritical fluid extraction (SFE) can also increase the stability of the grains and the immune response may be enhanced upon SFE treatment [262]. For crop plants, there is a well-established infrastructure for their cultivation, harvest and storage. The protein in cereal grains and oilseeds is stable as these seeds can be stored for long periods of time. Legumes such as alfalfa and soybean can fix nitrogen from the atmosphere and therefore have fewer chemical requirements for growth.
Plant production strategies involve either stable nuclear expression, stable chloroplast expression, transient expression or suspension cultures [263]. Nuclear expression allows for the stable integration of transgenes into the genome of plant cells, which is regulated by the transcription of the gene of interest and then translation in the cytoplasm. Agrobacterium tumefaciens-mediated transformation is the most widely used and offers the simplest method for the genetic modification of crops with horizontal transgene transfer and consistent recombinant protein expression. This method, however, can sometimes result in gene silencing, transgene contamination risk, potential interactions with natural products and low yields (<1% of total soluble protein) and can be time consuming [264]. The two methods used involve plant pathogens as vectors, plant viral infection or agroinfiltration by A. tumefaciens. High expression efficiency has been observed, but the risk of viral vector contamination and the environment must be carefully considered [265]. Plant cell cultures grown in suspension show more promise than using whole plants and biosafety and environmental concerns can be overcome by using bioreactors to prevent cross-fertilization and the transmission of pollen. Production costs are lower than mammalian systems as they require simple media [266].
The past twenty years has allowed plant expression systems to address technical issues such as expression, yield, purity and reproducibility as well as regulatory expectations like cGMP manufacture and the demonstration of efficacy in pre-clinical and clinical trials. This has allowed plant expression systems to become a commercially appealing approach to develop viral vaccines. In addition to this, there has been a recent shift in the expectation of plants to produce orally administered vaccines to established platforms to produce correctly folded, bioactive proteins, which can be used as injectable vaccines [259]. The major concerns associated with the plant expressed viral/vaccine antigens are oral tolerance, allergenicity, gene transfer to conventional food supplies and potential detrimental environmental impact.
The first USDA-approved plant expressed vaccine was against Newcastle disease in poultry produced by Dow Agro Sciences and was approved in 2006. The vaccine comprises the recombinant haemagglutinin–neuraminidase antigen expressed in transgenic tobacco suspension cells [267,268]. The CSF vaccine, HERBAVAC®, was licenced in South Korea. It is a fusion protein of the E2 antigen and the cellulose-binding domain (CBD) and does not contain the Erns antigen present in field strains or live-attenuated vaccine, making it possible for pigs vaccinated with HERBAVAC® to be distinguishable from pigs infected with field strains or vaccinated with the live-attenuated vaccines [269].

Applications of Plant Expression System

Several plant-expressed viral antigens are in development (Table 7).
H1 and H5 VLP vaccines produced in Nicotiana benthamiana elicited both strong antibody responses and poly-functional, cross-reactive memory T cells persisting for at least 6 months post vaccination [270]. Plant-derived VLP vaccines addressed several limitations of currently licenced vaccines. The proteins included in their VLPs are based on the genetic sequence of currently circulating human influenza viruses as opposed to egg-adapted influenza strains for optimal growth. With alum adjuvant, a single intramuscular (IM) injection of 3 μg of a plant-made H7 VLP vaccine induced a robust HI antibody response in both mice and ferrets [271]. The authors were also able to demonstrate clear induction of a strong, antigen-specific, T-cell response after immunization in ferrets. The humoral and cell-mediated immune responses provided 100% protection after lethal challenge with H7N9 in mice and a reduction in clinical signs and virus load ferret nasal and lung washes 3 dpi. Two doses of the unadjuvanted vaccine provided improved protection in mice [271]. This plant-expressed VLP vaccine contains low biological reactivity lipopolysaccharide (LPS) from Agrobacterium, which has been demonstrated to elicit immunological activity and may act as an adjuvant itself [271]. It is possible that this LPS may be partially responsible for the better protection that was observed after two immunizations with the unadjuvanted VLP. An H6 VLP produced in Nicotiana benthamiana was used in a challenge study with a heterologous H6N2 influenza virus in chickens [273]. A single dose elicited an immune response comparable to a commercial H6N2 (inactivated) vaccine administered with two doses. The H6 VLP vaccine also significantly reduced the proportion of chicken shedding virus as well as the level of shedding by >100-fold in oral and >6-fold in cloacal swabs.
VLPs displaying an S protein of the infectious bronchitis virus was produced using Agrobacterium-mediated Nicotiana benthamiana transient expression system [274]. When adjuvanted with Emulsigen®-P, specific pathogen-free chickens immunized intramuscularly seroconverted after two weeks with mean HA-inhibition titres of 9.1 to 10 log2 depending on the dose. Studies showing virus neutralization potential and/or protection have not been undertaken.
N. benthamiana was used to produce HAC1 and HAI-05, recombinant HA antigens of influenza A virus. Both subunits induced significant haemagglutinin inhibition and virus neutralizing serum antibody titres in mice, rabbits and ferrets. The use of AlhydrogelTM adjuvant also provided a dose-sparing effect [272].
A study used pmE2:pFc2 expressed using transgenic plants instead of a transient expression system to produce a CSFV E2 vaccine antigen [276]. While transient expression is generally expected to produce higher amount of target protein than stably expressing transgenic plants [277], the authors used transgenic plants instead as in transient expression systems, the expression level of the target genes is not uniform. Transgenic plants provide the stable expression of recombinant genes once homozygous transgenic lines are generated, which leads to easier large-scale production than in the transient system. Economically, upstream processes in transient expression require additional infrastructure, which can be eliminated in the transgenic approach allowing for inexpensive recombinant vaccine production [278], without lipopolysaccharide contamination [279].
Plant-based manufacturing systems have advantages with scalability compared to tissue-culture and egg-based production platforms and other recombinant technologies with capacity limitations of large-scale bioreactors. In 2012, Medicago demonstrated production of 10 million doses of plant-based H1N1 influenza vaccine within a month, demonstrating the capacity of this system [280,281].
The plant-based production of SARS-CoV-2 antigens has been well studied over the past 3 years, where some of the main drawbacks have been poor reproducibility, antigen stability and bioavailability [275,282,283,284,285,286,287]. A plant expression vector containing SARS-CoV-2 RBD was transformed into Agrobacterium tumefaciens strain GV3101 cells by electroporation then injected into 6-week-old N. benthamiana leaves [275]. The RBD was then used to immunize mice and NHPs with and without alum and was shown to induce neutralizing antibodies in mice and NHPs. Challenge studies were not performed; however, it was observed that a mixed Th1/Th2 response was induced, and even without alum, a SARS-CoV-2 RBD-specific T cell response was also generated [275].

8. Cell-Free Expression Systems

Cell-free expression systems (Figure 8) (CFSs) replicate the biology of transcription and translation in the absence of a cell. Protein synthesis is initiated in vitro using cell extracts prepared from cells by lysis and several washing steps to remove cell debris and cellular mRNA/DNA [288,289]. The extracts contain the components required for transcription and translation such as aminoacyl-tRNA synthetases, ribosomes, factors necessary for elongation, initiation and transcription. These are combined with amino acids, energy substrates, template DNA, cofactors, salts and nucleotides, and this is varies depending on the biochemical properties of the antigen. Translation is then initiated by adding the linear or circular template DNA or mRNA and the process is allowed to occur at a specified temperature and time period [290].
This work was pioneered in the 1960s [291,292] with major achievements made well into the 2000s, where fundamental ‘operating systems’ were established for proof-of-concept research [293]. The terms ‘cell-free biology’ and ‘cell-free expression systems’ have since evolved to mean much more than described above and now involves the addition of repressor molecules, chaperone proteins, non-native co-factors and substrates to improve the performance and productivity of expression reactions [294,295]. A plasmid containing the gene of interest is directly added to the cell-free expression system to initiate protein production [296]. CFSs can be stored as freeze-dried products allowing for simplified storage and applications [297,298].
Different CFS reaction formats have since been developed, from easily handled and scalable batch reactions with short reaction times but relatively low protein yields to complex dialysis systems known as continuous-flow cell-free and continuous exchange cell-free systems [299,300]. Due to the ease of manipulation of reagents and the scalability of the CFS, automated high-throughput systems are in development [301].
By removing the need to sustain life, CFSs allow control over the molecular machinery for gene expression, enabling the researcher to manipulate the system by adding non-native substrates such as recombinant DNA templates [302]. Cell-free experiments can also circumvent mechanisms involved in species survival and bypass limitations on molecular transport, providing the ability to focus the cellular machinery on the biosynthesis of a specific product. The details of the progress made regarding the preparation of cell-free extracts and reaction mixtures, optimization and more background on cell-free protein expression have been discussed previously [296,297,298,299,300,301]. The most common cell lysates used (Table 8) are acquired from E. coli [302,303,304,305,306,307], but other bacterial sources [308,309,310,311,312] or eukaryotic systems such as mammalian [313,314,315,316], insect [299,317,318] or plant cells [319,320,321] have also been adapted, although these are more expensive [322,323].
Prokaryotic CFSs such as Bacillus subtilis [310], Pseudomonas putida [312], Streptomyces [324] and Vibrio natriegens [308] have been recently optimized, and eukaryotic CFS based on Tobacco [325], Leishmania [326], Neurospora crassa [327], Saccharomyces cerevisiae [328] and erythrocytes [323] have been characterized and optimized for certain proteins at the laboratory level.
Most therapeutic proteins like monoclonal antibodies and viral antigens have glycan moieties. Synthesizing these glycoproteins in vitro is challenging since the most common cellular lysate source for CFSs, E. coli, lacks native glycosylation machinery. Several solutions have been suggested such as using a eukaryotic CFS, which can be supplemented with purified microsomes with the necessary components for glycosylation, but these seem to produce much less functional folded protein than bacterial systems [322]. To address these issues, a group developed a novel cell-free glycoprotein synthesis (CFGpS) system which integrates protein biosynthesis with asparagine-linked protein glycosylation. This involved a glycol-optimized E. coli strain as the source of cell extracts selectively enriched with glycosylation components such as oligosaccharyltransferases and lipid-linked oligosaccharides. The authors indicated that it enabled a ‘one-pot’ reaction scheme for the efficient and site-specific glycosylation of target proteins [329]. A major advantage of this system is that it allows a high level of control over all glycosylation components (catalysts, substrates and cofactors) in terms of important process variables such as input concentrations, time of addition and reaction time. Genome engineering makes the elimination of inhibitory enzymes, such as glycosidases, possible, which would hydrolyze necessary glycosidic linkages. The level of control with CFSs provides the opportunity to overcome bottlenecks that would otherwise limit glycosylation efficiency [330].
Various methods have been established to enhance the correct folding and solubility of transmembrane proteins such as supplementation with membrane-mimicking structures, such as micelle-forming detergents, nanodiscs, liposomes or exogenous microsomes [324,325,326]. Eukaryotic CFSs contain endogenous microsomes from the cellular endoplasmic reticulum (ER), which allows for the co-translational translocation of proteins and ER-associated post-translational modifications [331,332,333,334,335].
Table 8. Comparisons of commonly used CFSs.
Table 8. Comparisons of commonly used CFSs.
SystemAdvantagesDisadvantagesReference
E. coliHigh protein yield. Simple cultivation, rapid cell growth and easy lysate preparation. Cost-efficient. Well-established genetic engineering methods. High levels of VLP production.Post-translational modification issues. Lack of endogenous membrane structures for integral membrane protein synthesis. Only prokaryotic chaperones. Eukaryotic proteins often incorrectly folded.[306,307,329,330,331,332,336,337,338,339]
YeastPost-translational modifications possible. Rapid, easy propagation of cells and lysate preparation. Well-established genetic engineering methods.Low protein yield. No mammalian-like post-translational modifications. Slightly more costly than E. coli. Moderate levels of VLP production.[328,333,335,340,341,342]
Wheat germHigh yield of complex proteins. Synthesis of disulphide bridged proteins. Correct protein folding and high solubility. Well-established genetic engineering methods.Labour-intensive and expensive lysate preparation. Limited post-translational modifications. No endogenous membrane structures. Low protein yield compared to prokaryotic and cell-based wheat germ systems.[319,334,343]
TobaccoHigh yield of complex proteins. Rapid, simple lysate preparation. Glycosylation and disulphide bridge formation possible.Few studies on tobacco CFSs[320,325,344]
Insect cellRapid, simple lysate preparation. Post-translational modifications possible. Endogenous microsomes available. Direct synthesis and integration of membrane proteins.High cost of cell propagation. Moderate levels of VLP production.[299,345,346]
CHO cellContains endogenous microsomes. Mammalian PTMs. Direct production of membrane proteins. Well-established cell lines. IRES-mediated translation initiation allows high protein yield.Low yield compared to prokaryotic CFSs. High cost of cell propagation. Low levels of VLP production.[347,348]
Human cellOptimal environment for native protein folding and assembly of viral membrane proteins. Contain endogenous microsomes. Human post-translational modifications. Codon manipulation allows the synthesis of high molecular weight proteins.Low protein yield compared to prokaryotic CFS. High cell propagation costs. Labour intensive cell culture technologies needed as human cells are sensitive.[323,349,350]

Applications of Cell-Free Expression System

The E. coli-based CFS has also been used to rapidly produce domains from pH1N1 influenza virus HA. The stabilized HA head domain trimer was effectively recognized by antibodies from pH1N1 influenza vaccine recipients and bound to sialic acids, strongly indicating that the trimers were correctly formed and could be potentially effective as vaccines [351,352]. The E. coli CFS has been used to synthesize and assemble human norovirus (HuNoV) VLPs in 4 h with similar protein yield to other cell-based systems [353]. The extremely low level of endotoxin from the cell-free system could meet the demands for vaccine applications, referencing guidelines for toxoid-based vaccines [298,354]. Strains of E. coli such as ClearColiTM [37] or the use of an endotoxin binding column are also useful in reducing endotoxin levels.
A wheat germ CFS was used to produce highly soluble MERS-CoV nucleoprotein (NP) antigen, which was able to generate mAbs following the immunization of mice [355]. Hepatitis B core antigen VLPs produced in a Pichia pastoris cell-free expression system were found to have comparable characteristics to those previously produced in vivo and in vitro [356].
One study has produced the SARS-CoV-2 disulphide-bonded RBD protein in a wheat germ cell-free production system [357]. A wheat germ (WG) expression kit was used in a bilayer translation method [358], where by adding protein disulfide isomerase (PDI) and endoplasmic reticulum oxidase (ERO1α) to the translational reaction mixture, a functionally intact RBD protein that can interact with ACE2 was synthesized. The common problem with conventional WG expression is the use of dithiothreitol (DTT) in the wheat germ extract and substrate solution to enhance translation efficiency; however, DTT is known to disrupt disulphide bonds in the synthesized protein, which is detrimental to proteins where the disulphide linkage is important to its function. The modification of the WG (EP-WG) with the inclusion of the endoplasmic reticulum oxidoreductase-1 α (ERO1α) and protein disulfide isomerase (PDI) overcomes this drawback to produce biologically similar proteins with intact disulphide bonds. The RBD produced in this system had a different band shape compared to RBD produced in mammalian systems, which is possibly due to glycosylation or other post-translational modifications that occur in the mammalian system. The lack of glycosylation is one of the drawbacks of the WG system. RBD has two glycosylation sites, N331 and N343. In this study, RBD synthesized by EP-WG had slightly lowered affinity for ACE2 than RBD produced in the mammalian cell expression system. Vaccine-derived antibodies produced by conventional WG expression did not react with RBD whose conformation had been disrupted with DTT treatment, demonstrating that the disulfide bonds in the RBD are essential for its antigenicity.
The SARS-CoV-2 RBD has also been produced in a cell-free system derived from Nicotiana tabacum BY-2 cell culture (BY-2 lysate; BYL) within 48 h [344]. This work has improved on previous studies [320,325] and has now been optimized and commercialized by LenioBio GmbH as Almost Living Cell-free Extract (ALiCE)®. They were able to demonstrate that this system can easily and linearly scale across a 1000-fold difference from 100 μL reactions to 10 and 100 mL. The study also showed preliminary data from 1 L reactions in a commercial bioreactor, the first example of litre scale reactions from a eukaryotic cell-free protein synthesis system. In addition to SARS-CoV-2 RBD, they also produced a hepatitis B core VLP, the monoclonal antibody adalimumab, human epidermal growth factor and a G protein-coupled receptor membrane protein. These expressed proteins are monomeric and multimeric in nature with disulphide bonds, N-glycosylation and transmembrane domains, which demonstrates the versatility of BYL. Using anti-RBD antibodies from R&D systems, serum samples from COVID-19 patients and pure protein samples produced by the BYL-ALiCE cell-free system, RBD-binding and serological assays were performed by ELISA. RBD produced in BYL was directly comparable to RBD produced in HEK-293 cells. The work shows the potential of BYL as an end-to-end R&D protein synthesis platform, which could significantly reduce the time-to-market for vaccine antigens.

9. Future Directions

Taking into consideration the time and various complexities involved in traditional cell-based expression, cell-free systems are an emerging vaccine production technology compared in Table 9. Vaccine antigens can be processed for downstream applications like purification and functional analysis without the transfection, selection and expansion of clones. The availability of cell-free lysates from commercial sources has increased; however, there are major challenges with respect to scalability, cost effectiveness, protein folding and functionality.
The BEVS is one of the most efficient cell-based protein production platforms in use as most of the proteins synthesized in this system are functionally, antigenically and immunogenically similar to the native protein. This is because the insect cell system is able to perform several post-translational modifications. Based on the evidence, Spodoptera frugiperda CFSs have been developed [366,367]. The ER is not fully removed during lysate preparation but allows the structures to rearrange themselves into endogenous microsomes, which are translocationally active and provide a somewhat natural lipid membrane for protein translocation and embedding. Performing repeated protein synthesis allows the same batch of microsomes to be used multiple times and enrichment of the target proteins in the lumen and membrane of microsomes was achieved [368]. In this insect CFS, post-translational modifications can be achieved without additional enzymes and cofactors as the glycosylation, phosphorylation and disulphide bond formation have been shown [369].
Studies involving CFSs based on CHO cell lysates have shown that the combination of well-known properties and safety aspects of CHO cells [370,371] with the versatility of cell-free technology [314,347,372] provides many advantages for recombinant protein production. The ability to synthesize proteins easily and quickly and the flexibility of the DNA template allows for potential screening technologies. Vaccine antigens are usually membrane proteins originating from humans, and mammalian expression systems fulfil all the requirements such as post-translational modifications, cofactors and chaperones for correct folding and efficient production, although the overexpression of membrane proteins can be toxic for cell cultures and result in cell death or truncated or misfolded proteins [373].
Currently, CHO-lysate CFS reactions are performed coupled with transcription and translation occurring in one reaction, which allows for the fast and convenient production of proteins for high-throughput screening [372]. A major limitation of most eukaryotic CFS is low protein yield, due to limiting translational initiation, but integrating internal ribosome entry sites (IRESs) into the DNA template improved protein production by bypassing several yield-limiting steps in eukaryotic translation initiation [347].
Research into standardization is important as the CFS evolves into a technique used by large laboratories for protein expression and several teams have already begun this work [368,369,370,371,374,375,376,377]. This allows researchers to obtain more statistically significant results and those implementing CFS for the first time can easily and successfully contribute to the establishment of CFS as a major protein expression technology.
Transgenic animals, and more so, transgenic milk, are another recent protein expression platform. Mammary gland-specific promoters have been modified to express a wide range of biopharmaceutical proteins in rodents, rabbits and livestock [378,379,380,381]. The mammary gland is the organ of choice as milk is easily collected in large volumes. Protein expression is commonly reported to be produced at rates of several grams per litre [379]. Blood, egg white, seminal plasma and urine are other theoretically possible systems and have been discussed previously [3]. Interest in transgenic eggs has also been shown as a single hen can lay up to 330 eggs per year, and egg white naturally contains approximately 4 g of protein [382]. Urine has also been explored as an abundant biological fluid for protein expression. An advantage of using the bladder is that transgenic animals can urinate earlier than they can lactate; however, the major limitation of this system is yield, as although the bladder epithelium does secrete proteins, production rates are extremely low [383]. Seminal fluid is another relatively abundant biological fluid in some species such as pigs and it can easily be collected. Pig semen contains approximately 30 mg of protein/mL and boars can produce 200–300 mL of semen for a total of 6–9 g of protein per ejaculate [384]. Protein secretion in the male boar’s accessory sex glands is uniquely exocrine, minimizing the risk of a biologically active recombinant protein and upsetting the host’s own physiology; however, it is not well-understood how complex proteins are matured and secreted in semen [385].
In terms of the transgenic animal expression of vaccine antigens, very few studies exist. Enterovirus 71 (EV71) VP1 capsid protein has been expressed and secreted into the milk of transgenic mice with a yield of 2.51 mg/mL. Mouse pups that received VP1-transgenic milk orally showed better health conditions after challenge with EV71 compared to a non-transgenic milk-fed group, and bodyweight of the VP-1-fed group was similar to the uninfected group. Serum neutralization assays and serum antibody detection showed the VP-1-fed mice generated antibodies specific to EV71 [386]. Transgenic rabbits have been used to produce rotavirus VP2 and VP6 proteins in their milk from 50 to 250 µg/mL. These proteins were able to elicit significant protection against rotavirus infection in rabbit babies when orally administered in the whole milk. Protection levels were similar to other studies where VLP2/6 were used as immunogens in oral vaccination protocols.
Recombinant protein production in the milk of livestock seems to be the optimal direction for this type of expression system, which may lead to the creation of animal lines where genomes are modified by the insertion of sequences for the directed recombination of the expression cassette into a milk protein-encoding gene. The insertion can be achieved with high efficiency Via microinjection into the oocytes of a genetic construct carrying an expression cassette as well as the corresponding recombinant integrase or its mRNA. These animal lines can be created using the CRISPR/Cas9 system or its analogues which are highly efficient and simple to design and implement. This is one of the most promising approaches to the generation of transgenic animals producing recombinant antigens in milk as the CRISPR/Cas9 targeted integration of the transgene into the genes that encode milk proteins allows for transgene expression to be controlled by the endogenous regulatory sequences of the recipient gene. Applications of this technology will simplify and standardize transgenic animal production for the generation of recombinant antigens. Synthetic biology allows the use of mini-genes with artificial introns as transgenes to facilitate the efficient expression of the transgene and the production of the target protein, which simplifies the design and creation of genetic constructs for transgenesis [387,388].
The purification of recombinant vaccine antigens from milk creates undefined regulatory hurdles; however, using the modern technology available makes compliance with regulatory requirements describing the transformation event simple. Major hurdles are the requirements regarding the biological safety of recombinant protein in milk, which will require revising the standards for farm animal welfare and veterinary oversight to exclude the presence of extraneous agents as well as controlled parameters of manufactured proteins for use as subunit vaccines.

10. Conclusions

Both procaryotic and eucaryotic expression systems have been demonstrated to be useful for production of viral vaccine antigens. However, the number of successfully demonstrated vaccines is much greater in the eucaryotic expressions systems compared to the procaryotic systems. Further advances are underway in improving both procaryotic and eucaryotic protein expression systems to produce proteins with glycosylation patterns like mammalian cells and to be more economical for production. In addition, new cell free systems offer new approaches for protein expression.

Author Contributions

Conceptualization, J.R.V.S. and S.B., writing, J.R.V.S., Z.S., A.A., D.K., K.P. and S.B., images, Z.S. review, editing and draft preparation, J.R.V.S., Z.S., A.A., D.K., K.P. and S.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The data and information presented in the review are from papers published (and cited) in the References section, with abstracts accessible in PubMed.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Chadd, H.E.; Chamow, S.M. Therapeutic antibody expression technology. Curr. Opin. Biotechnol. 2001, 12, 188–194. [Google Scholar] [CrossRef] [PubMed]
  2. Andersen, D.C.; Krummen, L. Recombinant protein expression for therapeutic applications. Curr. Opin. Biotechnol. 2002, 13, 117–123. [Google Scholar] [CrossRef] [PubMed]
  3. Van Damme, P. Hepatitis B Vaccines BT—Pediatric Vaccines and Vaccinations: A European Textbook; Vesikari, T., Van Damme, P., Eds.; Springer International Publishing: Cham, Switzerland, 2017; pp. 109–116. [Google Scholar]
  4. McNeil, C. Who invented the VLP cervical cancer vaccines? J. Natl. Cancer Inst. 2006, 98, 433. [Google Scholar] [CrossRef] [PubMed]
  5. Roldão, A.; Mellado, M.C.M.; Castilho, L.R.; Carrondo, M.J.T.; Alves, P.M. Virus-like particles in vaccine development. Expert Rev. Vaccines 2010, 9, 1149–1176. [Google Scholar] [CrossRef] [PubMed]
  6. Crisci, E.; Bárcena, J.; Montoya, M. Virus-like particles: The new frontier of vaccines for animal viral infections. Vet. Immunol. Immunopathol. 2012, 148, 211–225. [Google Scholar] [CrossRef] [PubMed]
  7. Teffera, M.; Babiuk, S. Potential of Using Capripoxvirus Vectored Vaccines Against Arboviruses in Sheep, Goats, and Cattle. Front. Vet. Sci. 2019, 6, 450. Available online: https://www.frontiersin.org/article/10.3389/fvets.2019.00450 (accessed on 1 June 2024). [CrossRef]
  8. Singh, A. Modern Vaccines with DIVA Capability: An Overview. Res. Rev. A J. Immunol. 2019, 5, 195–205. [Google Scholar]
  9. Chu, L.; Robinson, D.K. Industrial choices for protein production by large-scale cell culture. Curr. Opin. Biotechnol. 2001, 12, 180–187. [Google Scholar] [CrossRef] [PubMed]
  10. Mascola, J.R.; Fauci, A.S. Novel vaccine technologies for the 21st century. Nat. Rev. Immunol. 2020, 20, 87–88. [Google Scholar] [CrossRef]
  11. Chan, C.Y.-Y.; Tambyah, P.A. Preflucel®: A Vero-cell culture-derived trivalent influenza vaccine. Expert Rev. Vaccines 2012, 11, 759–773. [Google Scholar] [CrossRef] [PubMed]
  12. Manini, I.; Domnich, A.; Amicizia, D.; Rossi, S. Flucelvax (Optaflu) for seasonal influenza. Expert Rev. Vaccines 2015, 14, 789–804. [Google Scholar] [CrossRef] [PubMed]
  13. Thomas, S.J.; Yoon, I.-K. A review of Dengvaxia®: Development to deployment. Hum. Vaccines Immunother. 2019, 15, 2295–2314. [Google Scholar] [CrossRef] [PubMed]
  14. Andréll, J.; Tate, C.G. Overexpression of membrane proteins in mammalian cells for structural studies. Mol. Membr. Biol. 2013, 30, 52–63. [Google Scholar] [CrossRef] [PubMed]
  15. Gupta, S.K.; Shukla, P. Glycosylation control technologies for recombinant therapeutic proteins. Appl. Microbiol. Biotechnol. 2018, 102, 10457–10468. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, D.; Liang, L.; Wang, S.; Nakao, T.; Li, Y.; Liu, L.; Guan, Y.; Fukuyama, S.; Bu, Z.; Kawaoka, Y.; et al. Glycosylation of the Hemagglutinin Protein of H5N1 Influenza Virus Increases Its Virulence in Mice by Exacerbating the Host Immune Response. J. Virol. 2017, 91, e02215-16. [Google Scholar] [CrossRef] [PubMed]
  17. Dalton, A.C.; Barton, W.A. Over-expression of secreted proteins from mammalian cell lines. Protein Sci. 2014, 23, 517–525. [Google Scholar] [CrossRef]
  18. Varki, A.; Cummings, R.D.; Esko, J.D.; Stanley, P.; Hart, G.W.; Aebi, M.; Darvill, A.G.; Kinoshita, T.; Packer, N.H.; Prestegard, J.H. Essentials of Glycobiology [Internet]. 2015. Available online: https://pubmed.ncbi.nlm.nih.gov/27010055/ (accessed on 1 June 2024).
  19. Qian, H.; Wu, Y.; Bian, G.; Zhang, Q.; Xu, Y.; Bai, Z.; Wu, S.; Che, Y.; Lv, Y.; Zha, Y. Porcine Epidemic Diarrhea Virus S Protein and Subunit Vaccine Thereof as Well as Method for Preparing Subunit Vaccine and Application Thereof. U.S. Patent App. 16/600,334, 18 June 2020. [Google Scholar]
  20. Fattom, A.I.; Hamouda, T.; Bitko, V.; Baker, J.R., Jr. Nanoemulsion Respiratory Syncytial Virus (RSV) Subunit Vaccine. U.S. Patent 10,596,251, 24 March 2020. [Google Scholar]
  21. Eldemery, F.; Joiner, K.S.; Toro, H.; Van Santen, V.L. Protection against infectious bronchitis virus by spike ectodomain subunit vaccine. Vaccine 2017, 35, 5864–5871. [Google Scholar] [CrossRef]
  22. Li, D.; von Schaewen, M.; Wang, X.; Tao, W.; Zhang, Y.; Li, L.; Heller, B.; Hrebikova, G.; Deng, Q.; Ploss, A. Altered glycosylation patterns increase immunogenicity of a subunit hepatitis C virus vaccine, inducing neutralizing antibodies which confer protection in mice. J. Virol. 2016, 90, 10486–10498. [Google Scholar] [CrossRef] [PubMed]
  23. Lehrer, A.T.; Wong, T.-A.S.; Lieberman, M.M.; Johns, L.; Medina, L.; Feldmann, F.; Feldmann, H.; Marzi, A. Recombinant subunit vaccines protect guinea pigs from lethal Ebola virus challenge. Vaccine 2019, 37, 6942–6950. [Google Scholar] [CrossRef] [PubMed]
  24. Morcol, T.; Nagappan, P.; Bell, S.J.D.; Cawthon, A.G. Influenza A (H5N1) virus subunit vaccine administered with CaPNP adjuvant induce high virus neutralization antibody titers in mice. AAPS PharmSciTech 2019, 20, 315. [Google Scholar] [CrossRef]
  25. Medina, L.O.; To, A.; Lieberman, M.M.; Wong, T.A.S.; Namekar, M.; Nakano, E.; Andersen, H.; Yalley-Ogunro, J.; Greenhouse, J.; Higgs, S. A recombinant subunit based zika virus vaccine is efficacious in non-human primates. Front. Immunol. 2018, 9, 2464. [Google Scholar] [CrossRef] [PubMed]
  26. Durbin, A.P.; Pierce, K.K.; Kirkpatrick, B.D.; Grier, P.; Sabundayo, B.P.; He, H.; Sausser, M.; Russell, A.F.; Martin, J.; Hyatt, D. Immunogenicity and Safety of a Tetravalent Recombinant Subunit Dengue Vaccine in Adults Previously Vaccinated with a Live Attenuated Tetravalent Dengue Vaccine: Results of a Phase-I Randomized Clinical Trial. Am. J. Trop. Med. Hyg. 2020, 103, 855–863. [Google Scholar] [CrossRef] [PubMed]
  27. Holmgren, J.; Bourgeois, L.; Carlin, N.; Clements, J.; Gustafsson, B.; Lundgren, A.; Nygren, E.; Tobias, J.; Walker, R.; Svennerholm, A.-M. Development and preclinical evaluation of safety and immunogenicity of an oral ETEC vaccine containing inactivated E. coli bacteria overexpressing colonization factors CFA/I, CS3, CS5 and CS6 combined with a hybrid LT/CT B subunit antigen, administered al. Vaccine 2013, 31, 2457–2464. [Google Scholar] [CrossRef]
  28. Huang, X.; Wang, X.; Zhang, J.; Xia, N.; Zhao, Q. Escherichia coli-derived virus-like particles in vaccine development. npj Vaccines 2017, 2, 3. [Google Scholar] [CrossRef]
  29. Zhang, X.; Xin, L.; Li, S.; Fang, M.; Zhang, J.; Xia, N.; Zhao, Q. Lessons learned from successful human vaccines: Delineating key epitopes by dissecting the capsid proteins. Hum. Vaccines Immunother. 2015, 11, 1277–1292. [Google Scholar] [CrossRef] [PubMed]
  30. Li, S.-W.; Zhao, Q.; Wu, T.; Chen, S.; Zhang, J.; Xia, N.-S. The development of a recombinant hepatitis E vaccine HEV 239. Hum. Vaccines Immunother. 2015, 11, 908–914. [Google Scholar] [CrossRef] [PubMed]
  31. Babiuk, S.; Asper, D.J.; Rogan, D.; Mutwiri, G.K.; Potter, A.A. Subcutaneous and intranasal immunization with type III secreted proteins can prevent colonization and shedding of Escherichia coli O157:H7 in mice. Microb. Pathog. 2008, 45, 7–11. [Google Scholar] [CrossRef]
  32. Lobstein, J.; Emrich, C.A.; Jeans, C.; Faulkner, M.; Riggs, P.; Berkmen, M. SHuffle, a novel Escherichia coli protein expression strain capable of correctly folding disulfide bonded proteins in its cytoplasm. Microb. Cell Fact. 2012, 11, 56. [Google Scholar] [CrossRef] [PubMed]
  33. Dopp, J.L.; Reuel, N.F. Simple, functional, inexpensive cell extract for in vitro prototyping of proteins with disulfide bonds. Biochem. Eng. J. 2020, 164, 107790. [Google Scholar] [CrossRef]
  34. Rosano, G.L.; Ceccarelli, E.A. Recombinant protein expression in Escherichia coli: Advances and challenges. Front. Microbiol. 2014, 5, 172. Available online: https://www.frontiersin.org/article/10.3389/fmicb.2014.00172 (accessed on 1 June 2024). [CrossRef]
  35. Zeltins, A. Construction and characterization of virus-like particles: A review. Mol. Biotechnol. 2013, 53, 92–107. [Google Scholar] [CrossRef] [PubMed]
  36. Mamat, U.; Wilke, K.; Bramhill, D.; Schromm, A.B.; Lindner, B.; Kohl, T.A.; Corchero, J.L.; Villaverde, A.; Schaffer, L.; Head, S.R.; et al. Detoxifying Escherichia coli for endotoxin-free production of recombinant proteins. Microb. Cell Fact. 2015, 14, 57. [Google Scholar] [CrossRef] [PubMed]
  37. Mamat, U.; Woodard, R.W.; Wilke, K.; Souvignier, C.; Mead, D.; Steinmetz, E.; Terry, K.; Kovacich, C.; Zegers, A.; Knox, C. Endotoxin-Free Protein Production—ClearColiTM Technology; Nature Publishing Group: New York, NY, USA, 2013. [Google Scholar]
  38. Schwarz, H.; Schmittner, M.; Duschl, A.; Horejs-Hoeck, J. Residual endotoxin contaminations in recombinant proteins are sufficient to activate human CD1c+ dendritic cells. PLoS ONE 2014, 9, e113840. [Google Scholar] [CrossRef]
  39. Palmer, I.; Wingfield, P.T. Preparation and extraction of insoluble (inclusion-body) proteins from Escherichia coli. Curr. Protoc. Protein Sci. 2012, 70, 3–6. [Google Scholar] [CrossRef] [PubMed]
  40. Sanchez-Garcia, L.; Martín, L.; Mangues, R.; Ferrer-Miralles, N.; Vázquez, E.; Villaverde, A. Recombinant pharmaceuticals from microbial cells: A 2015 update. Microb. Cell Fact. 2016, 15, 33. [Google Scholar] [CrossRef]
  41. Kyriakopoulos, S.; Kontoravdi, C. Analysis of the landscape of biologically-derived pharmaceuticals in Europe: Dominant production systems, molecule types on the rise and approval trends. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2013, 48, 428–441. [Google Scholar] [CrossRef] [PubMed]
  42. Proffitt, A. First HEV vaccine approved. Nat. Biotechnol. 2012, 30, 300. [Google Scholar] [CrossRef]
  43. Zhang, X.; Wei, M.; Pan, H.; Lin, Z.; Wang, K.; Weng, Z.; Zhu, Y.; Xin, L.; Zhang, J.; Li, S.; et al. Robust manufacturing and comprehensive characterization of recombinant hepatitis E virus-like particles in Hecolin(®). Vaccine 2014, 32, 4039–4050. [Google Scholar] [CrossRef] [PubMed]
  44. Guo, H.-C.; Sun, S.-Q.; Jin, Y.; Yang, S.-L.; Wei, Y.-Q.; Sun, D.-H.; Yin, S.-H.; Ma, J.-W.; Liu, Z.-X.; Guo, J.-H.; et al. Foot-and-mouth disease virus-like particles produced by a SUMO fusion protein system in Escherichia coli induce potent protective immune responses in guinea pigs, swine and cattle. Vet. Res. 2013, 44, 48. [Google Scholar] [CrossRef] [PubMed]
  45. Xiao, Y.; Chen, H.-Y.; Wang, Y.; Yin, B.; Lv, C.; Mo, X.; Yan, H.; Xuan, Y.; Huang, Y.; Pang, W.; et al. Large-scale production of foot-and-mouth disease virus (serotype Asia1) VLP vaccine in Escherichia coli and protection potency evaluation in cattle. BMC Biotechnol. 2016, 16, 56. [Google Scholar] [CrossRef] [PubMed]
  46. Li, S.; Zhao, R.; Song, H.; Pan, S.; Zhang, Y.; Dong, H.; Bai, M.; Sun, S.; Guo, H.; Yin, S. Local and systemic immune responses induced by intranasal immunization with biomineralized foot-and-mouth disease virus-like particles. Front. Microbiol. 2023, 14, 1112641. [Google Scholar] [CrossRef]
  47. Chathuranga, W.A.G.; Hewawaduge, C.; Nethmini, N.A.N.; Kim, T.-H.; Kim, J.H.; Ahn, Y.-H.; Yoon, I.-J.; Yoo, S.-S.; Park, J.-H.; Lee, J.-S. Efficacy of a Novel Multiepitope Vaccine Candidate against Foot-and-Mouth Disease Virus Serotype O and A. Vaccines 2022, 10, 2181. [Google Scholar] [CrossRef] [PubMed]
  48. Wibowo, N.; Hughes, F.K.; Fairmaid, E.J.; Lua, L.H.L.; Brown, L.E.; Middelberg, A.P.J. Protective efficacy of a bacterially produced modular capsomere presenting M2e from influenza: Extending the potential of broadly cross-protecting epitopes. Vaccine 2014, 32, 3651–3655. [Google Scholar] [CrossRef] [PubMed]
  49. Deng, L.; Cho, K.J.; Fiers, W.; Saelens, X. M2e-based universal influenza a vaccines. Vaccines 2015, 3, 105–136. [Google Scholar] [CrossRef] [PubMed]
  50. Watkins, H.C.; Pagan, C.L.; Childs, H.R.; Posada, S.; Chau, A.; Rios, J.; Guarino, C.; DeLisa, M.P.; Whittaker, G.R.; Putnam, D. A single dose and long lasting vaccine against pandemic influenza through the controlled release of a heterospecies tandem M2 sequence embedded within detoxified bacterial outer membrane vesicles. Vaccine 2017, 35, 5373–5380. [Google Scholar] [CrossRef]
  51. Liu, G.; Qiao, X.; Chang, C.; Hua, T.; Wang, J.; Tang, B.; Zhang, D. Reduction of Postweaning Multisystemic Wasting Syndrome-Associated Clinical Symptoms by Virus-Like Particle Vaccine Against Porcine Parvovirus and Porcine Circovirus Type 2. Viral Immunol. 2020, 33, 444–456. [Google Scholar] [CrossRef] [PubMed]
  52. Ke, Q.; Sun, P.; Wang, T.; Mi, T.; Xu, H.; Wu, J.; Liu, B. Non-glycosylated SARS-CoV-2 RBD elicited a robust neutralizing antibody response in mice. J. Immunol. Methods 2022, 506, 113279. [Google Scholar] [CrossRef]
  53. Liu, X.; Chang, X.; Rothen, D.; Derveni, M.; Krenger, P.; Roongta, S.; Wright, E.; Vogel, M.; Tars, K.; Mohsen, M.O. AP205 VLPs based on dimerized capsid proteins accommodate RBM domain of SARS-CoV-2 and serve as an attractive vaccine candidate. Vaccines 2021, 9, 403. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, W.S.; Kim, J.H.; Lee, J.; Ka, S.Y.; Chae, H.D.; Jung, I.; Jung, S.T.; Na, J.-H. Functional expression of the recombinant spike receptor binding domain of SARS-CoV-2 Omicron in the periplasm of Escherichia coli. Bioengineering 2022, 9, 670. [Google Scholar] [CrossRef] [PubMed]
  55. McGuire, B.E.; Mela, J.E.; Thompson, V.C.; Cucksey, L.R.; Stevens, C.E.; McWhinnie, R.L.; Winkler, D.F.H.; Pelech, S.; Nano, F.E. Escherichia coli recombinant expression of SARS-CoV-2 protein fragments. Microb. Cell Fact. 2022, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  56. Fitzgerald, G.A.; Komarov, A.; Kaznadzey, A.; Mazo, I.; Kireeva, M.L. Expression of SARS-CoV-2 surface glycoprotein fragment 319–640 in E. coli, and its refolding and purification. Protein Expr. Purif. 2021, 183, 105861. [Google Scholar] [CrossRef]
  57. He, Y.; Qi, J.; Xiao, L.; Shen, L.; Yu, W.; Hu, T. Purification and characterization of the receptor-binding domain of SARS-CoV-2 spike protein from Escherichia coli. Eng. Life Sci. 2021, 21, 453–460. [Google Scholar] [CrossRef]
  58. Clem, R.J.; Passarelli, A.L. Baculoviruses: Sophisticated pathogens of insects. PLoS Pathog. 2013, 9, e1003729. [Google Scholar] [CrossRef] [PubMed]
  59. Pennock, G.D.; Shoemaker, C.; Miller, L.K. Strong and regulated expression of Escherichia coli beta-galactosidase in insect cells with a baculovirus vector. Mol. Cell. Biol. 1984, 4, 399–406. [Google Scholar] [CrossRef] [PubMed]
  60. Smith, G.E.; Summers, M.D.; Fraser, M.J. Production of human beta interferon in insect cells infected with a baculovirus expression vector. Mol. Cell. Biol. 1983, 3, 2156–2165. [Google Scholar] [CrossRef] [PubMed]
  61. Hitchman, R.B.; Possee, R.D.; King, L.A. High-throughput baculovirus expression in insect cells. Methods Mol. Biol. 2012, 824, 609–627. [Google Scholar] [CrossRef] [PubMed]
  62. Hitchman, R.B.; Locanto, E.; Possee, R.D.; King, L.A. Optimizing the baculovirus expression vector system. Methods 2011, 55, 52–57. [Google Scholar] [CrossRef] [PubMed]
  63. Kitts, P.A.; Ayres, M.D.; Possee, R.D. Linearization of baculovirus DNA enhances the recovery of recombinant virus expression vectors. Nucleic Acids Res. 1990, 18, 5667–5672. [Google Scholar] [CrossRef] [PubMed]
  64. Kitts, P.A.; Possee, R.D. A method for producing recombinant baculovirus expression vectors at high frequency. Biotechniques 1993, 14, 810–817. [Google Scholar]
  65. Luckow, V.A.; Lee, S.C.; Barry, G.F.; Olins, P.O. Efficient generation of infectious recombinant baculoviruses by site-specific transposon-mediated insertion of foreign genes into a baculovirus genome propagated in Escherichia coli. J. Virol. 1993, 67, 4566–4579. [Google Scholar] [CrossRef] [PubMed]
  66. Berger, I.; Poterszman, A. Baculovirus expression: Old dog, new tricks. Bioengineered 2015, 6, 316–322. [Google Scholar] [CrossRef]
  67. Shi, X.; Jarvis, D.L. Protein N-glycosylation in the baculovirus-insect cell system. Curr. Drug Targets 2007, 8, 1116–1125. [Google Scholar] [CrossRef] [PubMed]
  68. Hong, M.; Li, T.; Xue, W.; Zhang, S.; Cui, L.; Wang, H.; Zhang, Y.; Zhou, L.; Gu, Y.; Xia, N. Genetic engineering of baculovirus-insect cell system to improve protein production. Front. Bioeng. Biotechnol. 2022, 10, 994743. [Google Scholar] [CrossRef] [PubMed]
  69. Geisler, C.; Mabashi-Asazuma, H.; Jarvis, D.L. An overview and history of glyco-engineering in insect expression systems. Glyco-Eng. Methods Protoc. 2015, 1321, 131–152. [Google Scholar]
  70. Palomares, L.A.; Srivastava, I.K.; Ramírez, O.T.; Cox, M.M.J. Glycobiotechnology of the insect cell-baculovirus expression system technology. In Advances in Glycobiotechnology; Springer: Berlin/Heidelberg, Germany, 2018; pp. 71–92. [Google Scholar]
  71. Palmberger, D.; Ashjaei, K.; Strell, S.; Hoffmann-Sommergruber, K.; Grabherr, R. Minimizing fucosylation in insect cell-derived glycoproteins reduces binding to IgE antibodies from the sera of patients with allergy. Biotechnol. J. 2014, 9, 1206–1214. [Google Scholar] [CrossRef] [PubMed]
  72. Jarvis, D.L. Developing baculovirus-insect cell expression systems for humanized recombinant glycoprotein production. Virology 2003, 310, 1–7. [Google Scholar] [CrossRef] [PubMed]
  73. Unger, T.; Peleg, Y. Recombinant protein expression in the baculovirus-infected insect cell system. In Chemical Genomics and Proteomics; Springer: Berlin/Heidelberg, Germany, 2012; pp. 187–199. [Google Scholar]
  74. Rendić, D.; Wilson, I.B.H.; Paschinger, K. The glycosylation capacity of insect cells. Croat. Chem. Acta 2008, 81, 7–21. [Google Scholar]
  75. Frenzel, A.; Hust, M.; Schirrmann, T. Expression of recombinant antibodies. Front. Immunol. 2013, 4, 217. [Google Scholar] [CrossRef]
  76. Futatsumori-Sugai, M.; Tsumoto, K. Signal peptide design for improving recombinant protein secretion in the baculovirus expression vector system. Biochem. Biophys. Res. Commun. 2010, 391, 931–935. [Google Scholar] [CrossRef] [PubMed]
  77. Soejima, Y.; Lee, J.M.; Nagata, Y.; Mon, H.; Iiyama, K.; Kitano, H.; Matsuyama, M.; Kusakabe, T. Comparison of signal peptides for efficient protein secretion in the baculovirus-silkworm system. Cent. Eur. J. Biol. 2013, 8, 1–7. [Google Scholar] [CrossRef]
  78. Mori, K.; Hamada, H.; Ogawa, T.; Ohmuro-Matsuyama, Y.; Katsuda, T.; Yamaji, H. Efficient production of antibody Fab fragment by transient gene expression in insect cells. J. Biosci. Bioeng. 2017, 124, 221–226. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Bleckmann, M.; Schmelz, S.; Schinkowski, C.; Scrima, A.; van den Heuvel, J. Fast plasmid based protein expression analysis in insect cells using an automated SplitGFP screen. Biotechnol. Bioeng. 2016, 113, 1975–1983. [Google Scholar] [CrossRef] [PubMed]
  81. 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] [PubMed]
  82. Shen, X.; Hacker, D.L.; Baldi, L.; Wurm, F.M. Virus-free transient protein production in Sf9 cells. J. Biotechnol. 2014, 171, 61–70. [Google Scholar] [CrossRef]
  83. Korn, J.; Schäckermann, D.; Kirmann, T.; Bertoglio, F.; Steinke, S.; Heisig, J.; Ruschig, M.; Rojas, G.; Langreder, N.; Wenzel, E.V.; et al. Baculovirus-free insect cell expression system for high yield antibody and antigen production. Sci. Rep. 2020, 10, 21393. [Google Scholar] [CrossRef]
  84. Ogay, I.D.; Lihoradova, O.A.; Azimova, S.S.; Abdukarimov, A.A.; Slack, J.M.; Lynn, D.E. Transfection of insect cell lines using polyethylenimine. Cytotechnology 2006, 51, 89–98. [Google Scholar]
  85. Puente-Massaguer, E.; Lecina, M.; Gòdia, F. Nanoscale characterization coupled to multi-parametric optimization of Hi5 cell transient gene expression. Appl. Microbiol. Biotechnol. 2018, 102, 10495–10510. [Google Scholar] [CrossRef] [PubMed]
  86. Kumar, N.; Pandey, D.; Halder, A. Preventive, Diagnostic and Therapeutic Applications of Baculovirus Expression Vector System BT—Trends in Insect Molecular Biology and Biotechnology; Kumar, D., Gong, C., Eds.; Springer International Publishing: Cham, Switzerland, 2018; pp. 163–191. [Google Scholar]
  87. Felberbaum, R.S. The baculovirus expression vector system: A commercial manufacturing platform for viral vaccines and gene therapy vectors. Biotechnol. J. 2015, 10, 702–714. [Google Scholar] [CrossRef] [PubMed]
  88. To, A.; Lai, C.-Y.; Wong, T.A.; Namekar, M.; Lehrer, A.T. A recombinant subunit Lassa virus vaccine elicits strong antibody responses. J. Immunol. 2020, 204, 167.30. [Google Scholar] [CrossRef]
  89. Qu, P.; Zhang, W.; Li, D.; Zhang, C.; Liu, Q.; Zhang, X.; Wang, X.; Dai, W.; Xu, Y.; Leng, Q.; et al. Insect cell-produced recombinant protein subunit vaccines protect against Zika virus infection. Antivir. Res. 2018, 154, 97–103. [Google Scholar] [CrossRef] [PubMed]
  90. Blome, S.; Meindl-Böhmer, A.; Loeffen, W.; Moennig, V. Assessment of classical swine fever diagnostics and vaccine performance. Int. Off. Epizoot. 2006, 25, 1025–1038. [Google Scholar]
  91. Monie, A.; Hung, C.-F.; Roden, R.; Wu, T.-C. Cervarix: A vaccine for the prevention of HPV 16, 18-associated cervical cancer. Biologics 2008, 2, 97–105. [Google Scholar] [CrossRef]
  92. Cox, M.M.J.; Patriarca, P.A.; Treanor, J. FluBlok, a recombinant hemagglutinin influenza vaccine. Influenza Other Respi. Viruses 2008, 2, 211–219. [Google Scholar] [CrossRef] [PubMed]
  93. Fort, M.; Sibila, M.; Pérez-Martín, E.; Nofrarías, M.; Mateu, E.; Segalés, J. One dose of a porcine circovirus 2 (PCV2) sub-unit vaccine administered to 3-week-old conventional piglets elicits cell-mediated immunity and significantly reduces PCV2 viremia in an experimental model. Vaccine 2009, 27, 4031–4037. [Google Scholar] [CrossRef]
  94. Coll, T.; Villalba, D.; Maass, P. Meta-Analysis of Globally Published Results on the Efficacy of Ingelvac CircoFLEX® Vaccination. 2010. Available online: https://hdl.handle.net/11299/135564 (accessed on 1 June 2024).
  95. Sno, M.; Cox, E.; Holtslag, H.; Nell, T.; Pel, S.; Segers, R.; Fachinger, V.; Witvliet, M. Efficacy and safety of a new intradermal PCV2 vaccine in pigs. Trials Vaccinol. 2016, 5, 24–31. [Google Scholar] [CrossRef]
  96. Metz, S.W.; Gardner, J.; Geertsema, C.; Le, T.T.; Goh, L.; Vlak, J.M.; Suhrbier, A.; Pijlman, G.P. Effective chikungunya virus-like particle vaccine produced in insect cells. PLoS Negl. Trop. Dis. 2013, 7, e2124. [Google Scholar] [CrossRef] [PubMed]
  97. Metz, S.W.; Martina, B.E.; van den Doel, P.; Geertsema, C.; Osterhaus, A.D.; Vlak, J.M.; Pijlman, G.P. Chikungunya virus-like particles are more immunogenic in a lethal AG129 mouse model compared to glycoprotein E1 or E2 subunits. Vaccine 2013, 31, 6092–6096. [Google Scholar] [CrossRef] [PubMed]
  98. Tran, H.T.T.; Truong, D.A.; Ly, V.D.; Vu, H.T.; Van Hoang, T.; Nguyen, C.T.; Chu, N.T.; Nguyen, V.T.; Nguyen, D.T.; Miyazawa, K.; et al. The potential efficacy of the E2-subunit vaccine to protect pigs against different genotypes of classical swine fever virus circulating in Vietnam. Clin. Exp. Vaccine Res. 2020, 9, 26–39. [Google Scholar] [CrossRef] [PubMed]
  99. Liu, Y.; Wen, Z.; Carrion, R.J.; Nunneley, J.; Staples, H.; Ticer, A.; Patterson, J.L.; Compans, R.W.; Ye, L.; Yang, C. Intradermal Immunization of EBOV VLPs in Guinea Pigs Induces Broader Antibody Responses Against GP Than Intramuscular Injection. Front. Microbiol. 2020, 11, 304. [Google Scholar] [CrossRef]
  100. Sunwoo, S.Y.; Noronha, L.E.; Morozov, I.; Trujillo, J.D.; Kim, I.J.; Schirtzinger, E.E.; Faburay, B.; Drolet, B.S.; Urbaniak, K.; Scott Mcvey, D.; et al. Evaluation of a baculovirus-expressed VP2 subunit vaccine for the protection of white-tailed deer (Odocoileus virginianus) from epizootic hemorrhagic disease. Vaccines 2020, 8, 59. [Google Scholar] [CrossRef] [PubMed]
  101. Pyo, H.-M.; Masic, A.; Woldeab, N.; Embury-Hyatt, C.; Lin, L.; Shin, Y.-K.; Song, J.-Y.; Babiuk, S.; Zhou, Y. Pandemic H1N1 influenza virus-like particles are immunogenic and provide protective immunity to pigs. Vaccine 2012, 30, 1297–1304. [Google Scholar] [CrossRef]
  102. Zhu, W.-Z.; Wen, Y.-C.; Lin, S.-Y.; Chen, T.-C.; Chen, H.-W. Anti-influenza protective efficacy of a H6 virus-like particle in chickens. Vaccines 2020, 8, 465. [Google Scholar] [CrossRef] [PubMed]
  103. Faburay, B.; Wilson, W.C.; Gaudreault, N.N.; Davis, A.S.; Shivanna, V.; Bawa, B.; Sunwoo, S.Y.; Ma, W.; Drolet, B.S.; Morozov, I.; et al. A Recombinant Rift Valley Fever Virus Glycoprotein Subunit Vaccine Confers Full Protection against Rift Valley Fever Challenge in Sheep. Sci. Rep. 2016, 6, 27719. [Google Scholar] [CrossRef]
  104. Faburay, B.; Lebedev, M.; McVey, D.S.; Wilson, W.; Morozov, I.; Young, A.; Richt, J.A. A Glycoprotein Subunit Vaccine Elicits a Strong Rift Valley Fever Virus Neutralizing Antibody Response in Sheep. Vector-Borne Zoonotic Dis. 2014, 14, 746–756. [Google Scholar] [CrossRef]
  105. Yang, J.; Wang, W.; Chen, Z.; Lu, S.; Yang, F.; Bi, Z.; Bao, L.; Mo, F.; Li, X.; Huang, Y. A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature 2020, 586, 572–577. [Google Scholar] [CrossRef] [PubMed]
  106. Hong, S.-H.; Oh, H.; Park, Y.W.; Kwak, H.W.; Oh, E.Y.; Park, H.-J.; Kang, K.W.; Kim, G.; Koo, B.-S.; Hwang, E.-H.; et al. Immunization with RBD-P2 and N protects against SARS-CoV-2 in nonhuman primates. Sci. Adv. 2023, 7, eabg7156. [Google Scholar] [CrossRef] [PubMed]
  107. Fernandes, B.; Castro, R.; Bhoelan, F.; Bemelman, D.; Gomes, R.A.; Costa, J.; Gomes-Alves, P.; Stegmann, T.; Amacker, M.; Alves, P.M. Insect cells for high-yield production of SARS-CoV-2 spike protein: Building a virosome-based COVID-19 vaccine candidate. Pharmaceutics 2022, 14, 854. [Google Scholar] [CrossRef] [PubMed]
  108. Mi, Y.; Xie, T.; Zhu, B.; Tan, J.; Li, X.; Luo, Y.; Li, F.; Niu, H.; Han, J.; Lv, W. Production of SARS-CoV-2 virus-like particles in insect cells. Vaccines 2021, 9, 554. [Google Scholar] [CrossRef]
  109. Jaron, M.; Lehky, M.; Zarà, M.; Zaydowicz, C.N.; Lak, A.; Ballmann, R.; Heine, P.A.; Wenzel, E.V.; Schneider, K.-T.; Bertoglio, F. Baculovirus-Free SARS-CoV-2 Virus-like Particle Production in Insect Cells for Rapid Neutralization Assessment. Viruses 2022, 14, 2087. [Google Scholar] [CrossRef] [PubMed]
  110. Struble, L.R.; Smith, A.L.; Lutz, W.E.; Grubbs, G.; Sagar, S.; Bayles, K.W.; Radhakrishnan, P.; Khurana, S.; El-Gamal, D.; Borgstahl, G.E.O. Insect cell expression and purification of recombinant SARS-COV-2 spike proteins that demonstrate ACE2 binding. Protein Sci. 2022, 31, e4300. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, Y.; Wu, Z.; Hu, W.; Hao, P.; Yang, S. Impact of expressing cells on glycosylation and glycan of the SARS-CoV-2 spike glycoprotein. ACS Omega 2021, 6, 15988–15999. [Google Scholar] [CrossRef] [PubMed]
  112. Li, T.; Zheng, Q.; Yu, H.; Wu, D.; Xue, W.; Xiong, H.; Huang, X.; Nie, M.; Yue, M.; Rong, R.; et al. SARS-CoV-2 spike produced in insect cells elicits high neutralization titres in non-human primates. Emerg. Microbes Infect. 2020, 9, 2076–2090. [Google Scholar] [CrossRef] [PubMed]
  113. Bandaranayake, A.D.; Almo, S.C. Recent advances in mammalian protein production. FEBS Lett. 2014, 588, 253–260. [Google Scholar] [CrossRef]
  114. Zhu, J. Mammalian cell protein expression for biopharmaceutical production. Biotechnol. Adv. 2012, 30, 1158–1170. [Google Scholar] [CrossRef] [PubMed]
  115. Jenkins, N.; Meleady, P.; Tyther, R.; Murphy, L. Strategies for analysing and improving the expression and quality of recombinant proteins made in mammalian cells. Biotechnol. Appl. Biochem. 2009, 53, 73–83. [Google Scholar] [CrossRef] [PubMed]
  116. Butler, M. Animal cell cultures: Recent achievements and perspectives in the production of biopharmaceuticals. Appl. Microbiol. Biotechnol. 2005, 68, 283–291. [Google Scholar] [CrossRef] [PubMed]
  117. Wiznerowicz, M.; Trono, D. Harnessing HIV for therapy, basic research and biotechnology. Trends Biotechnol. 2005, 23, 42–47. [Google Scholar] [CrossRef] [PubMed]
  118. Giry-Laterrière, M.; Verhoeyen, E.; Salmon, P. Lentiviral vectors. Methods Mol. Biol. 2011, 737, 183–209. [Google Scholar] [CrossRef]
  119. Ramezani, A.; Hawley, R.G. Overview of the HIV-1 Lentiviral Vector System. Curr. Protoc. Mol. Biol. 2002, 60, 16–21. [Google Scholar] [CrossRef]
  120. Kafri, T.; van Praag, H.; Gage, F.H.; Verma, I.M. Lentiviral Vectors: Regulated Gene Expression. Mol. Ther. 2000, 1, 516–521. [Google Scholar] [CrossRef] [PubMed]
  121. Tiscornia, G.; Singer, O.; Verma, I.M. Production and purification of lentiviral vectors. Nat. Protoc. 2006, 1, 241–245. [Google Scholar] [CrossRef]
  122. Rueckert, C.; Guzmán, C.A. Vaccines: From empirical development to rational design. PLoS Pathog. 2012, 8, e1003001. [Google Scholar] [CrossRef] [PubMed]
  123. Geisse, S. Reflections on more than 10 years of TGE approaches. Protein Expr. Purif. 2009, 64, 99–107. [Google Scholar] [CrossRef]
  124. Liu, C.; Dalby, B.; Chen, W.; Kilzer, J.M.; Chiou, H.C. Transient transfection factors for high-level recombinant protein production in suspension cultured mammalian cells. Mol. Biotechnol. 2008, 39, 141–153. [Google Scholar] [CrossRef] [PubMed]
  125. 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]
  126. Rozkov, A.; Larsson, B.; Gillström, S.; Björnestedt, R.; Schmidt, S.R. Large-scale production of endotoxin-free plasmids for transient expression in mammalian cell culture. Biotechnol. Bioeng. 2008, 99, 557–566. [Google Scholar] [CrossRef] [PubMed]
  127. Yarranton, G.T. Inducible vectors for expression in mammalian cells. Curr. Opin. Biotechnol. 1992, 3, 506–511. [Google Scholar] [CrossRef] [PubMed]
  128. Fike, R. Nutrient supplementation strategies for biopharmaceutical production, Part 3. Bioproc. Int. 2010, 8, 24–31. [Google Scholar]
  129. Lufino, M.M.P.; Edser, P.A.H.; Wade-Martins, R. Advances in high-capacity extrachromosomal vector technology: Episomal maintenance, vector delivery, and transgene expression. Mol. Ther. 2008, 16, 1525–1538. [Google Scholar] [CrossRef] [PubMed]
  130. Cacciatore, J.J.; Chasin, L.A.; Leonard, E.F. Gene amplification and vector engineering to achieve rapid and high-level therapeutic protein production using the Dhfr-based CHO cell selection system. Biotechnol. Adv. 2010, 28, 673–681. [Google Scholar] [CrossRef]
  131. Barnes, L.M.; Bentley, C.M.; Dickson, A.J. Advances in animal cell recombinant protein production: GS-NS0 expression system. Cytotechnology 2000, 32, 109–123. [Google Scholar] [CrossRef] [PubMed]
  132. Jadhav, V.; Hackl, M.; Druz, A.; Shridhar, S.; Chung, C.-Y.; Heffner, K.M.; Kreil, D.P.; Betenbaugh, M.; Shiloach, J.; Barron, N. CHO microRNA engineering is growing up: Recent successes and future challenges. Biotechnol. Adv. 2013, 31, 1501–1513. [Google Scholar] [CrossRef]
  133. Jefferis, R. Glycosylation as a strategy to improve antibody-based therapeutics. Nat. Rev. Drug Discov. 2009, 8, 226–234. [Google Scholar] [CrossRef] [PubMed]
  134. Costa, A.R.; Rodrigues, M.E.; Henriques, M.; Oliveira, R.; Azeredo, J. Glycosylation: Impact, control and improvement during therapeutic protein production. Crit. Rev. Biotechnol. 2014, 34, 281–299. [Google Scholar] [CrossRef] [PubMed]
  135. Beck, A.; Wagner-Rousset, E.; Bussat, M.-C.; Lokteff, M.; Klinguer-Hamour, C.; Haeuw, J.-F.; Goetsch, L.; Wurch, T.; Dorsselaer, A.V.; Corvaïa, N. Trends in glycosylation, glycoanalysis and glycoengineering of therapeutic antibodies and Fc-fusion proteins. Curr. Pharm. Biotechnol. 2008, 9, 482–501. [Google Scholar] [CrossRef]
  136. Bosques, C.J.; Collins, B.E.; Meador, J.W.; Sarvaiya, H.; Murphy, J.L.; DelloRusso, G.; Bulik, D.A.; Hsu, I.-H.; Washburn, N.; Sipsey, S.F. Chinese hamster ovary cells can produce galactose-α-1, 3-galactose antigens on proteins. Nat. Biotechnol. 2010, 28, 1153–1156. [Google Scholar] [CrossRef] [PubMed]
  137. Hills, A.E.; Patel, A.; Boyd, P.; James, D.C. Metabolic control of recombinant monoclonal antibody N-glycosylation in GS-NS0 cells. Biotechnol. Bioeng. 2001, 75, 239–251. [Google Scholar] [CrossRef]
  138. Baker, K.N.; Rendall, M.H.; Hills, A.E.; Hoare, M.; Freedman, R.B.; James, D.C. Metabolic control of recombinant protein N-glycan processing in NS0 and CHO cells. Biotechnol. Bioeng. 2001, 73, 188–202. [Google Scholar] [CrossRef] [PubMed]
  139. Sordo-Puga, Y.; Suárez-Pedroso, M.; Naranjo-Valdéz, P.; Pérez-Pérez, D.; Santana-Rodríguez, E.; Sardinas-Gonzalez, T.; Mendez-Orta, M.K.; Duarte-Cano, C.A.; Estrada-Garcia, M.P.; Rodríguez-Moltó, M.P. Porvac® Subunit Vaccine E2-CD154 Induces Remarkable Rapid Protection against Classical Swine Fever Virus. Vaccines 2021, 9, 167. [Google Scholar] [CrossRef] [PubMed]
  140. Pérez Rubio, A.; Eiros, J.M. Cell culture-derived flu vaccine: Present and future. Hum. Vaccines Immunother. 2018, 14, 1874–1882. [Google Scholar] [CrossRef]
  141. Partridge, J.; Kieny, M.P. Global production capacity of seasonal influenza vaccine in 2011. Vaccine 2013, 31, 728–731. [Google Scholar] [CrossRef] [PubMed]
  142. Lamb, Y.N. Cell-Based Quadrivalent Inactivated InfluenzaVirus Vaccine (Flucelvax® Tetra/FlucelvaxQuadrivalent®): A Review in the Prevention ofInfluenza. Drugs 2019, 79, 1337–1348. [Google Scholar] [CrossRef]
  143. Bouzya, B.; Rouxel, R.N.; Sacconnay, L.; Mascolo, R.; Nols, L.; Quique, S.; François, L.; Atas, A.; Warter, L.; Dezutter, N.; et al. Immunogenicity of an AS01-adjuvanted respiratory syncytial virus prefusion F (RSVPreF3) vaccine in animal models. npj Vaccines 2023, 8, 143. [Google Scholar] [CrossRef] [PubMed]
  144. Kotb, S.; Haranaka, M.; Folschweiller, N.; Nakanwagi, P.; Verheust, C.; De Schrevel, N.; David, M.-P.; Mesaros, N.; Hulstrøm, V. Safety and immunogenicity of a respiratory syncytial virus prefusion F protein (RSVPreF3) candidate vaccine in older Japanese adults: A phase I, randomized, observer-blind clinical trial. Respir. Investig. 2023, 61, 261–269. [Google Scholar] [CrossRef] [PubMed]
  145. Kopera, E.; Czajka, H.; Zapolnik, P.; Mazur, A. New Insights on Respiratory Syncytial Virus Prevention. Vaccines 2023, 11, 1797. [Google Scholar] [CrossRef] [PubMed]
  146. Leroux-Roels, I.; Davis, M.G.; Steenackers, K.; Essink, B.; Vandermeulen, C.; Fogarty, C.; Andrews, C.P.; Kerwin, E.; David, M.-P.; Fissette, L.; et al. Safety and Immunogenicity of a Respiratory Syncytial Virus Prefusion F (RSVPreF3) Candidate Vaccine in Older Adults: Phase 1/2 Randomized Clinical Trial. J. Infect. Dis. 2023, 227, 761–772. [Google Scholar] [CrossRef] [PubMed]
  147. Bebia, Z.; Reyes, O.; Jeanfreau, R.; Kantele, A.; De Leon, R.G.; Sánchez, M.G.; Banooni, P.; Gardener, G.J.; Rasero, J.L.B.; Pardilla, M.B.E.; et al. Safety and Immunogenicity of an Investigational Respiratory Syncytial Virus Vaccine (RSVPreF3) in Mothers and Their Infants: A Phase 2 Randomized Trial. J. Infect. Dis. 2023, 228, 299–310. [Google Scholar] [CrossRef] [PubMed]
  148. Fleming-Dutra, K.E. Use of the Pfizer respiratory syncytial virus vaccine during pregnancy for the prevention of respiratory syncytial virus–associated lower respiratory tract disease in infants: Recommendations of the Advisory Committee on Immunization Practices—United Stat. MMWR. Morb. Mortal. Wkly. Rep. 2023, 72, 1115–1122. [Google Scholar] [CrossRef] [PubMed]
  149. Che, Y.; Gribenko, A.V.; Song, X.; Handke, L.D.; Efferen, K.S.; Tompkins, K.; Kodali, S.; Nunez, L.; Prasad, A.K.; Phelan, L.M.; et al. Rational design of a highly immunogenic prefusion-stabilized F glycoprotein antigen for a respiratory syncytial virus vaccine. Sci. Transl. Med. 2024, 15, eade6422. [Google Scholar] [CrossRef] [PubMed]
  150. Walsh, E.E.; Falsey, A.R.; Scott, D.A.; Gurtman, A.; Zareba, A.M.; Jansen, K.U.; Gruber, W.C.; Dormitzer, P.R.; Swanson, K.A.; Radley, D.; et al. A Randomized Phase 1/2 Study of a Respiratory Syncytial Virus Prefusion F Vaccine. J. Infect. Dis. 2022, 225, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
  151. Melgar, M.; Britton, A. Evidence to Recommendations Framework: Respiratory Syncytial Virus (RSV) in Adults Pfizer bivalent RSVpreF vaccine in older adults GSK adjuvanted RSVPreF3 vaccine in older adults. In Proceedings of the ACIP Meeting, Atlanta, GA, USA, 21–23 June 2023. [Google Scholar]
  152. Li, J.; Li, X.; Ma, H.; Ren, X.; Hao, G.; Zhang, H.; Zhao, Z.; Fang, K.; Li, X.; Rong, Z.; et al. Efficient mucosal vaccination of a novel classical swine fever virus E2-Fc fusion protein mediated by neonatal Fc receptor. Vaccine 2020, 38, 4574–4583. [Google Scholar] [CrossRef] [PubMed]
  153. Bossart, K.N.; Crameri, G.; Dimitrov, A.S.; Mungall, B.A.; Feng, Y.-R.; Patch, J.R.; Choudhary, A.; Wang, L.-F.; Eaton, B.T.; Broder, C.C. Receptor binding, fusion inhibition, and induction of cross-reactive neutralizing antibodies by a soluble G glycoprotein of Hendra virus. J. Virol. 2005, 79, 6690–6702. [Google Scholar] [CrossRef] [PubMed]
  154. Bossart, K.N.; Rockx, B.; Feldmann, F.; Brining, D.; Scott, D.; LaCasse, R.; Geisbert, J.B.; Feng, Y.-R.; Chan, Y.-P.; Hickey, A.C.; et al. A Hendra virus G glycoprotein subunit vaccine protects African green monkeys from Nipah virus challenge. Sci. Transl. Med. 2012, 4, 146ra107. [Google Scholar] [CrossRef] [PubMed]
  155. McEachern, J.A.; Bingham, J.; Crameri, G.; Green, D.J.; Hancock, T.J.; Middleton, D.; Feng, Y.-R.; Broder, C.C.; Wang, L.-F.; Bossart, K.N. A recombinant subunit vaccine formulation protects against lethal Nipah virus challenge in cats. Vaccine 2008, 26, 3842–3852. [Google Scholar] [CrossRef] [PubMed]
  156. Pallister, J.; Middleton, D.; Wang, L.-F.; Klein, R.; Haining, J.; Robinson, R.; Yamada, M.; White, J.; Payne, J.; Feng, Y.-R.; et al. A recombinant Hendra virus G glycoprotein-based subunit vaccine protects ferrets from lethal Hendra virus challenge. Vaccine 2011, 29, 5623–5630. [Google Scholar] [CrossRef] [PubMed]
  157. Middleton, D.; Pallister, J.; Klein, R.; Feng, Y.-R.; Haining, J.; Arkinstall, R.; Frazer, L.; Huang, J.-A.; Edwards, N.; Wareing, M.; et al. Hendra virus vaccine, a one health approach to protecting horse, human, and environmental health. Emerg. Infect. Dis. 2014, 20, 372–379. [Google Scholar] [CrossRef] [PubMed]
  158. Pallister, J.A.; Klein, R.; Arkinstall, R.; Haining, J.; Long, F.; White, J.R.; Payne, J.; Feng, Y.-R.; Wang, L.-F.; Broder, C.C.; et al. Vaccination of ferrets with a recombinant G glycoprotein subunit vaccine provides protection against Nipah virus disease for over 12 months. Virol. J. 2013, 10, 237. [Google Scholar] [CrossRef]
  159. Young-Il, K.; Dokyun, K.; Kwang-Min, Y.; David, S.H.; Shin-Ae, L.; Casel, M.A.B.; Seung-Gyu, J.; Stephanie, K.; WooRam, J.; Chih-Jen, L.; et al. Development of Spike Receptor-Binding Domain Nanoparticles as a Vaccine Candidate against SARS-CoV-2 Infection in Ferrets. MBio 2021, 12, e00230-21. [Google Scholar] [CrossRef]
  160. Kassardjian, A.; Sun, E.; Sookhoo, J.; Muthuraman, K.; Boligan, K.F.; Kucharska, I.; Rujas, E.; Jetha, A.; Branch, D.R.; Babiuk, S. Modular adjuvant-free pan-HLA-DR-immunotargeting subunit vaccine against SARS-CoV-2 elicits broad sarbecovirus-neutralizing antibody responses. Cell Rep. 2023, 42, 112391. [Google Scholar] [CrossRef] [PubMed]
  161. Colgrave, M.L.; Snelling, H.J.; Shiell, B.J.; Feng, Y.-R.; Chan, Y.-P.; Bossart, K.N.; Xu, K.; Nikolov, D.B.; Broder, C.C.; Michalski, W.P. Site occupancy and glycan compositional analysis of two soluble recombinant forms of the attachment glycoprotein of Hendra virus. Glycobiology 2012, 22, 572–584. [Google Scholar] [CrossRef]
  162. Gstöttner, C.; Zhang, T.; Resemann, A.; Ruben, S.; Pengelley, S.; Suckau, D.; Welsink, T.; Wuhrer, M.; Domínguez-Vega, E. Structural and Functional Characterization of SARS-CoV-2 RBD Domains Produced in Mammalian Cells. Anal. Chem. 2021, 93, 6839–6847. [Google Scholar] [CrossRef] [PubMed]
  163. Arbeitman, C.R.; Auge, G.; Blaustein, M.; Bredeston, L.; Corapi, E.S.; Craig, P.O.; Cossio, L.A.; Dain, L.; D’Alessio, C.; Elias, F.; et al. Structural and functional comparison of SARS-CoV-2-spike receptor binding domain produced in Pichia pastoris and mammalian cells. Sci. Rep. 2020, 10, 21779. [Google Scholar] [CrossRef]
  164. Brindha, S.; Kuroda, Y. A Multi-Disulfide Receptor-Binding Domain (RBD) of the SARS-CoV-2 Spike Protein Expressed in E. coli Using a SEP-Tag Produces Antisera Interacting with the Mammalian Cell Expressed Spike (S1) Protein. Int. J. Mol. Sci. 2022, 23, 1703. [Google Scholar] [CrossRef]
  165. Merkuleva, I.A.; Shcherbakov, D.N.; Borgoyakova, M.B.; Shanshin, D.V.; Rudometov, A.P.; Karpenko, L.I.; Belenkaya, S.V.; Isaeva, A.A.; Nesmeyanova, V.S.; Kazachinskaia, E.I.; et al. Comparative Immunogenicity of the Recombinant Receptor-Binding Domain of Protein S SARS-CoV-2 Obtained in Prokaryotic and Mammalian Expression Systems. Vaccines 2022, 10, 96. [Google Scholar] [CrossRef] [PubMed]
  166. Arias-Arias, J.L.; Molina-Castro, S.E.; Monturiol-Gross, L.; Lomonte, B.; Corrales-Aguilar, E. Stable production of recombinant SARS-CoV-2 receptor-binding domain in mammalian cells with co-expression of a fluorescent reporter and its validation as antigenic target for COVID-19 serology testing. Biotechnol. Rep. 2023, 37, e00780. [Google Scholar] [CrossRef] [PubMed]
  167. Xu, R.; Shi, M.; Li, J.; Song, P.; Li, N. Construction of SARS-CoV-2 virus-like particles by mammalian expression system. Front. Bioeng. Biotechnol. 2020, 8, 862. [Google Scholar]
  168. García-Cordero, J.; Mendoza-Ramírez, J.; Fernández-Benavides, D.; Roa-Velazquez, D.; Filisola-Villaseñor, J.; Martínez-Frías, S.P.; Sanchez-Salguero, E.S.; Miguel-Rodríguez, C.E.; Maravillas Montero, J.L.; Torres-Ruiz, J.J.; et al. Recombinant Protein Expression and Purification of N, S1, and RBD of SARS-CoV-2 from Mammalian Cells and Their Potential Applications. Diagnostics 2021, 11, 1808. [Google Scholar] [CrossRef] [PubMed]
  169. Shouval, D. Hepatitis B vaccines. J. Hepatol. 2003, 39 (Suppl. 1), S70–S76. [Google Scholar] [CrossRef]
  170. Bryan, J.T. Developing an HPV vaccine to prevent cervical cancer and genital warts. Vaccine 2007, 25, 3001–3006. [Google Scholar] [CrossRef] [PubMed]
  171. Damasceno, L.M.; Huang, C.-J.; Batt, C.A. Protein secretion in Pichia pastoris and advances in protein production. Appl. Microbiol. Biotechnol. 2012, 93, 31–39. [Google Scholar] [CrossRef]
  172. Jeong, K.J.; Jang, S.H.; Velmurugan, N. Recombinant antibodies: Engineering and production in yeast and bacterial hosts. Biotechnol. J. 2011, 6, 16–27. [Google Scholar] [CrossRef]
  173. Cregg, J.M.; Tolstorukov, I.; Kusari, A.; Sunga, J.; Madden, K.; Chappell, T. Expression in the yeast Pichia pastoris. In Methods in Enzymology; Elsevier: Amsterdam, The Netherlands, 2009; Volume 463, pp. 169–189. [Google Scholar]
  174. Jahic, M.; Veide, A.; Charoenrat, T.; Teeri, T.; Enfors, S. Process technology for production and recovery of heterologous proteins with Pichia pastoris. Biotechnol. Prog. 2006, 22, 1465–1473. [Google Scholar] [CrossRef]
  175. Tomimoto, K.; Fujita, Y.; Iwaki, T.; Chiba, Y.; Jigami, Y.; Nakayama, K.; Nakajima, Y.; Abe, H. Protease-deficient Saccharomyces cerevisiae strains for the synthesis of human-compatible glycoproteins. Biosci. Biotechnol. Biochem. 2013, 77, 2461–2466. [Google Scholar] [CrossRef]
  176. Celik, E.; Calık, P. Production of recombinant proteins by yeast cells. Biotechnol. Adv. 2012, 30, 1108–1118. [Google Scholar] [CrossRef] [PubMed]
  177. Laurent, J.M.; Young, J.H.; Kachroo, A.H.; Marcotte, E.M. Efforts to make and apply humanized yeast. Brief. Funct. Genom. 2016, 15, 155–163. [Google Scholar] [CrossRef]
  178. Wildt, S.; Gerngross, T.U. The humanization of N-glycosylation pathways in yeast. Nat. Rev. Microbiol. 2005, 3, 119–128. [Google Scholar] [CrossRef] [PubMed]
  179. Li, X.; Shen, J.; Chen, X.; Chen, L.; Wan, S.; Qiu, X.; Chen, K.; Chen, C.; Tan, H. Humanization of yeasts for glycan-type end-products. Front. Microbiol. 2022, 13, 930658. [Google Scholar] [CrossRef] [PubMed]
  180. Bobrowicz, P.; Davidson, R.C.; Li, H.; Potgieter, T.I.; Nett, J.H.; Hamilton, S.R.; Stadheim, T.A.; Miele, R.G.; Bobrowicz, B.; Mitchell, T. Engineering of an artificial glycosylation pathway blocked in core oligosaccharide assembly in the yeast Pichia pastoris: Production of complex humanized glycoproteins with terminal galactose. Glycobiology 2004, 14, 757–766. [Google Scholar] [CrossRef] [PubMed]
  181. Singh, M.B.; Bhalla, P.L. Recombinant expression systems for allergen vaccines. Inflamm. Allergy-Drug Targets (Formerly Curr. Drug Targets-Inflamm. Allergy) 2006, 5, 53–59. [Google Scholar] [CrossRef]
  182. Bernstein, M.B.; Chakraborty, M.; Wansley, E.K.; Guo, Z.; Franzusoff, A.; Mostböck, S.; Sabzevari, H.; Schlom, J.; Hodge, J.W. Recombinant Saccharomyces cerevisiae (yeast-CEA) as a potent activator of murine dendritic cells. Vaccine 2008, 26, 509–521. [Google Scholar] [CrossRef]
  183. Munson, S.; Parker, J.; King, T.H.; Lu, Y.; Kelley, V.; Guo, Z.; Borges, V.; Franzusoff, A. Coupling innate and adaptive immunity with yeast-based cancer immunotherapy. In Cancer Vaccines and Tumor Immunity; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2008; pp. 131–149. [Google Scholar]
  184. Remondo, C.; Cereda, V.; Mostböck, S.; Sabzevari, H.; Franzusoff, A.; Schlom, J.; Tsang, K.-Y. Human dendritic cell maturation and activation by a heat-killed recombinant yeast (Saccharomyces cerevisiae) vector encoding carcinoembryonic antigen. Vaccine 2009, 27, 987–994. [Google Scholar] [CrossRef] [PubMed]
  185. Ueno, K.; Okawara, A.; Yamagoe, S.; Naka, T.; Umeyama, T.; Utena-Abe, Y.; Tarumoto, N.; Niimi, M.; Ohno, H.; Doe, M.; et al. The mannan of Candida albicans lacking β-1,2-linked oligomannosides increases the production of inflammatory cytokines by dendritic cells. Med. Mycol. 2013, 51, 385–395. [Google Scholar] [CrossRef] [PubMed]
  186. Roohvand, F.; Shokri, M.; Abdollahpour-Alitappeh, M.; Ehsani, P. Biomedical applications of yeast- a patent view, part one: Yeasts as workhorses for the production of therapeutics and vaccines. Expert Opin. Ther. Pat. 2017, 27, 929–951. [Google Scholar] [CrossRef] [PubMed]
  187. Schellenbacher, C.; Roden, R.B.S.; Kirnbauer, R. Developments in L2-based human papillomavirus (HPV) vaccines. Virus Res. 2017, 231, 166–175. [Google Scholar] [CrossRef]
  188. Liu, C.; Yao, Y.; Yang, X.; Bai, H.; Huang, W.; Xia, Y.; Ma, Y. Production of Recombinant Human Papillomavirus Type 52 L1 Protein in Hansenula polymorpha Formed Virus-Like Particles. J. Microbiol. Biotechnol. 2015, 25, 936–940. [Google Scholar] [CrossRef]
  189. Caetano, K.A.A.; Del-Rios, N.H.A.; Pinheiro, R.S.; Bergamaschi, F.P.R.; Carneiro, M.A.D.S.; Teles, S.A. Low Immunogenicity of Recombinant Hepatitis B Vaccine Derived from Hansenula polymorpha in Adults Aged Over 40 Years. Am. J. Trop. Med. Hyg. 2017, 96, 118–121. [Google Scholar] [CrossRef] [PubMed]
  190. Xiao, Y.; Zhao, P.; Du, J.; Li, X.; Lu, W.; Hao, X.; Dong, B.; Yu, Y.; Wang, L. High-level expression and immunogenicity of porcine circovirus type 2b capsid protein without nuclear localization signal expressed in Hansenula polymorpha. Biologicals 2018, 51, 18–24. [Google Scholar] [CrossRef] [PubMed]
  191. Ramasamy, V.; Arora, U.; Shukla, R.; Poddar, A.; Shanmugam, R.K.; White, L.J.; Mattocks, M.M.; Raut, R.; Perween, A.; Tyagi, P.; et al. A tetravalent virus-like particle vaccine designed to display domain III of dengue envelope proteins induces multi-serotype neutralizing antibodies in mice and macaques which confer protection against antibody dependent enhancement in AG129 mice. PLoS Negl. Trop. Dis. 2018, 12, e0006191. [Google Scholar] [CrossRef] [PubMed]
  192. Rajpoot, R.K.; Shukla, R.; Arora, U.; Swaminathan, S.; Khanna, N. Dengue envelope-based “four-in-one” virus-like particles produced using Pichia pastoris induce enhancement-lacking, domain III-directed tetravalent neutralising antibodies in mice. Sci. Rep. 2018, 8, 8643. [Google Scholar] [CrossRef] [PubMed]
  193. Liu, Y.; Zhou, J.; Yu, Z.; Fang, D.; Fu, C.; Zhu, X.; He, Z.; Yan, H.; Jiang, L. Tetravalent recombinant dengue virus-like particles as potential vaccine candidates: Immunological properties. BMC Microbiol. 2014, 14, 233. [Google Scholar] [CrossRef] [PubMed]
  194. Poddar, A.; Ramasamy, V.; Shukla, R.; Rajpoot, R.K.; Arora, U.; Jain, S.K.; Swaminathan, S.; Khanna, N. Virus-like particles derived from Pichia pastoris-expressed dengue virus type 1 glycoprotein elicit homotypic virus-neutralizing envelope domain III-directed antibodies. BMC Biotechnol. 2016, 16, 50. [Google Scholar] [CrossRef]
  195. Khetarpal, N.; Shukla, R.; Rajpoot, R.K.; Poddar, A.; Pal, M.; Swaminathan, S.; Arora, U.; Khanna, N. Recombinant Dengue Virus 4 Envelope Glycoprotein Virus-Like Particles Derived from Pichia pastoris are Capable of Eliciting Homotypic Domain III-Directed Neutralizing Antibodies. Am. J. Trop. Med. Hyg. 2017, 96, 126–134. [Google Scholar] [CrossRef]
  196. Zhang, C.; Zhang, X.; Zhang, W.; Dai, W.; Xie, J.; Ye, L.; Wang, H.; Chen, H.; Liu, Q.; Gong, S.; et al. Enterovirus D68 virus-like particles expressed in Pichia pastoris potently induce neutralizing antibody responses and confer protection against lethal viral infection in mice. Emerg. Microbes Infect. 2018, 7, 3. [Google Scholar] [CrossRef]
  197. Zahid, M.; Lünsdorf, H.; Rinas, U. Assessing stability and assembly of the hepatitis B surface antigen into virus-like particles during down-stream processing. Vaccine 2015, 33, 3739–3745. [Google Scholar] [CrossRef]
  198. Fazlalipour, M.; Keyvani, H.; Monavari, S.H.R.; Mollaie, H.R. Expression, Purification and Immunogenic Description of a Hepatitis C Virus Recombinant CoreE1E2 Protein Expressed by Yeast Pichia pastoris. Jundishapur J. Microbiol. 2015, 8, e17157. [Google Scholar] [CrossRef] [PubMed]
  199. Bredell, H.; Smith, J.J.; Görgens, J.F.; van Zyl, W.H. Expression of unique chimeric human papilloma virus type 16 (HPV-16) L1-L2 proteins in Pichia pastoris and Hansenula polymorpha. Yeast 2018, 35, 519–529. [Google Scholar] [CrossRef] [PubMed]
  200. Gupta, G.; Glueck, R.; Rishi, N. Physicochemical characterization and immunological properties of Pichia pastoris based HPV16L1 and 18L1 virus like particles. Biologicals 2017, 46, 11–22. [Google Scholar] [CrossRef] [PubMed]
  201. Athmaram, T.N.; Singh, A.K.; Saraswat, S.; Srivastava, S.; Misra, P.; Kameswara Rao, M.; Gopalan, N.; Rao, P.V.L. A simple Pichia pastoris fermentation and downstream processing strategy for making recombinant pandemic Swine Origin Influenza a virus Hemagglutinin protein. J. Ind. Microbiol. Biotechnol. 2013, 40, 245–255. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, S.-C.; Liao, H.-Y.; Zhang, J.-Y.; Cheng, T.-J.R.; Wong, C.-H. Development of a universal influenza vaccine using hemagglutinin stem protein produced from Pichia pastoris. Virology 2019, 526, 125–137. [Google Scholar] [CrossRef]
  203. Pietrzak, M.; Macioła, A.; Zdanowski, K.; Protas-Klukowska, A.M.; Olszewska, M.; Śmietanka, K.; Minta, Z.; Szewczyk, B.; Kopera, E. An avian influenza H5N1 virus vaccine candidate based on the extracellular domain produced in yeast system as subviral particles protects chickens from lethal challenge. Antivir. Res. 2016, 133, 242–249. [Google Scholar] [CrossRef]
  204. Cheng, C.-Y.; Wu, C.-W.; Lin, G.-J.; Lee, W.-C.; Chien, M.-S.; Huang, C. Enhancing expression of the classical swine fever virus glycoprotein E2 in yeast and its application to a blocking ELISA. J. Biotechnol. 2014, 174, 1–6. [Google Scholar] [CrossRef]
  205. Lin, G.-J.; Deng, M.-C.; Chen, Z.-W.; Liu, T.-Y.; Wu, C.-W.; Cheng, C.-Y.; Chien, M.-S.; Huang, C. Yeast expressed classical swine fever E2 subunit vaccine candidate provides complete protection against lethal challenge infection and prevents horizontal virus transmission. Vaccine 2012, 30, 2336–2341. [Google Scholar] [CrossRef]
  206. Saraswat, S.; Athmaram, T.N.; Parida, M.; Agarwal, A.; Saha, A.; Dash, P.K. Expression and Characterization of Yeast Derived Chikungunya Virus Like Particles (CHIK-VLPs) and Its Evaluation as a Potential Vaccine Candidate. PLoS Negl. Trop. Dis. 2016, 10, e0004782. [Google Scholar] [CrossRef] [PubMed]
  207. Zang, J.; Zhu, Y.; Zhou, Y.; Gu, C.; Yi, Y.; Wang, S.; Xu, S.; Hu, G.; Du, S.; Yin, Y.; et al. Yeast-produced RBD-based recombinant protein vaccines elicit broadly neutralizing antibodies and durable protective immunity against SARS-CoV-2 infection. Cell Discov. 2021, 7, 71. [Google Scholar] [CrossRef]
  208. Kim, H.J.; Lee, J.Y.; Kang, H.A.; Lee, Y.; Park, E.-J.; Kim, H.-J. Oral immunization with whole yeast producing viral capsid antigen provokes a stronger humoral immune response than purified viral capsid antigen. Lett. Appl. Microbiol. 2014, 58, 285–291. [Google Scholar] [CrossRef] [PubMed]
  209. Hadiji-Abbes, N.; Martin, M.; Benzina, W.; Karray-Hakim, H.; Gergely, C.; Gargouri, A.; Mokdad-Gargouri, R. Extraction and purification of hepatitis B virus-like M particles from a recombinant Saccharomyces cerevisiae strain using alumina powder. J. Virol. Methods 2013, 187, 132–137. [Google Scholar] [CrossRef]
  210. Pleckaityte, M.; Bremer, C.M.; Gedvilaite, A.; Kucinskaite-Kodze, I.; Glebe, D.; Zvirbliene, A. Construction of polyomavirus-derived pseudotype virus-like particles displaying a functionally active neutralizing antibody against hepatitis B virus surface antigen. BMC Biotechnol. 2015, 15, 85. [Google Scholar] [CrossRef] [PubMed]
  211. King, T.H.; Kemmler, C.B.; Guo, Z.; Mann, D.; Lu, Y.; Coeshott, C.; Gehring, A.J.; Bertoletti, A.; Ho, Z.Z.; Delaney, W.; et al. A whole recombinant yeast-based therapeutic vaccine elicits HBV X, S and Core specific T cells in mice and activates human T cells recognizing epitopes linked to viral clearance. PLoS ONE 2014, 9, e101904. [Google Scholar] [CrossRef] [PubMed]
  212. Zhang, L.; Liu, J.; Lu, J.; Yan, B.; Song, L.; Li, L.; Cui, F.; Zhang, G.; Wang, F.; Liang, X. Antibody response to revaccination among adult non-responders to primary Hepatitis B vaccination in China. Hum. Vaccines Immunother. 2015, 11, 2716–2722. [Google Scholar] [CrossRef] [PubMed]
  213. Gaggar, A.; Coeshott, C.; Apelian, D.; Rodell, T.; Armstrong, B.R.; Shen, G.; Subramanian, G.M.; McHutchison, J.G. Safety, tolerability and immunogenicity of GS-4774, a hepatitis B virus-specific therapeutic vaccine, in healthy subjects: A randomized study. Vaccine 2014, 32, 4925–4931. [Google Scholar] [CrossRef]
  214. Lin, T.; Xianyu, L.; Lyu, S. Monoclonal neutralizing antibodies against EV71 screened from mice immunized with yeast-produced virus-like particles. Virol. Sin. 2015, 30, 208–213. [Google Scholar] [CrossRef] [PubMed]
  215. Wang, X.; Xiao, X.; Zhao, M.; Liu, W.; Pang, L.; Sun, X.; Cen, S.; Yang, B.B.; Huang, Y.; Sheng, W. EV71 virus-like particles produced by co-expression of capsid proteins in yeast cells elicit humoral protective response against EV71 lethal challenge. BMC Res. Notes 2016, 9, 42. [Google Scholar] [CrossRef] [PubMed]
  216. Penkert, R.R.; Young, N.S.; Surman, S.L.; Sealy, R.E.; Rosch, J.; Dormitzer, P.R.; Settembre, E.C.; Chandramouli, S.; Wong, S.; Hankins, J.S.; et al. Saccharomyces cerevisiae-derived virus-like particle parvovirus B19 vaccine elicits binding and neutralizing antibodies in a mouse model for sickle cell disease. Vaccine 2017, 35, 3615–3620. [Google Scholar] [CrossRef] [PubMed]
  217. Chandramouli, S.; Medina-Selby, A.; Coit, D.; Schaefer, M.; Spencer, T.; Brito, L.A.; Zhang, P.; Otten, G.; Mandl, C.W.; Mason, P.W.; et al. Generation of a parvovirus B19 vaccine candidate. Vaccine 2013, 31, 3872–3878. [Google Scholar] [CrossRef] [PubMed]
  218. Nguyen, N.-L.; Kim, J.-M.; Park, J.-A.; Park, S.-M.; Jang, Y.-S.; Yang, M.-S.; Kim, D.-H. Expression and purification of an immunogenic dengue virus epitope using a synthetic consensus sequence of envelope domain III and Saccharomyces cerevisiae. Protein Expr. Purif. 2013, 88, 235–242. [Google Scholar] [CrossRef] [PubMed]
  219. Wang, H.; Chen, X.; Wang, D.; Yao, C.; Wang, Q.; Xie, J.; Shi, X.; Xiang, Y.; Liu, W.; Zhang, L. Epitope-focused immunogens against the CD4-binding site of HIV-1 envelope protein induce neutralizing antibodies against auto-and heterologous viruses. J. Biol. Chem. 2018, 293, 830–846. [Google Scholar] [CrossRef]
  220. Sablon, E.; Van Broekhoven, A.; Bosman, F.; Depla, E.; Deschamps, G. Constructs and Methods for Expression of Recombinant HCV Envelope Proteins. U.S. Patent 7,314,925, 1 January 2008. [Google Scholar]
  221. Evans, J.D.; Beatty, N.M. Yeast Expression of Flavivirus Virus-Like Particles and Use Thereof. U.S. Patent 4/241,530, 7 August 2014. [Google Scholar]
  222. Liu, Y.; Zhao, D.; Wang, Y.; Chen, Z.; Yang, L.; Li, W.; Gong, Y.; Gan, C.; Tang, J.; Zhang, T. A vaccine based on the yeast-expressed receptor-binding domain (RBD) elicits broad immune responses against SARS-CoV-2 variants. Front. Immunol. 2022, 13, 1011484. [Google Scholar] [CrossRef] [PubMed]
  223. Pino, M.; Abid, T.; Pereira Ribeiro, S.; Edara, V.V.; Floyd, K.; Smith, J.C.; Latif, M.B.; Pacheco-Sanchez, G.; Dutta, D.; Wang, S.; et al. A yeast-expressed RBD-based SARS-CoV-2 vaccine formulated with 3M-052-alum adjuvant promotes protective efficacy in non-human primates. Sci. Immunol. 2021, 6, eabh3634. [Google Scholar] [CrossRef] [PubMed]
  224. Pollet, J.; Chen, W.-H.; Versteeg, L.; Keegan, B.; Zhan, B.; Wei, J.; Liu, Z.; Lee, J.; Kundu, R.; Adhikari, R.; et al. SARS-CoV-2 RBD219-N1C1: A yeast-expressed SARS-CoV-2 recombinant receptor-binding domain candidate vaccine stimulates virus neutralizing antibodies and T-cell immunity in mice. Hum. Vaccines Immunother. 2021, 17, 2356–2366. [Google Scholar] [CrossRef]
  225. Chen, W.-H.; Pollet, J.; Strych, U.; Lee, J.; Liu, Z.; Kundu, R.T.; Versteeg, L.; Villar, M.J.; Adhikari, R.; Wei, J.; et al. Yeast-expressed recombinant SARS-CoV-2 receptor binding domain RBD203-N1 as a COVID-19 protein vaccine candidate. Protein Expr. Purif. 2022, 190, 106003. [Google Scholar] [CrossRef] [PubMed]
  226. Chen, W.-H.; Du, L.; Chag, S.M.; Ma, C.; Tricoche, N.; Tao, X.; Seid, C.A.; Hudspeth, E.M.; Lustigman, S.; Tseng, C.-T.K.; et al. Yeast-expressed recombinant protein of the receptor-binding domain in SARS-CoV spike protein with deglycosylated forms as a SARS vaccine candidate. Hum. Vaccines Immunother. 2014, 10, 648–658. [Google Scholar] [CrossRef]
  227. Dai, L.; Zheng, T.; Xu, K.; Han, Y.; Xu, L.; Huang, E.; An, Y.; Cheng, Y.; Li, S.; Liu, M. A universal design of betacoronavirus vaccines against COVID-19, MERS, and SARS. Cell 2020, 182, 722–733. [Google Scholar] [CrossRef]
  228. Thak, E.J.; Kim, J.; Lee, D.-J.; Kim, J.Y.; Kang, H.A. Structural analysis of N-/O-glycans assembled on proteins in yeasts. J. Microbiol. 2018, 56, 11–23. [Google Scholar] [CrossRef] [PubMed]
  229. Hamilton, S.R.; Cook, W.J.; Gomathinayagam, S.; Burnina, I.; Bukowski, J.; Hopkins, D.; Schwartz, S.; Du, M.; Sharkey, N.J.; Bobrowicz, P. Production of sialylated O-linked glycans in Pichia pastoris. Glycobiology 2013, 23, 1192–1203. [Google Scholar] [CrossRef]
  230. Shajahan, A.; Supekar, N.T.; Gleinich, A.S.; Azadi, P. Deducing the N-and O-glycosylation profile of the spike protein of novel coronavirus SARS-CoV-2. Glycobiology 2020, 30, 981–988. [Google Scholar] [CrossRef] [PubMed]
  231. Li, H.-Y.; Han, J.-F.; Qin, C.-F.; Chen, R. Virus-like particles for enterovirus 71 produced from Saccharomyces cerevisiae potently elicits protective immune responses in mice. Vaccine 2013, 31, 3281–3287. [Google Scholar] [CrossRef]
  232. Zhao, H.; Li, H.-Y.; Han, J.-F.; Deng, Y.-Q.; Li, Y.-X.; Zhu, S.-Y.; He, Y.-L.; Qin, E.-D.; Chen, R.; Qin, C.-F. Virus-like particles produced in Saccharomyces cerevisiae elicit protective immunity against Coxsackievirus A16 in mice. Appl. Microbiol. Biotechnol. 2013, 97, 10445–10452. [Google Scholar] [CrossRef] [PubMed]
  233. Bill, R.M. Recombinant protein subunit vaccine synthesis in microbes: A role for yeast? J. Pharm. Pharmacol. 2015, 67, 319–328. [Google Scholar] [CrossRef] [PubMed]
  234. Rosales-Mendoza, S.; Paz-Maldonado, L.M.T.; Soria-Guerra, R.E. Chlamydomonas reinhardtii as a viable platform for the production of recombinant proteins: Current status and perspectives. Plant Cell Rep. 2012, 31, 479–494. [Google Scholar] [CrossRef] [PubMed]
  235. Lauersen, K.J.; Kruse, O.; Berger, H.; Mussgnug, J.H. Efficient recombinant protein production and secretion from nuclear transgenes in Chlamydomonas reinhardtii. J. Biotechnol. 2013, 167, 101–110. [Google Scholar] [CrossRef] [PubMed]
  236. Gutiérrez, S.; Lauersen, K.J. Gene delivery technologies with applications in microalgal genetic engineering. Biology 2021, 10, 265. [Google Scholar] [CrossRef] [PubMed]
  237. Lanigan, T.M.; Kopera, H.C.; Saunders, T.L. Principles of genetic engineering. Genes 2020, 11, 291. [Google Scholar] [CrossRef] [PubMed]
  238. Zhou, H.-C.; Long, J.R.; Yaghi, O.M. Introduction to metal–organic frameworks. Chem. Rev. 2012, 112, 673–674. [Google Scholar] [CrossRef] [PubMed]
  239. Silva, S.; Almeida, A.J.; Vale, N. Combination of cell-penetrating peptides with nanoparticles for therapeutic application: A review. Biomolecules 2019, 9, 22. [Google Scholar] [CrossRef]
  240. Gregory, J.A.; Topol, A.B.; Doerner, D.Z.; Mayfield, S. Alga-Produced Cholera Toxin-Pfs25 Fusion Proteins as Oral Vaccines. Appl. Environ. Microbiol. 2013, 79, 3917–3925. [Google Scholar] [CrossRef]
  241. Dreesen, I.A.J.; Hamri, G.C.-E.; Fussenegger, M. Heat-stable oral alga-based vaccine protects mice from Staphylococcus aureus infection. J. Biotechnol. 2010, 145, 273–280. [Google Scholar] [CrossRef] [PubMed]
  242. Specht, E.; Mayfield, S. Algae-based oral recombinant vaccines. Front. Microbiol. 2014, 5, 60. Available online: https://www.frontiersin.org/article/10.3389/fmicb.2014.00060 (accessed on 1 June 2024). [CrossRef] [PubMed]
  243. Lössl, A.G.; Waheed, M.T. Chloroplast-derived vaccines against human diseases: Achievements, challenges and scopes. Plant Biotechnol. J. 2011, 9, 527–539. [Google Scholar] [CrossRef] [PubMed]
  244. Franklin, S.; Ngo, B.; Efuet, E.; Mayfield, S.P. Development of a GFP reporter gene for Chlamydomonas reinhardtii chloroplast. Plant J. 2002, 30, 733–744. [Google Scholar] [CrossRef] [PubMed]
  245. Mayfield, S.P.; Franklin, S.E.; Lerner, R.A. Expression and assembly of a fully active antibody in algae. Proc. Natl. Acad. Sci. USA 2003, 100, 438–442. [Google Scholar] [CrossRef]
  246. Butinar, L.; Sonjak, S.; Zalar, P.; Plemenitaš, A.; Gunde-Cimerman, N. Melanized halophilic fungi are eukaryotic members of microbial communities in hypersaline waters of solar salterns. Bot. Mar. 2005, 48, 73–79. [Google Scholar] [CrossRef]
  247. Ben-Amotz, A.; Avron, M. The biotechnology of cultivating the halotolerant algaDunaliella. Trends Biotechnol. 1990, 8, 121–126. [Google Scholar] [CrossRef]
  248. Qin, S.; Lin, H.; Jiang, P. Advances in genetic engineering of marine algae. Biotechnol. Adv. 2012, 30, 1602–1613. [Google Scholar] [CrossRef]
  249. Georgianna, D.R.; Mayfield, S.P. Exploiting diversity and synthetic biology for the production of algal biofuels. Nature 2012, 488, 329–335. [Google Scholar] [CrossRef]
  250. Ducat, D.C.; Way, J.C.; Silver, P.A. Engineering cyanobacteria to generate high-value products. Trends Biotechnol. 2011, 29, 95–103. [Google Scholar] [CrossRef] [PubMed]
  251. Gong, Y.; Hu, H.; Gao, Y.; Xu, X.; Gao, H. Microalgae as platforms for production of recombinant proteins and valuable compounds: Progress and prospects. J. Ind. Microbiol. Biotechnol. 2011, 38, 1879–1890. [Google Scholar] [CrossRef]
  252. Berndt, A.J.; Smalley, T.N.; Ren, B.; Simkovsky, R.; Badary, A.; Sproles, A.E.; Fields, F.J.; Torres-Tiji, Y.; Heredia, V.; Mayfield, S.P. Recombinant production of a functional SARS-CoV-2 spike receptor binding domain in the green algae Chlamydomonas reinhardtii. PLoS ONE 2021, 16, e0257089. [Google Scholar] [CrossRef] [PubMed]
  253. Govea-Alonso, D.O.; Malla, A.; Bolaños-Martínez, O.C.; Vimolmangkang, S.; Rosales-Mendoza, S. An Algae-Made RBD from SARS-CoV-2 Is Immunogenic in Mice. Pharmaceuticals 2022, 15, 1298. [Google Scholar] [CrossRef]
  254. 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] [PubMed]
  255. Slattery, S.S.; Giguere, D.J.; Stuckless, E.E.; Shrestha, A.; Briere, L.-A.K.; Galbraith, A.; Reaume, S.; Boyko, X.; Say, H.H.; Browne, T.S.; et al. Phosphate-regulated expression of the SARS-CoV-2 receptor-binding domain in the diatom Phaeodactylum tricornutum for pandemic diagnostics. Sci. Rep. 2022, 12, 7010. [Google Scholar] [CrossRef] [PubMed]
  256. Gimpel, J.A.; Hyun, J.S.; Schoepp, N.G.; Mayfield, S.P. Production of recombinant proteins in microalgae at pilot greenhouse scale. Biotechnol. Bioeng. 2015, 112, 339–345. [Google Scholar] [CrossRef] [PubMed]
  257. Rattanapisit, K.; Shanmugaraj, B.; Manopwisedjaroen, S.; Purwono, P.B.; Siriwattananon, K.; Khorattanakulchai, N.; Hanittinan, O.; Boonyayothin, W.; Thitithanyanont, A.; Smith, D.R. 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] [PubMed]
  258. Yusibov, V.; Rabindran, S. Recent progress in the development of plant derived vaccines. Expert Rev. Vaccines 2008, 7, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
  259. Yusibov, V.; Streatfield, S.J.; Kushnir, N. Clinical development of plant-produced recombinant pharmaceuticals: Vaccines, antibodies and beyond. Hum. Vaccines 2011, 7, 313–321. [Google Scholar] [CrossRef] [PubMed]
  260. Egelkrout, E.; Hayden, C.; Fake, G.; Keener, T.; Arruda, P.; Saltzman, R.; Walker, J.; Howard, J. Oral delivery of maize-produced porcine epidemic diarrhea virus spike protein elicits neutralizing antibodies in pigs. Plant Cell. Tissue Organ Cult. 2020, 142, 79–86. [Google Scholar] [CrossRef] [PubMed]
  261. Hayden, C.A.; Egelkrout, E.M.; Moscoso, A.M.; Enrique, C.; Keener, T.K.; Jimenez-Flores, R.; Wong, J.C.; Howard, J.A. Production of highly concentrated, heat-stable hepatitis B surface antigen in maize. Plant Biotechnol. J. 2012, 10, 979–984. [Google Scholar] [CrossRef] [PubMed]
  262. Hayden, C.A.; Smith, E.M.; Turner, D.D.; Keener, T.K.; Wong, J.C.; Walker, J.H.; Tizard, I.R.; Jimenez-Flores, R.; Howard, J.A. Supercritical fluid extraction provides an enhancement to the immune response for orally-delivered hepatitis B surface antigen. Vaccine 2014, 32, 1240–1246. [Google Scholar] [CrossRef]
  263. Shahid, N.; Daniell, H. Plant-based oral vaccines against zoonotic and non-zoonotic diseases. Plant Biotechnol. J. 2016, 14, 2079–2099. [Google Scholar] [CrossRef]
  264. Gehl, C.; Li, G.; Serek, M. An efficient protocol for Agrobacterium-mediated transformation and regeneration of Campanula medium (Canterbury bells) based on leaf disc explants. Plant Cell Tissue Organ Cult. 2020, 140, 635–645. [Google Scholar] [CrossRef]
  265. Ariga, H.; Toki, S.; Ishibashi, K. Potato virus X vector-mediated DNA-free genome editing in plants. Plant Cell Physiol. 2020, 61, 1946–1953. [Google Scholar] [CrossRef]
  266. Santos, R.B.; Abranches, R.; Fischer, R.; Sack, M.; Holland, T. Putting the spotlight back on plant suspension cultures. Front. Plant Sci. 2016, 7, 297. [Google Scholar] [CrossRef]
  267. Kolotilin, I.; Topp, E.; Cox, E.; Devriendt, B.; Conrad, U.; Joensuu, J.; Stöger, E.; Warzecha, H.; McAllister, T.; Potter, A.; et al. Plant-based solutions for veterinary immunotherapeutics and prophylactics. Vet. Res. 2014, 45, 117. [Google Scholar] [CrossRef] [PubMed]
  268. Floss, D.M.; Falkenburg, D.; Conrad, U. Production of vaccines and therapeutic antibodies for veterinary applications in transgenic plants: An overview. Transgenic Res. 2007, 16, 315–332. [Google Scholar] [CrossRef]
  269. Oh, Y.; Park, Y.; Choi, B.-H.; Park, S.; Gu, S.; Park, J.; Kim, J.-K.; Sohn, E.-J. Field Application of a New CSF Vaccine Based on Plant-Produced Recombinant E2 Marker Proteins on Pigs in Areas with Two Different Control Strategies. Vaccines 2021, 9, 537. [Google Scholar] [CrossRef]
  270. Landry, N.; Pillet, S.; Favre, D.; Poulin, J.-F.; Trépanier, S.; Yassine-Diab, B.; Ward, B.J. Influenza virus-like particle vaccines made in Nicotiana benthamiana elicit durable, poly-functional and cross-reactive T cell responses to influenza HA antigens. Clin. Immunol. 2014, 154, 164–177. [Google Scholar] [CrossRef] [PubMed]
  271. Pillet, S.; Racine, T.; Nfon, C.; Di Lenardo, T.Z.; Babiuk, S.; Ward, B.J.; Kobinger, G.P.; Landry, N. Plant-derived H7 VLP vaccine elicits protective immune response against H7N9 influenza virus in mice and ferrets. Vaccine 2015, 33, 6282–6289. [Google Scholar] [CrossRef] [PubMed]
  272. Shoji, Y.; Chichester, J.A.; Jones, M.; Manceva, S.D.; Damon, E.; Mett, V.; Musiychuk, K.; Bi, H.; Farrance, C.; Shamloul, M.; et al. Plant-based rapid production of recombinant subunit hemagglutinin vaccines targeting H1N1 and H5N1 influenza. Hum. Vaccines 2011, 7 (Suppl. 1), 41–50. [Google Scholar] [CrossRef] [PubMed]
  273. Smith, T.; O’Kennedy, M.M.; Wandrag, D.B.R.; Adeyemi, M.; Abolnik, C. Efficacy of a plant-produced virus-like particle vaccine in chickens challenged with Influenza A H6N2 virus. Plant Biotechnol. J. 2020, 18, 502–512. [Google Scholar] [CrossRef] [PubMed]
  274. Sepotokele, K.M.; O’Kennedy, M.M.; Wandrag, D.B.R.; Abolnik, C. Optimization of infectious bronchitis virus-like particle expression in Nicotiana benthamiana as potential poultry vaccines. PLoS ONE 2023, 18, e0288970. [Google Scholar] [CrossRef] [PubMed]
  275. Siriwattananon, K.; Manopwisedjaroen, S.; Shanmugaraj, B.; Rattanapisit, K.; Phumiamorn, S.; Sapsutthipas, S.; Trisiriwanich, S.; Prompetchara, E.; Ketloy, C.; Buranapraditkun, S. Plant-produced receptor-binding domain of SARS-CoV-2 elicits potent neutralizing responses in mice and non-human primates. Front. Plant Sci. 2021, 12, 682953. [Google Scholar] [CrossRef] [PubMed]
  276. Park, Y.; Lee, S.; Kang, H.; Park, M.; Min, K.; Kim, N.H.; Gu, S.; Kim, J.K.; An, D.-J.; Choe, S. A classical swine fever virus E2 fusion protein produced in plants elicits a neutralizing humoral immune response in mice and pigs. Biotechnol. Lett. 2020, 42, 1247–1261. [Google Scholar] [CrossRef]
  277. 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]
  278. Garabagi, F.; McLean, M.D.; Hall, J.C. Transient and stable expression of antibodies in Nicotiana species. Methods Mol. Biol. 2012, 907, 389–408. [Google Scholar] [CrossRef] [PubMed]
  279. Darveau, R.P.; Chilton, P.M. Naturally occurring low biological reactivity lipopolysaccharides as vaccine adjuvants. Expert Rev. Vaccines 2013, 12, 707–709. [Google Scholar] [CrossRef] [PubMed]
  280. Global Biodefense Staff. DARPA Hits Milestone in Plant-Based Vaccines for Pandemics. Glob. Biodefense. Available online: https://globalbiodefense.com/2012/07/28/darpa-program-hits-milestone-in-plant-based-vaccines-for-pandemics/ (accessed on 3 June 2020).
  281. Potera, C. Vaccine Manufacturing Gets Boost from Tobacco Plants. Genet. Eng. Biotechnol. News 2012, 32, 8–10. [Google Scholar] [CrossRef]
  282. Shin, Y.-J.; König-Beihammer, J.; Vavra, U.; Schwestka, J.; Kienzl, N.F.; Klausberger, M.; Laurent, E.; Grünwald-Gruber, C.; Vierlinger, K.; Hofner, M. N-glycosylation of the SARS-CoV-2 receptor binding domain is important for functional expression in plants. Front. Plant Sci. 2021, 12, 689104. [Google Scholar] [CrossRef] [PubMed]
  283. Makatsa, M.S.; Tincho, M.B.; Wendoh, J.M.; Ismail, S.D.; Nesamari, R.; Pera, F.; De Beer, S.; David, A.; Jugwanth, S.; Gededzha, M.P. SARS-CoV-2 antigens expressed in plants detect antibody responses in COVID-19 patients. Front. Plant Sci. 2021, 12, 589940. [Google Scholar] [CrossRef]
  284. Jung, J.; Zahmanova, G.; Minkov, I.; Lomonossoff, G.P. Plant-based expression and characterization of SARS-CoV-2 virus-like particles presenting a native spike protein. Plant Biotechnol. J. 2022, 20, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  285. Capell, T.; Twyman, R.M.; Armario-Najera, V.; Ma, J.K.-C.; Schillberg, S.; Christou, P. Potential applications of plant biotechnology against SARS-CoV-2. Trends Plant Sci. 2020, 25, 635–643. [Google Scholar] [CrossRef] [PubMed]
  286. Maharjan, P.M.; Cheon, J.; Jung, J.; Kim, H.; Lee, J.; Song, M.; Jeong, G.U.; Kwon, Y.; Shim, B.; Choe, S. Plant-expressed receptor binding domain of the SARS-CoV-2 spike protein elicits Humoral immunity in mice. Vaccines 2021, 9, 978. [Google Scholar] [CrossRef]
  287. Moon, K.-B.; Jeon, J.-H.; Choi, H.; Park, J.-S.; Park, S.-J.; Lee, H.-J.; Park, J.M.; Cho, H.S.; Moon, J.S.; Oh, H. Construction of SARS-CoV-2 virus-like particles in plant. Sci. Rep. 2022, 12, 1005. [Google Scholar] [CrossRef] [PubMed]
  288. Gregorio, N.E.; Levine, M.Z.; Oza, J.P. A User’s Guide to Cell-Free Protein Synthesis. Methods Protoc. 2019, 2, 24. [Google Scholar] [CrossRef] [PubMed]
  289. Jin, X.; Hong, S.H. Cell-free protein synthesis for producing ‘difficult-to-express’ proteins. Biochem. Eng. J. 2018, 138, 156–164. [Google Scholar] [CrossRef]
  290. Rosenblum, G.; Cooperman, B.S. Engine out of the chassis: Cell-free protein synthesis and its uses. FEBS Lett. 2014, 588, 261–268. [Google Scholar] [CrossRef] [PubMed]
  291. Bretthauer, R.K.; Marcus, L.; Chaloupka, J.; Halvorson, H.O.; Bock, R.M. Amino acid incorporation into protein by cell-free extracts of yeast. Biochemistry 1963, 2, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  292. Zubay, G. In Vitro Synthesis of Protein in Microbial Systems. Annu. Rev. Genet. 1973, 7, 267–287. [Google Scholar] [CrossRef]
  293. Shin, J.; Noireaux, V. An E. coli Cell-Free Expression Toolbox: Application to Synthetic Gene Circuits and Artificial Cells. ACS Synth. Biol. 2012, 1, 29–41. [Google Scholar] [CrossRef] [PubMed]
  294. Chan, P.; Thomas, C.J.; Sprang, S.R.; Tall, G.G. Molecular chaperoning function of Ric-8 is to fold nascent heterotrimeric G protein α subunits. Proc. Natl. Acad. Sci. USA 2013, 110, 3794–3799. [Google Scholar] [CrossRef]
  295. Li, J.; Gu, L.; Aach, J.; Church, G.M. Improved Cell-Free RNA and Protein Synthesis System. PLoS ONE 2014, 9, e106232. [Google Scholar] [CrossRef] [PubMed]
  296. Tinafar, A.; Jaenes, K.; Pardee, K. Synthetic Biology Goes Cell-Free. BMC Biol. 2019, 17, 64. [Google Scholar] [CrossRef] [PubMed]
  297. Smith, M.T.; Berkheimer, S.D.; Werner, C.J.; Bundy, B.C. Lyophilized Escherichia coli-based cell-free systems for robust, high-density, long-term storage. Biotechniques 2014, 56, 186–193. [Google Scholar] [CrossRef] [PubMed]
  298. Pardee, K.; Slomovic, S.; Nguyen, P.Q.; Lee, J.W.; Donghia, N.; Burrill, D.; Ferrante, T.; McSorley, F.R.; Furuta, Y.; Vernet, A.; et al. Portable, On-Demand Biomolecular Manufacturing. Cell 2016, 167, 248–259.e12. [Google Scholar] [CrossRef]
  299. Stech, M.; Quast, R.B.; Sachse, R.; Schulze, C.; Wüstenhagen, D.A.; Kubick, S. A Continuous-Exchange Cell-Free Protein Synthesis System Based on Extracts from Cultured Insect Cells. PLoS ONE 2014, 9, e96635. [Google Scholar] [CrossRef] [PubMed]
  300. Chen, Y.; Emileh, A.; Brideau, N.; Britton, J. Cell-free reactions in continuous manufacturing systems. Curr. Opin. Green Sustain. Chem. 2020, 25, 100380. [Google Scholar] [CrossRef]
  301. Quast, R.B.; Kortt, O.; Henkel, J.; Dondapati, S.K.; Wüstenhagen, D.A.; Stech, M.; Kubick, S. Automated production of functional membrane proteins using eukaryotic cell-free translation systems. J. Biotechnol. 2015, 203, 45–53. [Google Scholar] [CrossRef] [PubMed]
  302. Silverman, A.D.; Karim, A.S.; Jewett, M.C. Cell-free gene expression: An expanded repertoire of applications. Nat. Rev. Genet. 2020, 21, 151–170. [Google Scholar] [CrossRef] [PubMed]
  303. Dopp, B.J.L.; Tamiev, D.D.; Reuel, N.F. Cell-free supplement mixtures: Elucidating the history and biochemical utility of additives used to support in vitro protein synthesis in E. coli extract. Biotechnol. Adv. 2019, 37, 246–258. [Google Scholar] [CrossRef] [PubMed]
  304. Carlson, E.D.; Gan, R.; Hodgman, C.E.; Jewett, M.C. Cell-free protein synthesis: Applications come of age. Biotechnol. Adv. 2012, 30, 1185–1194. [Google Scholar] [CrossRef]
  305. Chiao, A.C.; Murray, R.M.; Sun, Z.Z. Development of prokaryotic cell-free systems for synthetic biology. bioRxiv 2016, 48710. [Google Scholar] [CrossRef]
  306. Yang, W.C.; Patel, K.G.; Wong, H.E.; Swartz, J.R. Simplifying and streamlining Escherichia coli-based cell-free protein synthesis. Biotechnol. Prog. 2012, 28, 413–420. [Google Scholar] [CrossRef] [PubMed]
  307. Caschera, F.; Noireaux, V. Synthesis of 2.3 mg/ml of protein with an all Escherichia coli cell-free transcription–translation system. Biochimie 2014, 99, 162–168. [Google Scholar] [CrossRef]
  308. Des Soye, B.J.; Davidson, S.R.; Weinstock, M.T.; Gibson, D.G.; Jewett, M.C. Establishing a High-Yielding Cell-Free Protein Synthesis Platform Derived from Vibrio natriegens. ACS Synth. Biol. 2018, 7, 2245–2255. [Google Scholar] [CrossRef] [PubMed]
  309. Xu, H.; Liu, W.-Q.; Li, J. Translation Related Factors Improve the Productivity of a Streptomyces-Based Cell-Free Protein Synthesis System. ACS Synth. Biol. 2020, 9, 1221–1224. [Google Scholar] [CrossRef] [PubMed]
  310. Kelwick, R.; Webb, A.J.; MacDonald, J.T.; Freemont, P.S. Development of a Bacillus subtilis cell-free transcription-translation system for prototyping regulatory elements. Metab. Eng. 2016, 38, 370–381. [Google Scholar] [CrossRef] [PubMed]
  311. Moore, S.J.; MacDonald, J.T.; Wienecke, S.; Ishwarbhai, A.; Tsipa, A.; Aw, R.; Kylilis, N.; Bell, D.J.; McClymont, D.W.; Jensen, K.; et al. Rapid acquisition and model-based analysis of cell-free transcription–translation reactions from nonmodel bacteria. Proc. Natl. Acad. Sci. USA 2018, 115, E4340–E4349. [Google Scholar] [CrossRef] [PubMed]
  312. Wang, H.; Li, J.; Jewett, M.C. Development of a Pseudomonas putida cell-free protein synthesis platform for rapid screening of gene regulatory elements. Synth. Biol. 2018, 3, ysy003. [Google Scholar] [CrossRef]
  313. Tran, K.; Gurramkonda, C.; Cooper, M.A.; Pilli, M.; Taris, J.E.; Selock, N.; Han, T.-C.; Tolosa, M.; Zuber, A.; Peñalber-Johnstone, C.; et al. Cell-free production of a therapeutic protein: Expression, purification, and characterization of recombinant streptokinase using a CHO lysate. Biotechnol. Bioeng. 2018, 115, 92–102. [Google Scholar] [CrossRef] [PubMed]
  314. Martin, R.W.; Majewska, N.I.; Chen, C.X.; Albanetti, T.E.; Jimenez, R.B.C.; Schmelzer, A.E.; Jewett, M.C.; Roy, V. Development of a CHO-Based Cell-Free Platform for Synthesis of Active Monoclonal Antibodies. ACS Synth. Biol. 2017, 6, 1370–1379. [Google Scholar] [CrossRef] [PubMed]
  315. Liu, K.; Hu, J. Host-regulated Hepatitis B Virus Capsid Assembly in a Mammalian Cell-free System. Bio-protocol 2018, 8, e2813. [Google Scholar] [CrossRef] [PubMed]
  316. Kopniczky, M.B.; Canavan, C.; McClymont, D.W.; Crone, M.A.; Suckling, L.; Goetzmann, B.; Siciliano, V.; MacDonald, J.T.; Jensen, K.; Freemont, P.S. Cell-Free Protein Synthesis as a Prototyping Platform for Mammalian Synthetic Biology. ACS Synth. Biol. 2020, 9, 144–156. [Google Scholar] [CrossRef] [PubMed]
  317. Ezure, T.; Suzuki, T.; Higashide, S.; Shintani, E.; Endo, K.; Kobayashi, S.; Shikata, M.; Ito, M.; Tanimizu, K.; Nishimura, O. Cell-Free Protein Synthesis System Prepared from Insect Cells by Freeze-Thawing. Biotechnol. Prog. 2006, 22, 1570–1577. [Google Scholar] [CrossRef] [PubMed]
  318. Ezure, T.; Suzuki, T.; Shikata, M.; Ito, M.; Ando, E. A cell-free protein synthesis system from insect cells. Methods Mol. Biol. 2010, 607, 31–42. [Google Scholar] [CrossRef] [PubMed]
  319. Harbers, M. Wheat germ systems for cell-free protein expression. FEBS Lett. 2014, 588, 2762–2773. [Google Scholar] [CrossRef] [PubMed]
  320. Buntru, M.; Vogel, S.; Stoff, K.; Spiegel, H.; Schillberg, S. A versatile coupled cell-free transcription–translation system based on tobacco BY-2 cell lysates. Biotechnol. Bioeng. 2015, 112, 867–878. [Google Scholar] [CrossRef] [PubMed]
  321. Nomoto, M.; Tada, Y. Cell-free protein synthesis of plant transcription factors. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2018; Volume 1830, pp. 337–349. [Google Scholar]
  322. Zemella, A.; Thoring, L.; Hoffmeister, C.; Šamalíková, M.; Ehren, P.; Wüstenhagen, D.A.; Kubick, S. Cell-free protein synthesis as a novel tool for directed glycoengineering of active erythropoietin. Sci. Rep. 2018, 8, 8514. [Google Scholar] [CrossRef] [PubMed]
  323. Burgenson, D.; Gurramkonda, C.; Pilli, M.; Ge, X.; Andar, A.; Kostov, Y.; Tolosa, L.; Rao, G. Rapid recombinant protein expression in cell-free extracts from human blood. Sci. Rep. 2018, 8, 9569. [Google Scholar] [CrossRef]
  324. Li, J.; Wang, H.; Kwon, Y.-C.; Jewett, M.C. Establishing a high yielding streptomyces-based cell-free protein synthesis system. Biotechnol. Bioeng. 2017, 114, 1343–1353. [Google Scholar] [CrossRef] [PubMed]
  325. Buntru, M.; Vogel, S.; Spiegel, H.; Schillberg, S. Tobacco BY-2 cell-free lysate: An alternative and highly-productive plant-based in vitro translation system. BMC Biotechnol. 2014, 14, 37. [Google Scholar] [CrossRef]
  326. Kovtun, O.; Mureev, S.; Jung, W.; Kubala, M.H.; Johnston, W.; Alexandrov, K. Leishmania cell-free protein expression system. Methods 2011, 55, 58–64. [Google Scholar] [CrossRef] [PubMed]
  327. Wu, C.; Dasgupta, A.; Shen, L.; Bell-Pedersen, D.; Sachs, M.S. The cell free protein synthesis system from the model filamentous fungus Neurospora crassa. Methods 2018, 137, 11–19. [Google Scholar] [CrossRef] [PubMed]
  328. Gan, R.; Jewett, M.C. A combined cell-free transcription-translation system from Saccharomyces cerevisiae for rapid and robust protein synthe. Biotechnol. J. 2014, 9, 641–651. [Google Scholar] [CrossRef] [PubMed]
  329. Jaroentomeechai, T.; Stark, J.C.; Natarajan, A.; Glasscock, C.J.; Yates, L.E.; Hsu, K.J.; Mrksich, M.; Jewett, M.C.; DeLisa, M.P. Single-pot glycoprotein biosynthesis using a cell-free transcription-translation system enriched with glycosylation machinery. Nat. Commun. 2018, 9, 2686. [Google Scholar] [CrossRef] [PubMed]
  330. Martin, R.W.; Des Soye, B.J.; Kwon, Y.-C.; Kay, J.; Davis, R.G.; Thomas, P.M.; Majewska, N.I.; Chen, C.X.; Marcum, R.D.; Weiss, M.G.; et al. Cell-free protein synthesis from genomically recoded bacteria enables multisite incorporation of noncanonical amino acids. Nat. Commun. 2018, 9, 1203. [Google Scholar] [CrossRef]
  331. Shinoda, T.; Shinya, N.; Ito, K.; Ishizuka-Katsura, Y.; Ohsawa, N.; Terada, T.; Hirata, K.; Kawano, Y.; Yamamoto, M.; Tomita, T.; et al. Cell-free methods to produce structurally intact mammalian membrane proteins. Sci. Rep. 2016, 6, 30442. [Google Scholar] [CrossRef] [PubMed]
  332. Focke, P.J.; Hein, C.; Hoffmann, B.; Matulef, K.; Bernhard, F.; Dötsch, V.; Valiyaveetil, F.I. Combining in Vitro Folding with Cell Free Protein Synthesis for Membrane Protein Expression. Biochemistry 2016, 55, 4212–4219. [Google Scholar] [CrossRef] [PubMed]
  333. Baumann, A.; Kerruth, S.; Fitter, J.; Büldt, G.; Heberle, J.; Schlesinger, R.; Ataka, K. In-Situ Observation of Membrane Protein Folding during Cell-Free Expression. PLoS ONE 2016, 11, e0151051. [Google Scholar] [CrossRef]
  334. Fogeron, M.-L.; Badillo, A.; Penin, F.; Böckmann, A. Wheat Germ Cell-Free Overexpression for the Production of Membrane Proteins. Methods Mol. Biol. 2017, 1635, 91–108. [Google Scholar] [CrossRef] [PubMed]
  335. Junge, F.; Haberstock, S.; Roos, C.; Stefer, S.; Proverbio, D.; Dötsch, V.; Bernhard, F. Advances in cell-free protein synthesis for the functional and structural analysis of membrane proteins. New Biotechnol. 2011, 28, 262–271. [Google Scholar] [CrossRef] [PubMed]
  336. Marshall, R.; Noireaux, V. Quantitative modeling of transcription and translation of an all-E. coli cell-free system. Sci. Rep. 2019, 9, 11980. [Google Scholar] [CrossRef]
  337. Marshall, R.; Maxwell, C.S.; Collins, S.P.; Jacobsen, T.; Luo, M.L.; Begemann, M.B.; Gray, B.N.; January, E.; Singer, A.; He, Y.; et al. Rapid and Scalable Characterization of CRISPR Technologies Using an E. coli Cell-Free Transcription-Translation System. Mol. Cell 2018, 69, 146–157.e3. [Google Scholar] [CrossRef] [PubMed]
  338. Des Soye, B.J.; Gerbasi, V.R.; Thomas, P.M.; Kelleher, N.L.; Jewett, M.C. A Highly Productive, One-Pot Cell-Free Protein Synthesis Platform Based on Genomically Recoded Escherichia coli. Cell Chem. Biol. 2019, 26, 1743–1754.e9. [Google Scholar] [CrossRef]
  339. Vilkhovoy, M.; Horvath, N.; Shih, C.-H.; Wayman, J.A.; Calhoun, K.; Swartz, J.; Varner, J.D. Sequence Specific Modeling of E. coli Cell-Free Protein Synthesis. ACS Synth. Biol. 2018, 7, 1844–1857. [Google Scholar] [CrossRef]
  340. Hodgman, C.E.; Jewett, M.C. Optimized extract preparation methods and reaction conditions for improved yeast cell-free protein synthesis. Biotechnol. Bioeng. 2013, 110, 2643–2654. [Google Scholar] [CrossRef]
  341. Hodgman, C.E.; Jewett, M.C. Characterizing IGR IRES-mediated translation initiation for use in yeast cell-free protein synthesis. New Biotechnol. 2014, 31, 499–505. [Google Scholar] [CrossRef] [PubMed]
  342. Choudhury, A.; Hodgman, C.E.; Anderson, M.J.; Jewett, M.C. Evaluating fermentation effects on cell growth and crude extract metabolic activity for improved yeast cell-free protein synthesis. Biochem. Eng. J. 2014, 91, 140–148. [Google Scholar] [CrossRef]
  343. Miura, K.; Tachibana, M.; Takashima, E.; Morita, M.; Kanoi, B.N.; Nagaoka, H.; Baba, M.; Torii, M.; Ishino, T.; Tsuboi, T. Malaria transmission-blocking vaccines: Wheat germ cell-free technology can accelerate vaccine development. Expert Rev. Vaccines 2019, 18, 1017–1027. [Google Scholar] [CrossRef]
  344. Das Gupta, M.; Flaskamp, Y.; Roentgen, R.; Juergens, H.; Armero-Gimenez, J.; Albrecht, F.; Hemmerich, J.; Arfi, Z.A.; Neuser, J.; Spiegel, H. Scaling eukaryotic cell-free protein synthesis achieved with the versatile and high-yielding tobacco BY-2 cell lysate. Biotechnol. Bioeng. 2023, 120, 2890–2906. [Google Scholar] [CrossRef]
  345. Zemella, A.; Thoring, L.; Hoffmeister, C.; Kubick, S. Cell-Free Protein Synthesis: Pros and Cons of Prokaryotic and Eukaryotic Systems. Chembiochem 2015, 16, 2420–2431. [Google Scholar] [CrossRef] [PubMed]
  346. Kubick, S.; Schacherl, J.; Fleischer-Notter, H.; Royall, E.; Roberts, L.O.; Stiege, W. In Vitro Translation in an Insect-Based Cell-Free System. In Cell-Free Protein Expression; Springer: Berlin/Heidelberg, Germany, 2003; pp. 209–217. [Google Scholar]
  347. Thoring, L.; Dondapati, S.K.; Stech, M.; Wüstenhagen, D.A.; Kubick, S. High-yield production of “difficult-to-express” proteins in a continuous exchange cell-free system based on CHO cell lysates. Sci. Rep. 2017, 7, 11710. [Google Scholar] [CrossRef]
  348. Thoring, L.; Wüstenhagen, D.A.; Borowiak, M.; Stech, M.; Sonnabend, A.; Kubick, S. Cell-Free Systems Based on CHO Cell Lysates: Optimization Strategies, Synthesis of “Difficult-to-Express” Proteins and Future Perspectives. PLoS ONE 2016, 11, e0163670. [Google Scholar] [CrossRef] [PubMed]
  349. Brödel, A.K.; Sonnabend, A.; Roberts, L.O.; Stech, M.; Wüstenhagen, D.A.; Kubick, S. IRES-Mediated Translation of Membrane Proteins and Glycoproteins in Eukaryotic Cell-Free Systems. PLoS ONE 2013, 8, e82234. [Google Scholar] [CrossRef] [PubMed]
  350. Burgenson, D.; Linton, J.; Ge, X.; Kostov, Y.; Tolosa, L.; Szeto, G.L.; Rao, G. A Cell-Free Protein Expression System Derived from Human Primary Peripheral Blood Mononuclear Cells. ACS Synth. Biol. 2020, 9, 2188–2196. [Google Scholar] [CrossRef] [PubMed]
  351. Welsh, J.P.; Lu, Y.; He, X.-S.; Greenberg, H.B.; Swartz, J.R. Cell-free production of trimeric influenza hemagglutinin head domain proteins as vaccine antigens. Biotechnol. Bioeng. 2012, 109, 2962–2969. [Google Scholar] [CrossRef] [PubMed]
  352. Lu, Y.; Welsh, J.P.; Swartz, J.R. Production and stabilization of the trimeric influenza hemagglutinin stem domain for potentially broadly protective influenza vaccines. Proc. Natl. Acad. Sci. USA 2014, 111, 125–130. [Google Scholar] [CrossRef] [PubMed]
  353. Sheng, J.; Lei, S.; Yuan, L.; Feng, X. Cell-free protein synthesis of norovirus virus-like particles. RSC Adv. 2017, 7, 28837–28840. [Google Scholar] [CrossRef]
  354. Brito, L.A.; Singh, M. Acceptable levels of endotoxin in vaccine formulations during preclinical research. J. Pharm. Sci. 2011, 100, 34–37. [Google Scholar] [CrossRef]
  355. Yamaoka, Y.; Matsuyama, S.; Fukushi, S.; Matsunaga, S.; Matsushima, Y.; Kuroyama, H.; Kimura, H.; Takeda, M.; Chimuro, T.; Ryo, A. Development of Monoclonal Antibody and Diagnostic Test for Middle East Respiratory Syndrome Coronavirus Using Cell-Free Synthesized Nucleocapsid Antigen. Front. Microbiol. 2016, 7, 509. Available online: https://www.frontiersin.org/article/10.3389/fmicb.2016.00509 (accessed on 5 June 2024). [CrossRef] [PubMed]
  356. Spice, A.J.; Aw, R.; Bracewell, D.G.; Polizzi, K.M. Synthesis and Assembly of Hepatitis B Virus-Like Particles in a Pichia pastoris Cell-Free System. Front. Bioeng. Biotechnol. 2020, 8, 72. Available online: https://www.frontiersin.org/article/10.3389/fbioe.2020.00072 (accessed on 5 June 2024). [CrossRef] [PubMed]
  357. Yamaoka, Y.; Jeremiah, S.S.; Funabashi, R.; Miyakawa, K.; Morita, T.; Mihana, Y.; Kato, H.; Ryo, A. Characterization and Utilization of Disulfide-Bonded SARS-CoV-2 Receptor Binding Domain of Spike Protein Synthesized by Wheat Germ Cell-Free Production System. Viruses 2022, 14, 1461. [Google Scholar] [CrossRef]
  358. Takai, K.; Sawasaki, T.; Endo, Y. The wheat-germ cell-free expression system. Curr. Pharm. Biotechnol. 2010, 11, 272–278. [Google Scholar] [CrossRef]
  359. Kaur, J.; Kumar, A.; Kaur, J. Strategies for optimization of heterologous protein expression in E. coli: Roadblocks and reinforcements. Int. J. Biol. Macromol. 2018, 106, 803–822. [Google Scholar] [CrossRef] [PubMed]
  360. Dondapati, S.K.; Wüstenhagen, D.A.; Kubick, S. Functional Analysis of Membrane Proteins Produced by Cell-Free Translation. In Protein Engineering: Methods and Protocols; Bornscheuer, U.T., Höhne, M., Eds.; Springer: New York, NY, USA, 2018; pp. 171–186. [Google Scholar]
  361. Venkat, S.; Chen, H.; Gan, Q.; Fan, C. The Application of Cell-Free Protein Synthesis in Genetic Code Expansion for Post-translational Modifications. Front. Pharmacol. 2019, 10, 248. Available online: https://www.frontiersin.org/article/10.3389/fphar.2019.00248 (accessed on 5 June 2024). [CrossRef] [PubMed]
  362. Karim, A.S.; Jewett, M.C. Chapter Two—Cell-Free Synthetic Biology for Pathway Prototyping. In Enzymes in Synthetic Biology; Scrutton, N., Ed.; Academic Press: Cambridge, MA, USA, 2018; Volume 608, pp. 31–57. [Google Scholar]
  363. Bundy, B.C.; Hunt, J.P.; Jewett, M.C.; Swartz, J.R.; Wood, D.W.; Frey, D.D.; Rao, G. Cell-free biomanufacturing. Curr. Opin. Chem. Eng. 2018, 22, 177–183. [Google Scholar] [CrossRef]
  364. Lu, Y. Cell-free synthetic biology: Engineering in an open world. Synth. Syst. Biotechnol. 2017, 2, 23–27. [Google Scholar] [CrossRef] [PubMed]
  365. Adiga, R.; Al-Adhami, M.; Andar, A.; Borhani, S.; Brown, S.; Burgenson, D.; Cooper, M.A.; Deldari, S.; Frey, D.D.; Ge, X.; et al. Point-of-care production of therapeutic proteins of good-manufacturing-practice quality. Nat. Biomed. Eng. 2018, 2, 675–686. [Google Scholar] [CrossRef] [PubMed]
  366. Maghodia, A.B.; Geisler, C.; Jarvis, D.L. Characterization of an Sf-rhabdovirus-negative Spodoptera frugiperda cell line as an alternative host for recombinant protein production in the baculovirus-insect cell system. Protein Expr. Purif. 2016, 122, 45–55. [Google Scholar] [CrossRef] [PubMed]
  367. Sonnabend, A.; Spahn, V.; Stech, M.; Zemella, A.; Stein, C.; Kubick, S. Production of G protein-coupled receptors in an insect-based cell-free system. Biotechnol. Bioeng. 2017, 114, 2328–2338. [Google Scholar] [CrossRef] [PubMed]
  368. Stech, M.; Merk, H.; Schenk, J.A.; Stöcklein, W.F.M.; Wüstenhagen, D.A.; Micheel, B.; Duschl, C.; Bier, F.F.; Kubick, S. Production of functional antibody fragments in a vesicle-based eukaryotic cell-free translation system. J. Biotechnol. 2012, 164, 220–231. [Google Scholar] [CrossRef] [PubMed]
  369. Merk, H.; Gless, C.; Maertens, B.; Gerrits, M.; Stiege, W. Cell-free synthesis of functional and endotoxin-free antibody Fab fragments by translocation into microsomes. Biotechniques 2012, 53, 153–160. [Google Scholar] [CrossRef]
  370. Lai, T.; Yang, Y.; Ng, S. Advances in Mammalian Cell Line Development Technologies for Recombinant Protein Production. Pharmaceuticals 2013, 6, 579–603. [Google Scholar] [CrossRef] [PubMed]
  371. 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]
  372. Brödel, A.K.; Sonnabend, A.; Kubick, S. Cell-free protein expression based on extracts from CHO cells. Biotechnol. Bioeng. 2014, 111, 25–36. [Google Scholar] [CrossRef] [PubMed]
  373. Sachse, R.; Dondapati, S.K.; Fenz, S.F.; Schmidt, T.; Kubick, S. Membrane protein synthesis in cell-free systems: From bio-mimetic systems to bio-membranes. FEBS Lett. 2014, 588, 2774–2781. [Google Scholar] [CrossRef]
  374. Cole, S.D.; Beabout, K.; Turner, K.B.; Smith, Z.K.; Funk, V.L.; Harbaugh, S.V.; Liem, A.T.; Roth, P.A.; Geier, B.A.; Emanuel, P.A.; et al. Quantification of Interlaboratory Cell-Free Protein Synthesis Variability. ACS Synth. Biol. 2019, 8, 2080–2091. [Google Scholar] [CrossRef] [PubMed]
  375. Chizzolini, F.; Forlin, M.; Yeh Martín, N.; Berloffa, G.; Cecchi, D.; Mansy, S.S. Cell-Free Translation Is More Variable than Transcription. ACS Synth. Biol. 2017, 6, 638–647. [Google Scholar] [CrossRef] [PubMed]
  376. Dopp, J.L.; Reuel, N.F. Process optimization for scalable E. coli extract preparation for cell-free protein synthesis. Biochem. Eng. J. 2018, 138, 21–28. [Google Scholar] [CrossRef]
  377. Dopp, J.L.; Jo, Y.R.; Reuel, N.F. Methods to reduce variability in E. Coli-based cell-free protein expression experiments. Synth. Syst. Biotechnol. 2019, 4, 204–211. [Google Scholar] [CrossRef]
  378. Wang, Y.; Zhao, S.; Bai, L.; Fan, J.; Liu, E. Expression systems and species used for transgenic animal bioreactors. Biomed Res. Int. 2013, 2013, 580463. [Google Scholar] [CrossRef] [PubMed]
  379. Shepelev, M.V.; Kalinichenko, S.V.; Deykin, A.V.; Korobko, I.V. Production of Recombinant Proteins in the Milk of Transgenic Animals: Current State and Prospects. Acta Naturae 2018, 10, 40–47. [Google Scholar] [CrossRef] [PubMed]
  380. Pollock, D.P.; Kutzko, J.P.; Birck-Wilson, E.; Williams, J.L.; Echelard, Y.; Meade, H.M. Transgenic milk as a method for the production of recombinant antibodies. J. Immunol. Methods 1999, 231, 147–157. [Google Scholar] [CrossRef] [PubMed]
  381. Meade, H.M.; Echelard, Y.; Ziomek, C.A.; Young, M.W.; Harvey, M.; Cole, E.S.; Groet, S.; Smith, T.E.; Curling, J.M. Expression of Recombinant Proteins in the Milk of Transgenic Animals. Gene Expr. Syst. 1999, 10, 399–427. [Google Scholar] [CrossRef]
  382. Lillico, S.G.; Sherman, A.; McGrew, M.J.; Robertson, C.D.; Smith, J.; Haslam, C.; Barnard, P.; Radcliffe, P.A.; Mitrophanous, K.A.; Elliot, E.A. Oviduct-specific expression of two therapeutic proteins in transgenic hens. Proc. Natl. Acad. Sci. USA 2007, 104, 1771–1776. [Google Scholar] [CrossRef]
  383. Kerr, D.E.; Liang, F.; Bondioli, K.R.; Zhao, H.; Kreibich, G.; Wall, R.J.; Sun, T.-T. The bladder as a bioreactor: Urothelium production and secretion of growth hormone into urine. Nat. Biotechnol. 1998, 16, 75–79. [Google Scholar] [CrossRef] [PubMed]
  384. Dyck, M.K.; Lacroix, D.; Pothier, F.; Sirard, M.-A. Making recombinant proteins in animals–different systems, different applications. TRENDS Biotechnol. 2003, 21, 394–399. [Google Scholar] [CrossRef] [PubMed]
  385. CHRENEK, P.P.; Makarevich, A.V.; Pivko, J.; Bulla, J. Transgenic farm animal production and application. Slovak J. Anim. Sci. 2010, 43, 45–49. [Google Scholar]
  386. Chen, H.-L.; Huang, J.-Y.; Chu, T.-W.; Tsai, T.-C.; Hung, C.-M.; Lin, C.-C.; Liu, F.-C.; Wang, L.-C.; Chen, Y.-J.; Lin, M.-F.; et al. Expression of VP1 protein in the milk of transgenic mice: A potential oral vaccine protects against enterovirus 71 infection. Vaccine 2008, 26, 2882–2889. [Google Scholar] [CrossRef] [PubMed]
  387. Nakade, S.; Tsubota, T.; Sakane, Y.; Kume, S.; Sakamoto, N.; Obara, M.; Daimon, T.; Sezutsu, H.; Yamamoto, T.; Sakuma, T. Microhomology-mediated end-joining-dependent integration of donor DNA in cells and animals using TALENs and CRISPR/Cas9. Nat. Commun. 2014, 5, 5560. [Google Scholar] [CrossRef]
  388. Cristea, S.; Freyvert, Y.; Santiago, Y.; Holmes, M.C.; Urnov, F.D.; Gregory, P.D.; Cost, G.J. In vivo cleavage of transgene donors promotes nuclease-mediated targeted integration. Biotechnol. Bioeng. 2013, 110, 871–880. [Google Scholar] [CrossRef]
Figure 1. Molecular cloning overview.
Figure 1. Molecular cloning overview.
Vaccines 12 01344 g001
Figure 2. Bacterial expression system.
Figure 2. Bacterial expression system.
Vaccines 12 01344 g002
Figure 3. Insect cell (baculovirus) expression system.
Figure 3. Insect cell (baculovirus) expression system.
Vaccines 12 01344 g003
Figure 4. Mammalian expression system.
Figure 4. Mammalian expression system.
Vaccines 12 01344 g004
Figure 5. Yeast expression system.
Figure 5. Yeast expression system.
Vaccines 12 01344 g005
Figure 6. Algal expression system.
Figure 6. Algal expression system.
Vaccines 12 01344 g006
Figure 7. Plant expression system.
Figure 7. Plant expression system.
Vaccines 12 01344 g007
Figure 8. Cell-free expression system.
Figure 8. Cell-free expression system.
Vaccines 12 01344 g008
Table 1. In-development viral vaccines produced using E. coli expression.
Table 1. In-development viral vaccines produced using E. coli expression.
VirusAntigenFindingsReference
Foot and mouth disease virusVP0, VP1 and VP3 VLPsDemonstrated protection in guinea pigs, swine and cattle. Local mucosal and systemic immune responses in mice via IN immunization.[43,44,45,46]
VP1Protected mice and pigs from challenge.[47]
Influenza A virusACAM-FLU-APhase I clinical trials.[48,49,50]
Porcine circovirus type 2PCV2-Cap VLPExperimental vaccine demonstrating efficacy in piglets.[51]
Porcine parvovirusPPV-VP2 VLPDemonstrated efficacy in piglets.[51]
SARS-CoV-2Dual-adjuvanted RBDDemonstrated robust neutralizing antibody response in mice.[52]
Table 2. Licenced BEVS-expressed vaccines.
Table 2. Licenced BEVS-expressed vaccines.
VirusVaccineAntigenManufacturerReference
Classical swine fever virusPorcilis® PestiE2MSD Animal Health (Rahway, NJ, USA)[90]
BAYOVAC CSF E2®E2Bayer/Pfizer Animal Health (Leverkusen, Germany)
Human papillomavirusCevarix®L1GlaxoSmithKline (Mississauga, ON, Canada)[91]
Influenza A virusFlublok®HAProtein Sciences Corporation
(Meriden, CT, USA)
[92]
Porcine circovirus type 2Circumvent® PCVG2Merck Animal Health (Rahway, NJ, USA)[93]
Ingelvac CircoFLEX®ORF2Boehringer Ingelheim Vetmedica
(Duluth, GA, USA)
[94]
Porcilis® PCVORF2MSD Animal Health (Rahway, NJ, USA)[95]
Table 3. In-development BEVS-expressed vaccines.
Table 3. In-development BEVS-expressed vaccines.
VirusAntigenApplicationReference
Chikungunya virusCHIKV-S27 structural polyprotein (C, E3, E2, 6K, E1)Experimental vaccine preventing viremia and inflammation in mice[96]
Chikungunya virusE1 and E2 proteinsExperimental vaccine demonstrating protection in mice[97]
Classical swine fever virusE2 proteinExperimental vaccine demonstrating protective immunity in pigs[98]
EbolavirusGP and VP40 VLPsExperimental vaccine demonstrating protection in guinea pigs[99]
Epizootic hemorrhagic disease virusVP2 proteinExperimental vaccine preventing clinical disease or viremia in deer[100]
Influenza A virusHA, NA and M1 VLPsExperimental vaccine demonstrating protection in pigs against pH1N1 and in chickens against H6N1[101,102]
Lassa virusGlycoprotein (GP)Experimental vaccine eliciting high antibody titres in mice[88]
Rift Valley fever virusGn and Gc GlycoproteinsExperimental vaccine demonstrating complete protection in sheep[103,104]
SARS-CoV-2RBDExperimental vaccine demonstrating protection in NHPs[105,106]
Zika virusE80 and EDIII proteinsExperimental vaccine demonstrating protection in mice[89]
Table 4. Licenced mammalian cell-expressed viral antigens.
Table 4. Licenced mammalian cell-expressed viral antigens.
VirusVaccineCell LineAntigenManufacturerReference
Classical swine feverPorvac®HEK 293E2-CD154The Centre for Genetic Engineering and Biotechnology (CIGB) (Havana, Cuba)[139]
Dengue virusDengvaxia®VeropreM and ESanofi Pasteur (Val de Reuil
France)
[13]
Influenza A (H1N1) 2009Celvapan®VeroWhole virionBaxter (Orth an der Donau Austria)[140,141]
Influenza A (trivalent)Flucelvax®MDCKHA and NANovartis Vaccines and Diagnostics (Basel, Switzerland) [12,142]
Influenza A (trivalent)Preflucel®VeroHABaxter (Orth an der Donau Austria)[11]
Influenza H5N1Celvapan®VeroWhole virionBaxter (Orth an der Donau Austria)[140,141]
Respiratory syncytial virusAREXEVY®CHORSVPreFGlaxoSmithKline (London, UK)[143,144,145,146,147]
Respiratory syncytial virusABRYSVO®CHORSVPreFPfizer (New York, NY, USA)[148,149,150,151]
Table 5. In-development mammalian cell-expressed viral antigens.
Table 5. In-development mammalian cell-expressed viral antigens.
VirusAntigenCell LineApplicationReference
Classical swine fever virusE2 ProteinCHOExperimental mucosal vaccine demonstrating protection in pigs.[152]
Hendra virussGHeVHeLaExperimental vaccine demonstrating protective efficacy in African green monkeys and cats.[153,154,155]
Hendra virussGHeV293FExperimental vaccine demonstrating protective efficacy in ferrets and horses.[156,157,158]
SARS-CoV-2RBD-nanoparticleHEK-293Experimental vaccine eliciting strong neutralizing antibody responses and rapid viral clearance in nasal washes.[159]
SARS-CoV-2Pan-HLA-DR mAb fused to RBDHEK-293Experimental vaccine demonstrating robust protection, strong neutralizing antibody responses and viral clearance in ferret nasal washes.[160]
Table 6. In-development yeast-expressed viral antigens.
Table 6. In-development yeast-expressed viral antigens.
Yeast SpeciesVirusAntigenReference
H. polymorphaPapillomavirusL1 protein[188]
Hepatitis B virusVrHB-IB[189]
Porcine circovirus type 2PCV2b capsid protein[190]
P. pastorisDengue virusEnvelope glycoproteins [191,192,193,194,195]
Hand, foot and mouth disease (HFMD)P1 and 3CD proteins [196]
Hepatitis B virusHBsAg[197]
Hepatitis C virusHCV Core protein[198]
PapillomavirusHPV 16L1, 18L1[199,200]
Influenza virusHemagglutinin protein[201,202,203]
Classical swine fever virusE2 glycoprotein[204,205]
Chikungunya virusStructural protein VLPs[206]
SARS-CoV-2RBD monomers and dimers[207]
S. cerevisiaePapillomavirusVLPs of hrHPV16 and 18, and lrHPV6 and 11—Gardasil®[187]
HPV16[208]
Hepatitis B virusHepatitis B surface antigen (HBsAg)—Recombivax®[209,210]
HBV X, S and C antigens[211,212]
Surface protein GS-4774[213]
Enterovirus 71 (hand, foot and mouth disease)EV71 Structural antigens[214,215]
Parvovirus B19VP[216,217]
Dengue virusDengue envelope domain III[218]
Human immunodeficiency virus-1Envelope glycoprotein[219]
Table 7. In-development plant-expressed viral antigens.
Table 7. In-development plant-expressed viral antigens.
PlantVirusAntigenApplicationReference
Nicotiana benthamianaInfluenzaH1 and H5 VLPsExperimental vaccine eliciting strong humoral and long-term cell-mediated responses in adults.[270]
InfluenzaH7N9 VLPExperimental vaccine demonstrating strong antibody response in mice and ferrets.[271]
InfluenzaHA subunitExperimental vaccine demonstrating immunogenicity in mice, rabbits and ferrets. [272]
InfluenzaH6 VLPExperimental vaccine demonstrating reduced viral shedding in chickens.[273]
Infectious bronchitis virusS protein VLPExperimental vaccine demonstrating immunogenicity in chickens.[274]
SARS-CoV-2RBDExperimental vaccine shown to induce neutralizing antibodies in mice and NHPs.[275]
Table 9. Comparison of cellular vs. cell-free antigen expression systems.
Table 9. Comparison of cellular vs. cell-free antigen expression systems.
Cellular ExpressionCell-Free ExpressionReferences
Time1–2 weeks24–72 h[308,344]
Protein ToxicityOften a major issueHigh tolerance for toxic proteins[289,359]
Membrane ProteinsOverexpression can cause cytotoxicity and cell deathVariety of different sizes can be produced depending on lysate source[331,332,360]
Protein YieldHigh yields (mg/mL)Yield varies depending on the protein from µg/mL to mg/mL[288,304]
Post-translational ModificationsAll possible depending on the system.Mainly in eukaryotic CF systems with translationally active microsomes. Limited in prokaryotic and eukaryotic lysates without endogenous microsomes. O-linked glycosylation does not occur.[302,361]
ScalabilityMinimum volume of 5 mL to several litres.Range from 5 µL (chip-based, Eppendorf tube) to 100 L (fermenter, commercial bioreactor)[362,363]
Flexibility and ManipulationClosed system, difficult to manipulateOpen system, easy to manipulate reaction conditions, no cell membrane constraints.[364]
Point-of-care protein productionDifficult due to time-consuming process, infrastructural facilities and cold storage.Lyophilization allows for removal of cold-chain and on-site production without major facilities.[298,365]
ApplicationsCells need to be lysed for membrane protein applications.Simple as protein can be purified and reconstituted immediately post-synthesis.[288,364]
AcceptanceReliable, current standard for protein production approved by regulatory authorities.Currently limited to research at laboratory level with few, limited commercial applications[289,304,344]
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

Sookhoo, J.R.V.; Schiffman, Z.; Ambagala, A.; Kobasa, D.; Pardee, K.; Babiuk, S. Protein Expression Platforms and the Challenges of Viral Antigen Production. Vaccines 2024, 12, 1344. https://doi.org/10.3390/vaccines12121344

AMA Style

Sookhoo JRV, Schiffman Z, Ambagala A, Kobasa D, Pardee K, Babiuk S. Protein Expression Platforms and the Challenges of Viral Antigen Production. Vaccines. 2024; 12(12):1344. https://doi.org/10.3390/vaccines12121344

Chicago/Turabian Style

Sookhoo, Jamie R. V., Zachary Schiffman, Aruna Ambagala, Darwyn Kobasa, Keith Pardee, and Shawn Babiuk. 2024. "Protein Expression Platforms and the Challenges of Viral Antigen Production" Vaccines 12, no. 12: 1344. https://doi.org/10.3390/vaccines12121344

APA Style

Sookhoo, J. R. V., Schiffman, Z., Ambagala, A., Kobasa, D., Pardee, K., & Babiuk, S. (2024). Protein Expression Platforms and the Challenges of Viral Antigen Production. Vaccines, 12(12), 1344. https://doi.org/10.3390/vaccines12121344

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