Next Article in Journal
Assessment of the Interferon-Lambda-3 Polymorphism in the Antibody Response to COVID-19 in Older Adults Seropositive for CMV
Next Article in Special Issue
Candida parapsilosis Cell Wall Proteome Characterization and Effectiveness against Hematogenously Disseminated Candidiasis in a Murine Model
Previous Article in Journal
Parental Decision Making Regarding COVID-19 Vaccines for Children under Age 5: Does Decision Self-Efficacy Play a Role?
Previous Article in Special Issue
Vaccinating Children against SARS-CoV-2: A Literature Review and Survey of International Experts to Assess Safety, Efficacy and Perceptions of Vaccine Use in Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery

1
Department of Clinical Microbiology and Infectious Diseases, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
2
Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
3
Infection Control, Charlotte Maxeke Johannesburg Academic Hospital, National Health Laboratory Service, Johannesburg 2193, South Africa
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current address: Department of Pathology, Collage of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
Vaccines 2023, 11(2), 479; https://doi.org/10.3390/vaccines11020479
Submission received: 2 January 2023 / Revised: 6 February 2023 / Accepted: 14 February 2023 / Published: 18 February 2023
(This article belongs to the Special Issue New Trends in Vaccine Characterization, Formulations, and Development)

Abstract

:
Virus-like particles (VLPs) are empty, nanoscale structures morphologically resembling viruses. Internal cavity, noninfectious, and particulate nature with a high density of repeating epitopes, make them an ideal platform for vaccine development and drug delivery. Commercial use of Gardasil-9 and Cervarix showed the usefulness of VLPs in vaccine formulation. Further, chimeric VLPs allow the raising of an immune response against different immunogens and thereby can help reduce the generation of medical or clinical waste. The economically viable production of VLPs significantly impacts their usage, application, and availability. To this end, several hosts have been used and tested. The present review will discuss VLPs produced using different yeasts as fermentation hosts. We also compile a list of studies highlighting the expression and purification of VLPs using a yeast-based platform. We also discuss the advantages of using yeast to generate VLPs over other available systems. Further, the issues or limitations of yeasts for producing VLPs are also summarized. The review also compiles a list of yeast-derived VLP-based vaccines that are presently in public use or in different phases of clinical trials.

1. Background of Vaccines

The term vaccine originated from the Latin word “Vacca”, meaning cow. Vaccines are a biological formulation of weakened or killed pathogens (sometimes closely related species of the target pathogen), which, upon administration, help protect an individual against the target disease(s) by imitating an infection. Generally, vaccines are prophylactic except for the vaccine against rabies, which is curative [1]. Primarily, whole microbial pathogens were used in vaccine formulation along with other constituents such as preservatives or stabilizers, adjuvants, or immune boosters [2]. However, the conventional vaccine development strategy is not suitable for combating those diseases where the associated pathogen defies the current microbiological growth procedures on a commercial scale (for example, Mycobacterium leprae and species of Plasmodium) [3,4].
The arrival of subunit vaccines, which involved only a few components of the pathogen, overcame the loophole associated with the traditional vaccine development regime. Apart from the surface proteins (from bacteria, protozoa, and viruses), subunit vaccines may also include pathogen-specific toxin(s) and polysaccharides [5]. The most recent and dramatic development in vaccine formulation is nucleic acid-based vaccines (which include DNA and mRNA) [6]. These vaccines overcome the issues associated with conventional and subunit vaccine development processes, such as the expression and purification of target proteins and the need to propagate serious pathogens [7,8,9]. Thus, nucleic acid-based vaccines gave a new horizon to the therapeutic regimen in healthcare settings; for instance, mRNA-based vaccines against COVID-19 (manufactured by Moderna and Pfizer) are an advancement over the traditional vaccine development against infectious diseases [10,11,12,13]. Thus, there is continuous improvement in the methods and strategies used in vaccine development and delivery. Due to the arrival of newer methods of vaccine design and development, there is a need to redefine vaccines. Today, vaccines can be redefined as biological preparations containing attenuated or killed pathogens or any component, including peptides, proteins, DNA, mRNA, or polysaccharides, which, upon injection, help prevent an individual from any possible future infection [6]. Although subunit and nucleic acid-based vaccines offer several advantages over conventional methods of vaccine development, they still suffer from issues like the need for a cold chain or deep refrigeration and a short shelf life like other common vaccines [14]. Figure 1 shows the different methods of antiviral vaccine formulation.
Initially, vaccines were used only against infectious diseases (viral or bacterial), but now vaccines are considered the most promising means to combat certain types of cancer. For example, Gardasil-9 (the L1 protein of the human papillomavirus, or HPV), is expressed and purified from yeast against HPV, whose infection may lead to cervical cancer. Thus, in the contemporary world, vaccines are not just a tool to fight against infectious diseases but are also used to prevent certain types of cancer associated with viral infections [15,16,17]. Apart from papilloma, several other anticancer vaccines are in different stages of clinical trials [18,19,20,21,22].
Vaccine development platforms based on subunits or nucleic acids have actualized the development of vaccines against diseases considered unfeasible a few decades ago (for example, a vaccine against malaria) [23]. Additionally, these approaches have expedited vaccine development programs and made them safer for individuals; the development and application of an mRNA-based vaccine against COVID-19 is the best-suited example for this kind of platform [10,11,12,13]. A continuous cold chain is needed to maintain the efficacy or potency of vaccines. To tackle this, researchers are exploring further advancements to fix the problem associated with the thermolabile nature of vaccines and minimize the requirement of a cold chain, as discussed elsewhere [14,24,25].
After briefly introducing vaccines, the different development platforms used, and their shortcomings, it is now time to focus on one of the emerging platforms in the field of vaccine development. The intended purpose of this review is to discuss the suitability of VLPs as a means of vaccine development and drug delivery, with a particular focus on VLPs derived from yeast. We will also discuss how VLPs are a better platform than purified proteins for triggering immune responses in the host. The present review also highlights the reasons that make yeast the most acceptable model for the expression and purification of VLPs; the review also includes associated concerns and success stories about this concept. We also discuss the major yeast species used to express and purify VLPs. All of this is discussed under separate headings.

2. Introduction to Virus-Like Particles

VLPs, ghost viruses, or dummy viruses lacking genetic material are nanostructures first observed in sera samples from hepatitis patients in 1968 [26]. VLPs can exist naturally (in a virally infected host) and can be generated in the laboratory. The size of VLPs may vary from 20 nm to 200 nm or more [27]. The size of VLPs depends on the virus species and viral proteins used for developing these particles [27]. It is essential to mention that VLPs can be formed using the capsid, envelope, or core viral proteins [27]. These particles are usually formed naturally by folding viral proteins under appropriate conditions, including the optimum pH, salt concentration, temperature, and so on [28,29,30]. The VLPs form when monomeric proteins fold into a pentameric form, also called capsomers, which are then assembled to form VLPs [30]. VLPs can exist either non-enveloped, as seen in the case of HPV VLPs, or enveloped with a lipid membrane (eVLPs), such as SARS coronavirus VLPs [31,32]. The shape of VLPs also differs from icosahedral to rod-shaped [33]. These particles may be composed of a single protein or can be a fusion of two different proteins (chimeric VLPs) [34,35].
The feature that makes VLPs important in vaccine development is their high density of epitopes, particulate nature, and lack of genetic material that restricts their replication and makes them safe for the host [26,36,37,38]. In addition to their use in vaccine development, VLPs are also widely investigated to deliver drugs and other small molecules inside the host system. This property is attributed to the internal cavity in VLPs. Several studies also showed the feasibility of using VLPs in photo imaging [37,38]. Different applications of VLPs are shown in Figure 2.
The high epitope density and particulate nature of VLPs make them ideal systems for mounting immune responses. It is essential to mention that VLPs can mount both humoral and cellular immune responses [39,40,41,42,43,44]. Because of their natural resemblance to viruses, VLPs act as pathogen-associated structural patterns (PASP) and are easily recognized and taken up by host immune cells [39,40]. Additionally, their structural properties help activate immune cells like dendritic cells [45]. Due to their ability to mount both humoral and cellular immune responses, VLPs appear to be a better choice for vaccine delivery than purified proteins [46].
Due to their importance in vaccine development and drug delivery, efforts were made to express and purify VLPs from different biological systems and identify the most suitable hosts for producing VLPs commercially. To this end, VLPs are successfully expressed and purified from both the prokaryotic system (for example, Escherichia coli) [34] as well as from the eukaryotic system, including yeast (for example, S. cerevisiae) [47], insect cell lines [48,49], mammalian cell lines [39,50], and plants [51] (as shown in Figure 3).
Apart from whole cells, several studies have shown that VLPs can be produced by in vitro protein translation systems [52]. The advantages and disadvantages of in vitro protein translation-based VLP generation are discussed by others [22,52,53]. In Table 1, the advantages and disadvantages of different systems used for the expression and purification of VLPs are compared.

3. Yeast, Host to Produce VLPs on a Commercial Scale

As mentioned above, several biological systems have been evaluated over the years for a suitable host to produce VLPs on a commercial scale. An ideal host for the commercial purification of VLPs should be nonpathogenic, easy to handle, able to grow on economic media, able to express the protein of interest in the maximum amount, allow proper folding of a large amount of expressed protein, be genetically responsive, and be easily scaled up to industrial measures [54]. The secretion of the expressed protein(s) and VLPs (both enveloped and non-enveloped) into the medium will be another beneficial facet. Looking at these attributes, a yeast-based system appears as an ideal host for the commercial production of VLPs. In Table 1, the advantages, and disadvantages of yeast over other hosts are compared.
In the past, several yeasts were successfully used to express and purify clinically relevant proteins. Yeast species, S. cerevisiae and K. phaffii (formerly known as Pichia pastoris), fall under GARS (Generally Recognized as Safe), which is another significant advantage of using yeast-based systems for the development of VLPs [55]. Unlike the bacterial system, the yeast-based system does not suffer from endotoxin problems [56], and the solubility of expressed proteins and folding are much better in yeast compared to bacteria [57]. Furthermore, utilizing mammalian and insect cells for VLP production is expensive due to the high cost of media and poor scalability [58]. In contrast, yeast can quickly grow on simple media [59]. A yeast species, K. phaffii, can be grown to a high cell density on a commercial scale, which is not feasible with animal cells. The rapid growth of yeast cells (unlike animal cells, yeast cells grow faster with a doubling time of around 90–120 min, whereas animal cells have a doubling time of 16–18 h or more) is another advantage [60]. The growth of plants is slow and may take several weeks, months, or even years to reach the desired maturity. Another issue is the varying expression levels in different organs or tissues and more batch-to-batch variation. Other concerns include the possibility of escape into the natural environment [61]. The seasonal variation may severely impact plant growth, so the expression of the protein of interest remains an important consideration. Due to several advantages (as mentioned), yeast has been extensively used for expressing and purifying VLPs, especially S. cerevisiae (Table 2) and K. phaffii (Table 3).
Apart from the advantages mentioned above, yeast offers several other benefits, including a fully sequenced genome, the availability of vast genetic information, and molecular tools (in terms of vectors, promoters, and markers). Yeasts such as S. cerevisiae and K. phaffii are accessible and responsive in terms of genetic manipulation. Technology for introducing foreign genes is well established and therefore allows integration of the gene of interest at a precise location in the genome, contrary to animal or plant cells [60]. Under certain conditions, several copies of a gene can be introduced in the same cell, thereby increasing the yield per cell [122]. The yeast-based system also gives the option of constitutive and inducive expression. Compared to plant or animal cells, screening positive clones with a high yield is relatively fast and more economical in yeast [122]. Although in most of the studies, proteins of interest were expressed intracellularly, on several occasions, a protein expressed for VLP formation also gets secreted in culture media; this is another advantage as it is relatively easier to purify proteins from growth media [54]. Apart from S. cerevisiae and K. phaffii, several other yeast species have been tested for expression and purification of VLPs. (See Table 4 and Table 5).
Table 3. Studies where K. phaffii yeast was used to generate VLPs.
Table 3. Studies where K. phaffii yeast was used to generate VLPs.
S. NoProtein AntigenVirusProtein
Localization
PromoterReferences
1Capsid proteinRed-spotted grouper nervous necrosis virusICPw42-2[123]
2ZS and SZika virusICAOX1[124]
3112-608aa of the ORF2Hepatitis E virusECAOX1[125]
4P1 and CD3Poliovirus type IICAOX1[126,127]
5Chimeric HPV-HIV L1P18 proteinHPV and HIVICGAP[128]
6NY-ESO-1 cancer testis antigenNorovirusECAOX1[129]
7Surface antigenHepatitis C virusICAOX1[130,131]
8P1 and 3CDEnterovirus 71ECAOX1[132,133]
9E domain IIIDengue VirusICAOX1[134,135]
10P1 and 3CD Coxsackievirus A16ECGAP[136]
11VP1NorovirusECAOX1[137]
12E antigenDengue virusICAOX1[138,139,140,141,142,143]
13prMEJapanese encephalitis virus ECAOX1[144]
14E antigenDengue virusECGAP[145]
15Matrix proteinNipah virusICAOX1[146]
16P1 and 3CDEnterovirus D68ICAOX1[147]
17prM and E proteinTick-borne encephalitis virusECGAP[148]
18P1 and 3CDCoxsackievirus A10ICAOX1[149]
19Surface antigenHepatitis B virusICAOX1[150,151,152,153,154,155,156]
20prM and E proteinDengue virusICGAP[157]
21L1HPV16 and 18ICAOX1[158,159,160]
22L1HPV 52 AOX1[161]
23Capsid proteinCowpea chlorotic mottle virusECAOX1[162]
24L1HPV 58ICAOX1[163]
25Envelope protein domain III (EDIII), hepatitis B surface antigenDengue virusICAOX1[164]
26VP2Infectious bursal disease virusICAOX1[165]
27P1 and 3CDCoxsackievirus A16 ICAOX1[166]
28Core proteinHepatitis B virusICAOX1[167,168]
29E proteinDengue virus ICAOX1[169]
30L2Grapevine fanleaf virusECAOX1[170]
31Capsid protein (VP60)Rabbit hemorrhagic disease virusICAOX1[171]
32HBc-influenza virus LAH domainHepatitis B/Influenza H3N2 virusICAOX1[172]
33CoreE1E2 ProteinHepatitis C virusECAOX1[173]
34P1 and 3CDCoxsackievirus A6ICAOX1[174]
35L1, L2HPV 16ICAOX1[175]
36prM/EnvJapanese encephalitis virusICAOX1[176]
37Den2E-HBsAgDengue/Hepatitis B virusICAOX1[177]
38prM and E proteinDengue virusICGAP[178]
39PolyproteinChikungunya virusECAOX1[179]
40L1HPV 16 [180]
41Major capsid protein Iridovirus ECAOX1[181]
42Surface antigen Hepatitis B virus [182]
43VP1Rabbit hemorrhagic disease virusICAOX1[183]
44L1Bovine papillomavirus 1,2,4 [184]
45L1HPV 16ICAOX1[185]
46Capsid proteinNorovirusICAOX1[186]
47L1HPV 16ECPGK1[187]
48Core protein Hepatitis B virus [188]
49VP1Calicivirus virusECAOX1[189]
50Core protein Hepatitis C virusICAOX1[190,191,192,193]
IC: intracellular; EC: extracellular.

4. Bottleneck in the Use of a Yeast-Based System on a Commercial Scale

In the previous section, we discussed some salient features that make yeast a suitable host for the commercial production of VLPs. Indeed, many commercially available vaccines are produced using yeast (discussed in detail in the next section). Despite this, several issues limit the use of the yeast-based system for commercial expression and purification of VLPs. In this section, we will point out the reasons that hamper the full utilization of yeast-based platforms for the commercial production of VLPs. One of the critical issues with using yeast to produce proteins (which will be used as vaccines) is the inability of yeast cells to perform protein glycosylation like humans [207].
Further, proteins in yeast cells are, in general, more heavily glycosylated compared to animal cells, including human cells [208]. Additionally, the glycosylation pattern differs in humans and yeast, which may have significant implications in terms of the antigenicity, solubility, and stability of proteins, especially when injected into the body [209]. However, this issue can be addressed to a certain extent by treating purified proteins with deglycosylase enzymes or modifying the primary amino acid sequence where possible. Apart from this, efforts are made to engineer yeast cells (through pathway modification) to minimize the problem of heavy mannosylation of proteins. For example, the glycoengineered strain of K. phaffii from Biogrammatics showed a significant reduction in glycosylation of expressed proteins (https://www.biogrammatics.com (accessed on 17 December 2022).
Another issue limiting the use of yeast for producing VLPs is that many viruses are enveloped. To date, only one study has shown the possibility of producing enveloped VLPs (eVLPs) using yeast as a host [210]. The success of producing eVLPs using the yeast-based system on an industrial scale remains to be determined. Indeed, this is where animal cells have advantages over yeast-based systems; however, future research can help manipulate cell membranes and may open the way for the secretion of eVLPs in yeast.

5. In Vivo VLP Assembly and Secretion

Secretion of fully assembled VLPs into culture media is desirable as it enables an economical and straightforward purification of VLPs from the host system. So far, insects and mammalian cells are known to secrete fully assembled VLPs into culture media [45,211]. This feature is especially important when one is working with eVLPs. However, the high media cost and level to which scale-up is possible with animal cell lines remain significant concerns (reviewed in [212]). S. cerevisiae and K. phaffii are known to grow on cheap media and can grow well in big industrial vessels (especially K. phaffii); however, the secretion of fully assembled VLPs into culture media is possible in a few cases. (Table 2, Table 3, Table 4 and Table 5). Even before secretion, it is important to see whether VLPs can be assembled within yeast cells in the cytoplasm. Several studies in the past have already shown the in vivo assembly of VLPs followed by their secretion [129,144,145,148,179,210]. Again, this showed the advantage of yeast over the bacterial system. Based on published studies, some VLPs get secreted into the media while others are not (Table 2, Table 3, Table 4 and Table 5). Secretion of VLPs depends upon the proteins used for VLP generation and VLP size. Therefore, one must check the possibility of secretion of VLPs for each virus and protein used.

6. Aggregation of VLPs, a Matter of Concern

The production of VLPs on a commercial scale using the different hosts mentioned above is now becoming common practice. Several companies dealing in vaccines or drugs are producing VLPs commercially. Almost all VLP production suffers from the common issue of VLP aggregation when stored at a low-salt concentration and at 2–8 °C [213,214,215,216,217]. Dynamic Light Scattering (DLS), Transmission Electron Microscopes (TEM), and Atomic Force Microscopes (AFM) can detect the aggregation of VLPs, approaches commonly used for VLP characterization (reviewed by [28]). In addition, turbidity during elution or storage can also inform about possible VLP aggregation. The main problem with the aggregation of VLPs is that it affects immunogenicity and the level of an immune response. Aggregation of VLPs reduces their recovery using POROS resin used for the purification of VLPs on a commercial scale [111]. It was observed that the immune response raised by aggregated VLPs is lower than that raised by non-aggregated VLPs [213,218,219,220,221]. Additionally, aggregation can affect dose formulation. To obtain highly monodisperse VLPs, the manufacturer performs an ultracentrifuge or size exclusion to separate clean and aggregated VLPs. However, this leads to considerable wastage of VLPs and reduces the recovery of useful VLPs.
To prevent or minimize the aggregation of VLPs, different approaches have been used in the past. For example, adding L-arginine and glycine improves VLP recovery [221,222]. Sugars like sorbitol and trehalose are also shown to solubilize and improve VLP recovery by preventing their aggregation and by solubilizing protein monomers [210,212]. The use of polyethylene glycol (PEG) or glycerol also improves the solubilization of VLPs [223,224]. Apart from this, using surfactants or emulsifiers like Tween-80 is also helpful in preventing the aggregation of VLPs and improving the recovery of VLPs [225]. In several published studies, the author has added these compounds at one or another step in the purification of VLPs. Sometimes changes in the pH of the buffer and the buffer composition were also found helpful in preventing the aggregation of VLPs [226]. Additionally, to obtain VLPs of a uniform size, in vitro disassembly followed by reassembly is recommended [227]. Therefore, it requires substantial effort to find suitable conditions that improve the recovery of VLPs while simultaneously preventing their aggregation.

7. Success Story

In previous sections, we provided a descriptive background of VLPs, various hosts available for their production, issues, and the suitability of yeast to produce VLPs. We also mentioned one of the most common problems encountered in the purification of VLPs. Now is the time to discuss some cases where VLPs have been used successfully as a vaccine or drug delivery system. Although we will mainly focus on VLPs produced using yeast that are currently on the market for public use, it is essential to mention that VLPs purified from other systems are also floating on the market [22].
The first VLP-based vaccine to hit the market was Gardasil, followed by Gardasil9; both products were branded under Merck and were against HPV-induced cancer. GlaxoSmithKline introduced another VLP-based vaccine (Cervarix) against HPV. It is important to note that Cervarix was effective against both the HPV16 and HPV18 viral strains. Cervarix from GlaxoSmithKline was taken off the market (USA) in 2016, while Gardasil and Gardasil9 are still available for public use. Notably, Gardasil and Gardasil9 are from yeast, while Cervarix is from the Baculovirus expression system, which uses High Five Rix4446 cells derived from the insect Trichoplusia ni. Apart from the two vaccines (from yeast) mentioned, another vaccine, Mosquirix against Plasmodium falciparum, responsible for malaria, is commercially approved [228].
Above, we mentioned the VLP-based vaccines currently in public use. Apart from this, several VLP-based vaccines are under clinical trials. An example vaccine against HIV is under clinical trial [43]. Apart from yeast-derived VLPs, several VLP-based vaccines derived from non-yeast hosts are also in public use or clinical trials [22]. Table 6 compiles the list of VLPs produced in yeast currently in use and those in different phases of clinical trials.

8. Conclusions and Future Direction

Based on the points discussed above, during the last few years, VLPs have gained significant importance and acceptance as a means of vaccine and drug delivery. Apart from their use in a vaccine, VLPs are also noted for their potential use in drug delivery (chemical salts or small molecules) as well as for the delivery of nucleic acids (DNA/RNA) [22,229,230,231,232,233]. This approach to drug delivery offers the advantage of being biodegradable (unlike metal-based nanoparticles). Several studies from different labs have already showed proof of concept where VLPs were used to deliver drugs and nucleic acids [22]. Because they resemble viruses, VLPs can be used to study virus-host interactions and virus internalization by host cells. This aspect can be a game changer as it will eliminate or minimize the use of handling infectious entities. Since VLPs can be obtained from companies, these can be ordered as standard reagents (like antibodies), which can speed up research activity (Figure 2). For example, Advance ImmunoChemical Inn supplies HPV18 L1 VLPs (https://www.advimmuno.com).
Interestingly, more and more VLPs are now expressed in yeast. Apart from S. cerevisiae and K. phaffii, other yeast species, including H. polymorpha, species of Kluyveromyces, Y. lipolytica, X. dendrorhous, and S. pombe, have also been used for VLP expression and purification. Additionally, over the years, more and more studies have appeared in which K. phaffii was used to produce VLPs. Although eVLPs have yet to be produced using yeast (on a commercial scale), this remains one of the concerns with using yeast for VLP production. Since considerable effort and resources are used during the purification of VLPs, the secretion of fully assembled VLPs into culture media is ideal, making the purification of VLPs economical. The secretion of VLPs is common in the case of animal cells, but in the case of yeast, only a few studies are available where VLPs have been successfully secreted into media [64,125,144,145,189]. Although yeast species other than S. cerevisiae and K. phaffii are also used for producing VLPs, whether using those species on an industrial scale is feasible remains to be determined. VLPs purified from S. cerevisiae are already approved by the FDA and are currently in public use. It will be interesting to see when we will have VLPs purified from K. phaffii, which is FDA-approved and available for public use. It is anticipated that we may soon have a VLP-based vaccine where K. phaffii is used as a host. Preclinical evaluation of HBc VLPs (purified from K. phaffii) against hepatocellular carcinoma is underway [234,235]. This is important as more and more VLPs are purified using K. phaffii. Since K. phaffii can grow to high cell densities on cheap media, the use of VLPs purified from K. phaffii may also reduce the cost of VLP-based vaccines.
On a commercial scale, the purification of VLPs uses a chromatography column (such as POROS HS50). These columns get clogged by yeast lipids (due to the irreversible binding of lipids to resin) and lipid droplets. Therefore, to prevent or minimize any such issue, it is essential to use yeast strains that do not form lipid droplets [236]. In most of the studies using K. phaffii, the authors used the GS115 strain. However, using protease-deficient strains (for example, SMD1163) to minimize the degradation of an expressed protein is recommended [24]. Intracellular degradation of an expressed protein can affect the assembly and recovery of VLPs.
In the end, several hosts are available for the production of VLPs, each offering some advantages over the others. Certainly, yeast has some significant benefits as a host for the commercial production of VLPs compared to other available hosts. However, the inability of yeast to secrete VLPs of important viruses remains a critical bottleneck. Therefore, more work in strain engineering will be needed to fully tap the potential of yeast for the production of VLPs. It may involve host engineering, antigen engineering, modification of culture media, and buffers, thus representing several channelings and opportunities.

Author Contributions

Conceptualization, R.K.; manuscript writing—draft, including preparation of tables and figures, V.S., K.N.N. and A.A.; draft manuscript writing—editing and revision, R.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

R.K. is grateful to RPI Troy, NY, for providing space and other necessary facilities to complete the draft.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Zhu, S.; Guo, C. Rabies control and treatment: From prophylaxis to strategies with curative potential. Viruses 2016, 8, 279. [Google Scholar] [CrossRef] [PubMed]
  2. Kocourkova, A.; Honegr, J.; Kuca, K.; Danova, J. Vaccine Ingredients: Components that Influence Vaccine Efficacy. Mini-Rev. Med. Chem. 2017, 17, 451–466. [Google Scholar] [CrossRef] [PubMed]
  3. Scollard, D.M.; Adams, L.B.; Gillis, T.P.; Krahenbuhl, J.L.; Truman, R.W.; Williams, D.L. The continuing challenges of leprosy. Clin. Microbiol. Rev. 2006, 19, 338–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Cox, F.E. Malaria vaccines—Progress and problems. Trends Biotechnol. 1991, 9, 389–394. [Google Scholar] [CrossRef]
  5. Raffatellu, M.; Chessa, D.; Wilson, R.P.; Dusold, R.; Rubino, S.; Bäumler, A.J. The Vi capsular antigen of Salmonella enterica serotype Typhi reduces Toll-like receptor-dependent interleukin-8 expression in the intestinal mucosa. Infect. Immun. 2005, 73, 3367–3374. [Google Scholar] [CrossRef] [Green Version]
  6. Plotkin, S.A. Rubella vaccine. In Vaccines, 3rd ed.; Plotkin, S.A., Orenstein, W.A., Eds.; WB Saunders Co.: Philadelphia, PA, USA, 1998; pp. 409–439. [Google Scholar]
  7. Vogel, F.R.; Sarver, N. Nucleic acid vaccines. Clin. Microbiol. Rev. 1995, 8, 406–410. [Google Scholar] [CrossRef]
  8. Restifo, N.P.; Ying, H.; Hwang, L.; Leitner, W.W. The promise of nucleic acid vaccines. Gene Ther. 2000, 7, 89–92. [Google Scholar] [CrossRef] [Green Version]
  9. Pardi, N.; Hogan, M.J.; Porter, F.W.; Weissman, D. mRNA vaccines—A new era in vaccinology. Nat. Rev. Drug Discov. 2018, 17, 261–279. [Google Scholar] [CrossRef] [Green Version]
  10. Schlake, T.; Thess, A.; Fotin-Mleczek, M.; Kallen, K.J. Developing mRNA-vaccine technologies. RNA Biol. 2012, 9, 1319–1330. [Google Scholar] [CrossRef] [Green Version]
  11. Qin, S.; Tang, X.; Chen, Y.; Chen, K.; Fan, N.; Xiao, W.; Zheng, Q.; Li, G.; Teng, Y.; Wu, M.; et al. mRNA-based therapeutics: Powerful and versatile tools to combat diseases. Signal Transduct. Target. Ther. 2022, 7, 166. [Google Scholar] [CrossRef]
  12. Kaur, S.P.; Gupta, V. COVID-19 Vaccine: A comprehensive status report. Virus Res. 2020, 288, 198114. [Google Scholar] [CrossRef] [PubMed]
  13. Callaway, E. The race for coronavirus vaccines: A graphical guide. Nature 2020, 580, 576–577. [Google Scholar] [CrossRef] [PubMed]
  14. Kumar, R.; Srivastava, V.; Baindara, P.; Ahmad, A. Thermostable vaccines: An innovative concept in vaccine development. Expert Rev. Vaccines 2022, 21, 811–824. [Google Scholar] [CrossRef] [PubMed]
  15. McLaughlin-Drubin, M.E.; Munger, K. Viruses associated with human cancer. Biochim. Biophys. Acta 2008, 1782, 127–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Schlom, J.; Hodge, J.W.; Palena, C.; Tsang, K.Y.; Jochems, C.; Greiner, J.W.; Farsaci, B.; Madan, R.A.; Heery, C.R.; Gulley, J.L. Therapeutic cancer vaccines. Adv. Cancer Res. 2014, 121, 67–124. [Google Scholar] [PubMed]
  17. Cuzick, J. Gardasil 9 joins the fight against cervix cancer. Expert Rev. Vaccines 2015, 14, 1047–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Pouyanfard, S.; Spagnoli, G.; Bulli, L.; Balz, K.; Yang, F.; Odenwald, C.; Seitz, H.; Mariz, F.C.; Bolchi, A.; Ottonello, S.; et al. Minor Capsid Protein L2 Polytope Induces Broad Protection against Oncogenic and Mucosal Human Papillomaviruses. J. Virol. 2018, 92, e01930-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Rolih, V.; Caldeira, J.; Bolli, E.; Salameh, A.; Conti, L.; Barutello, G.; Riccardo, F.; Magri, J.; Lamolinara, A.; Parra, K.; et al. Development of a VLP-Based Vaccine Displaying an xCT Extracellular Domain for the Treatment of Metastatic Breast Cancer. Cancers 2020, 12, 1492. [Google Scholar] [CrossRef]
  20. Zhang, S.; Yong, L.K.; Li, D.; Cubas, R.; Chen, C.; Yao, Q. Mesothelin virus-like particle immunization controls pancreatic cancer growth through CD8+ T cell induction and reduction in the frequency of CD4+foxp3+ICOS regulatory T cells. PLoS ONE 2013, 8, e68303. [Google Scholar] [CrossRef]
  21. Cubas, R.; Zhang, S.; Li, M.; Chen, C.; Yao, Q. Chimeric Trop2 virus-like particles: A potential immunotherapeutic approach against pancreatic cancer. J. Immunother. 2011, 34, 251–263. [Google Scholar] [CrossRef]
  22. Nooraei, S.; Bahrulolum, H.; Hoseini, Z.S.; Katalani, C.; Hajizade, A.; Easton, A.J.; Ahmadian, G. Virus-like particles: Preparation, immunogenicity and their roles as nanovaccines and drug nanocarriers. J. Nanobiotechnol. 2021, 19, 59. [Google Scholar] [CrossRef] [PubMed]
  23. Draper, S.J.; Sack, B.K.; King, C.R.; Nielsen, C.M.; Rayner, J.C.; Higgins, M.K.; Long, C.A.; Seder, R.A. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe 2018, 24, 43–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Kumar, R.; Kharbikar, B.N. Lyophilized yeast powder for adjuvant free thermostable vaccine delivery. Appl. Microbiol. Biotechnol. 2021, 105, 3131–3143. [Google Scholar] [CrossRef] [PubMed]
  25. Kumar, R. Investigating the long-term stability of protein immunogen(s) for whole recombinant yeast-based vaccines. FEMS Yeast Res. 2018, 18, foy071. [Google Scholar] [CrossRef] [Green Version]
  26. Bayer, M.E.; Blumberg, B.S.; Werner, B. Particles associated with Australia antigen in the sera of patients with leukaemia, Down’s Syndrome and Hepatitis. Nature 1968, 218, 1057–1059. [Google Scholar] [CrossRef]
  27. Grgacic, E.V.; Anderson, D.A. Virus-like particles: Passport to immune recognition. Methods 2006, 40, 60–65. [Google Scholar] [CrossRef]
  28. Mejía-Méndez, J.L.; Vazquez-Duhalt, R.; Hernández, L.R.; Sánchez-Arreola, E.; Bach, H. Virus-like Particles: Fundamentals and Biomedical Applications. Int. J. Mol. Sci. 2022, 23, 8579. [Google Scholar] [CrossRef]
  29. Lan, K.; Luo, M.H. Herpesviruses: Epidemiology, pathogenesis, and interventions. Virol. Sin. 2017, 32, 347–348. [Google Scholar] [CrossRef] [Green Version]
  30. Le, D.T.; Müller, K.M. In Vitro Assembly of Virus-Like Particles and Their Applications. Life 2021, 11, 334. [Google Scholar] [CrossRef]
  31. Jeong, H.; Seong, B.L. Exploiting virus-like particles as innovative vaccines against emerging viral infections. J. Microbiol. 2017, 55, 220–230. [Google Scholar] [CrossRef]
  32. Mortola, E.; Roy, P. Efficient assembly and release of SARS coronavirus-like particles by a heterologous expression system. FEBS Lett. 2004, 576, 174–178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Pushko, P.; Pumpens, P.; Grens, E. Development of virus-like particle technology from small highly symmetric to large complex virus-like particle structures. Intervirology 2013, 56, 141–165. [Google Scholar] [CrossRef] [PubMed]
  34. Latham, T.; Galarza, J.M. Formation of wild-type and chimeric influenza virus-like particles following simultaneous expression of only four structural proteins. J. Virol. 2001, 75, 6154–6165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Gedvilaite, A.; Kucinskaite-Kodze, I.; Lasickiene, R.; Timinskas, A.; Vaitiekaite, A.; Ziogiene, D.; Zvirbliene, A. Evaluation of Trichodysplasia Spinulosa-Associated Polyomavirus Capsid Protein as a New Carrier for Construction of Chimeric Virus-Like Particles Harboring Foreign Epitopes. Viruses 2015, 7, 4204–4229. [Google Scholar] [CrossRef] [Green Version]
  36. Bai, B.; Hu, Q.; Hu, H.; Zhou, P.; Shi, Z.; Meng, J.; Lu, B.; Huang, Y.; Mao, P.; Wang, H. Virus-like particles of SARS-like coronavirus formed by membrane proteins from different origins demonstrate stimulating activity in human dendritic cells. PLoS ONE 2008, 3, e2685. [Google Scholar] [CrossRef] [Green Version]
  37. Chung, Y.H.; Cai, H.; Steinmetz, N.F. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv. Drug Deliv. Rev. 2020, 156, 214–235. [Google Scholar] [CrossRef]
  38. Steinmetz, N.F. Viral nanoparticles as platforms for next-generation therapeutics and imaging devices. Nanomedicine 2010, 6, 634–641. [Google Scholar] [CrossRef] [Green Version]
  39. Mohsen, M.O.; Gomes, A.C.; Vogel, M.; Bachmann, M.F. Interaction of viral capsid-derived virus-like particles (VLPs) with the innate immune system. Vaccines 2018, 6, 37. [Google Scholar] [CrossRef] [Green Version]
  40. Silva, A.L.; Peres, C.; Conniot, J.; Matos, A.I.; Moura, L.; Carreira, B.; Sainz, V.; Scomparin, A.; Satchi-Fainaro, R.; Préat, V.; et al. Nanoparticle impact on innate immune cell pattern-recognition receptors and inflammasomes activation. Semin. Immunol. 2017, 34, 3–24. [Google Scholar] [CrossRef]
  41. Lee, Y.T.; Ko, E.J.; Lee, Y.; Kim, K.H.; Kim, M.C.; Lee, Y.N.; Kang, S.M. Intranasal vaccination with M2e5x virus-like particles induces humoral and cellular immune responses conferring cross-protection against heterosubtypic influenza viruses. PLoS ONE 2018, 13, e0190868. [Google Scholar] [CrossRef] [Green Version]
  42. Wang, C.; Zheng, X.; Gai, W.; Wong, G.; Wang, H.; Jin, H.; Feng, N.; Zhao, Y.; Zhang, W.; Li, N.; et al. Novel chimeric virus-like particles vaccine displaying MERS-CoV receptor-binding domain induce specific humoral and cellular immune response in mice. Antiviral Res. 2017, 140, 55–61. [Google Scholar] [CrossRef] [PubMed]
  43. Weber, J.; Cheinsong-Popov, R.; Callow, D.; Adams, S.; Patou, G.; Hodgkin, K.; Martin, S.; Gotch, F.; Kingsman, A. Immunogenicity of the yeast recombinant p17p24: Ty virus-like particles (p24-VLP) in healthy volunteers. Vaccine 1995, 13, 831–834. [Google Scholar] [CrossRef] [PubMed]
  44. Yong, C.Y.; Yeap, S.K.; Goh, Z.H.; Ho, K.L.; Omar, A.R.; Tan, W.S. Induction of humoral and cell-mediated immune responses by hepatitis B virus epitope displayed on the virus-like particles of prawn nodavirus. Appl. Environ. Microbiol. 2015, 81, 882–889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Ye, L.; Lin, J.; Sun, Y.; Bennouna, S.; Lo, M.; Wu, Q.; Bu, Z.; Pulendran, B.; Compans, R.W.; Yang, C. Ebola virus-like particles produced in insect cells exhibit dendritic cell stimulating activity and induce neutralizing antibodies. Virology 2006, 351, 260–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Available online: https://www.gavi.org/vaccineswork/what-are-protein-subunit-vaccines-and-how-could-they-be-used-against-covid-19 (accessed on 10 November 2022).
  47. Sailaja, G.; Skountzou, I.; Quan, F.S.; Compans, R.W.; Kang, S.M. Human immunodeficiency virus-like particles activate multiple types of immune cells. Virology 2007, 362, 331–341. [Google Scholar] [CrossRef] [Green Version]
  48. Le, D.T.; Radukic, M.T.; Mller, K.M. Adeno-associated virus capsid protein expression in Escherichia coli and chemically defined capsid assembly. Sci. Rep. 2019, 9, 18631. [Google Scholar] [CrossRef] [Green Version]
  49. Joe, C.C.; Chatterjee, S.; Lovrecz, G.; Adams, T.E.; Thaysen-Andersen, M.; Walsh, R.; Locarnini, S.A.; Smooker, P.; Netter, H.J. Glycoengineered hepatitis B virus-like particles with enhanced immunogenicity. Vaccine 2020, 38, 3892–3901. [Google Scholar] [CrossRef]
  50. Shiri, F.; Petersen, K.E.; Romanov, V.; Zou, Q.; Gale, B.K. Characterization and differential retention of Q beta bacteriophage virus-like particles using cyclical electrical field–flow fractionation and asymmetrical flow fieldflow fractionation. Anal. Bioanal. Chem. 2020, 412, 1563–1572. [Google Scholar] [CrossRef]
  51. Zhai, L.; Yadav, R.; Kunda, N.K.; Anderson, D.; Bruckner, E.; Miller, E.K.; Basu, R.; Muttil, P.; Tumban, E. Oral immunization with bacteriophage MS2-L2 VLPs protects against oral and genital infection with multiple HPV types associated with head & neck cancers and cervical cancer. Antiviral Res. 2019, 166, 56–65. [Google Scholar]
  52. Glass, P.J.; White, L.J.; Ball, J.M.; Leparc-Goffart, I.; Hardy, M.E.; Estes, M.K. Norwalk virus open reading frame 3 encodes a minor structural protein. J. Virol. 2000, 74, 6581–6591. [Google Scholar] [CrossRef] [Green Version]
  53. Donaldson, B.; Al-Barwani, F.; Young, V.; Scullion, S.; Ward, V.; Young, S. Virus-Like Particles, a Versatile Subunit Vaccine Platform. In Subunit Vaccine Delivery; Part of the Advances in Delivery Science and Technology Book Series; Springer: New York, NY, USA, 2014; pp. 159–180. [Google Scholar]
  54. Kim, H.J.; Kim, H.J. Yeast as an expression system for producing virus-like particles: What factors do we need to consider? Lett. Appl. Microbiol. 2017, 64, 111–123. [Google Scholar] [CrossRef] [PubMed]
  55. Kumar, R.; Kumar, P. Yeast-based vaccines: New perspective in vaccine development and application. FEMS Yeast Res. 2019, 19, foz007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Magalhães, P.O.; Lopes, A.M.; Mazzola, P.G.; Rangel-Yagui, C.; Penna, T.C.; Pessoa, A., Jr. Methods of endotoxin removal from biological preparations: A review. J. Pharm. Pharm. Sci. 2007, 10, 388–404. [Google Scholar] [PubMed]
  57. Baneyx, F.; Mujacic, M. Recombinant protein folding and misfolding in Escherichia coli. Nat. Biotechnol. 2004, 22, 1399–1408. [Google Scholar] [CrossRef] [PubMed]
  58. Dalton, A.C.; Barton, W.A. Over-expression of secreted proteins from mammalian cell lines. Protein Sci. 2014, 23, 517–525. [Google Scholar] [CrossRef]
  59. Bill, R.M. Recombinant protein subunit vaccine synthesis in microbes: A role for yeast? J. Pharm. Pharmacol. 2015, 67, 319–328. [Google Scholar] [CrossRef] [Green Version]
  60. Karbalaei, M.; Rezaee, S.A.; Farsiani, H. Pichia pastoris: A highly successful expression system for optimal synthesis of heterologous proteins. J. Cell. Physiol. 2020, 235, 5867–5881. [Google Scholar] [CrossRef]
  61. Desai, P.N.; Shrivastava, N.; Padh, H. Production of heterologous proteins in plants: Strategies for optimal expression. Biotechnol. Adv. 2010, 28, 427–435. [Google Scholar] [CrossRef]
  62. Lazutka, J.; Simutis, K.; Matulis, P.; Petraitytė-Burneikienė, R.; Kučinskaitė-Kodzė, I.; Simanavičius, M.; Tamošiunas, P.L. Antigenicity study of the yeast-generated human parvovirus 4 (PARV4) virus-like particles. Virus Res. 2021, 292, 198236. [Google Scholar] [CrossRef]
  63. Simanavicius, M.; Tamosiunas, P.L.; Petraityte-Burneikiene, R.; Johne, R.; Ulrich, R.G.; Zvirbliene, A.; Kucinskaite-Kodze, I. Generation in yeast and antigenic characterization of hepatitis E virus capsid protein virus-like particles. Appl. Microbiol. Biotechnol. 2018, 102, 185–198. [Google Scholar] [CrossRef]
  64. Chen, P.; Zhang, L.; Chang, N.; Shi, P.; Gao, T.; Zhang, L.; Huang, J. Preparation of virus-like particles for porcine circovirus type 2 by Yeast Fab Assembly. Virus Genes 2018, 54, 246–255. [Google Scholar] [CrossRef] [PubMed]
  65. Juozapaitis, M.; Slibinskas, R.; Staniulis, J.; Sakaguchi, T.; Sasnauskas, K. Generation of Sendai virus nucleocapsid-like particles in yeast. Virus Res. 2005, 108, 221–224. [Google Scholar] [CrossRef] [PubMed]
  66. Tamošiūnas, P.L.; Petraitytė-Burneikienė, R.; Bulavaitė, A.; Marcinkevičiūtė, K.; Simutis, K.; Lasickienė, R.; Firantienė, R.; Ėmužytė, R.; Žvirblienė, A.; Sasnauskas, K. Yeast-generated virus-like particles as antigens for detection of human bocavirus 1-4 specific antibodies in human serum. Appl. Microbiol. Biotechnol. 2016, 100, 4935–4946. [Google Scholar] [CrossRef] [PubMed]
  67. Burden, C.S.; Jin, J.; Podgornik, A.; Bracewell, D.G. A monolith purification process for virus-like particles from yeast homogenate. J. Chromatogr. B 2012, 880, 82–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kee, G.S.; Jin, J.; Balasundaram, B.; Bracewell, D.G.; Pujar, N.S.; Titchener-Hooker, N.J. Exploiting the intracellular compartmentalization characteristics of the S. cerevisiae host cell for enhancing primary purification of lipid-envelope virus-like particles. Biotechnol. Prog. 2010, 26, 26–33. [Google Scholar] [PubMed]
  69. Kee, G.S.; Pujar, N.S.; Titchener-Hooker, N.J. Study of detergent-mediated liberation of hepatitis B virus-like particles from S. cerevisiae homogenate: Identifying a framework for the design of future-generation lipoprotein vaccine processes. Biotechnol. Prog. 2008, 24, 623–631. [Google Scholar] [CrossRef] [PubMed]
  70. Tsunetsugu-Yokota, Y.; Morikawa, Y.; Isogai, M.; Kawana-Tachikawa, A.; Odawara, T.; Nakamura, T.; Grassi, F.; Autran, B.; Iwamoto, A. Yeast-derived human immunodeficiency virus type 1 p55(gag) virus-like particles activate dendritic cells (DCs) and induce perforin expression in Gag-specific CD8+ T cells by cross-presentation of DCs. J. Virol. 2003, 77, 10250–10259. [Google Scholar] [CrossRef] [Green Version]
  71. Norkiene, M.; Stonyte, J.; Ziogiene, D.; Mazeike, E.; Sasnauskas, K.; Gedvilaite, A. Production of recombinant VP1-derived virus-like particles from novel human polyomaviruses in yeast. BMC Biotechnol. 2015, 15, 68. [Google Scholar] [CrossRef] [Green Version]
  72. Kim, H.J.; Cho, S.Y.; Park, M.H.; Kim, H.J. Comparison of the size distributions and immunogenicity of human papillomavirus type 16 L1 virus-like particles produced in insect and yeast cells. Arch. Pharm. Res. 2018, 41, 544–553. [Google Scholar] [CrossRef]
  73. Kim, H.J.; Jin, Y.; Kim, H.J. The concentration of carbon source in the medium affects the quality of virus-like particles of human papillomavirus type 16 produced in Saccharomyces cerevisiae. PLoS ONE 2014, 9, e94467. [Google Scholar] [CrossRef] [Green Version]
  74. Petraityte, R.; Tamosiunas, P.L.; Juozapaitis, M.; Zvirbliene, A.; Sasnauskas, K.; Shiell, B.; Russell, G.; Bingham, J.; Michalski, W.P. Generation of Tioman virus nucleocapsid-like particles in yeast Saccharomyces cerevisiae. Virus Res. 2009, 145, 92–96. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, H.L.; Chen, P.J.; Mu, J.J.; Chi, W.K.; Kao, T.L.; Hwang, L.H.; Chen, D.S. Assembly of hepatitis delta virus-like empty particles in yeast. Virology 1997, 236, 374–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Nainys, J.; Lasickiene, R.; Petraityte-Burneikiene, R.; Dabrisius, J.; Lelesius, R.; Sereika, V.; Zvirbliene, A.; Sasnauskas, K.; Gedvilaite, A. Generation in yeast of recombinant virus-like particles of porcine circovirus type 2 capsid protein and their use for a serologic assay and development of monoclonal antibodies. BMC Biotechnol. 2014, 14, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, X.; Xiao, X.; Zhao, M.; Liu, W.; Pang, L.; Sun, X.; Cen, S.; Yang, B.B.; Huang, Y.; Sheng, W.; et al. 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] [Green Version]
  78. Sasnauskas, K.; Bulavaite, A.; Hale, A.; Jin, L.; Knowles, W.A.; Gedvilaite, A.; Dargeviciūte, A.; Bartkeviciūte, D.; Zvirbliene, A.; Staniulis, J.; et al. Generation of recombinant virus-like particles of human and non-human polyomaviruses in yeast Saccharomyces cerevisiae. Intervirology 2002, 45, 308–317. [Google Scholar] [CrossRef] [PubMed]
  79. Backovic, A.; Cervelli, T.; Salvetti, A.; Zentilin, L.; Giacca, M.; Galli, A. Capsid protein expression and adeno-associated virus like particles assembly in Saccharomyces cerevisiae. Microb. Cell Fact. 2012, 11, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Bulavaitė, A.; Lasickienė, R.; Tamošiūnas, P.L.; Simanavičius, M.; Sasnauskas, K.; Žvirblienė, A. Synthesis of human parainfluenza virus 4 nucleocapsid-like particles in yeast and their use for detection of virus-specific antibodies in human serum. Appl. Microbiol. Biotechnol. 2017, 101, 2991–3004. [Google Scholar] [CrossRef] [PubMed]
  81. Freivalds, J.; Dislers, A.; Ose, V.; Skrastina, D.; Cielens, I.; Pumpens, P.; Sasnauskas, K.; Kazaks, A. Assembly of bacteriophage Qbeta virus-like particles in yeast Saccharomyces cerevisiae and Pichia pastoris. J. Biotechnol. 2006, 123, 297–303. [Google Scholar] [CrossRef] [PubMed]
  82. Wi, G.R.; Hwang, J.Y.; Kwon, M.G.; Kim, H.J.; Kang, H.A.; Kim, H.J. Protective immunity against nervous necrosis virus in convict grouper Epinephelus septemfasciatus following vaccination with virus-like particles produced in yeast Saccharomyces cerevisiae. Vet. Microbiol. 2015, 177, 214–218. [Google Scholar] [CrossRef]
  83. 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]
  84. 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]
  85. Zielonka, A.; Gedvilaite, A.; Reetz, J.; Rösler, U.; Müller, H.; Johne, R. Serological cross-reactions between four polyomaviruses of birds using virus-like particles expressed in yeast. J. Gen. Virol. 2012, 93, 2658–2667. [Google Scholar] [CrossRef] [PubMed]
  86. Zhao, H.; Li, H.Y.; Han, J.F.; Deng, Y.Q.; Zhu, S.Y.; Li, X.F.; Yang, H.Q.; Li, Y.X.; Zhang, Y.; Qin, E.D.; et al. Novel recombinant chimeric virus-like particle is immunogenic and protective against both enterovirus 71 and coxsackievirus A16 in mice. Sci. Rep. 2015, 5, 7878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Bucarey, S.A.; Noriega, J.; Reyes, P.; Tapia, C.; Sáenz, L.; Zuñiga, A.; Tobar, J.A. The optimized capsid gene of porcine circovirus type 2 expressed in yeast forms virus-like particles and elicits antibody responses in mice fed with recombinant yeast extracts. Vaccine 2009, 27, 5781–5790. [Google Scholar] [CrossRef]
  88. Rodríguez-Limas, W.A.; Tyo, K.E.; Nielsen, J.; Ramírez, O.T.; Palomares, L.A. Molecular and process design for rotavirus-like particle production in Saccharomyces cerevisiae. Microb. Cell Fact. 2011, 10, 33. [Google Scholar] [CrossRef] [Green Version]
  89. Rodríguez-Limas, W.A.; Pastor, A.R.; Esquivel-Soto, E.; Esquivel-Guadarrama, F.; Ramírez, O.T.; Palomares, L.A. Immunogenicity and protective efficacy of yeast extracts containing rotavirus-like particles: A potential veterinary vaccine. Vaccine 2014, 32, 2794–2798. [Google Scholar] [CrossRef]
  90. Tamošiūnas, P.L.; Simutis, K.; Kodzė, I.; Firantienė, R.; Emužytė, R.; Petraitytė-Burneikienė, R.; Zvirblienė, A.; Sasnauskas, K. Production of human parvovirus 4 VP2 virus-like particles in yeast and their evaluation as an antigen for detection of virus-specific antibodies in human serum. Intervirology 2013, 56, 271–277. [Google Scholar] [CrossRef]
  91. Bulavaitė, A.; Lasickienė, R.; Vaitiekaitė, A.; Sasnauskas, K.; Žvirblienė, A. Synthesis of human parainfluenza virus 2 nucleocapsid protein in yeast as nucleocapsid-like particles and investigation of its antigenic structure. Appl. Microbiol. Biotechnol. 2016, 100, 4523–4534. [Google Scholar] [CrossRef]
  92. Juozapaitis, M.; Serva, A.; Kucinskaite, I.; Zvirbliene, A.; Slibinskas, R.; Staniulis, J.; Sasnauskas, K.; Shiell, B.J.; Bowden, T.R.; Michalski, W.P. Generation of menangle virus nucleocapsid-like particles in yeast Saccharomyces cerevisiae. J. Biotechnol. 2007, 130, 441–447. [Google Scholar] [CrossRef]
  93. Tomo, N.; Goto, T.; Morikawa, Y. Trans-packaging of human immunodeficiency virus type 1 genome into Gag virus-like particles in Saccharomyces cerevisiae. Microb. Cell Fact. 2013, 12, 28. [Google Scholar] [CrossRef] [Green Version]
  94. Tamošiūnas, P.L.; Petraitytė-Burneikienė, R.; Lasickienė, R.; Akatov, A.; Kundrotas, G.; Sereika, V.; Lelešius, R.; Žvirblienė, A.; Sasnauskas, K. Generation of recombinant porcine parvovirus virus-like particles in Saccharomyces cerevisiae and development of virus-specific monoclonal antibodies. J. Immunol. Res. 2014, 2014, 573531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. 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]
  96. Zaveckas, M.; Snipaitis, S.; Pesliakas, H.; Nainys, J.; Gedvilaite, A. Purification of recombinant virus-like particles of porcine circovirus type 2 capsid protein using ion-exchange monolith chromatography. J. Chromatogr. B 2015, 991, 21–28. [Google Scholar] [CrossRef] [PubMed]
  97. 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] [Green Version]
  98. Sasnauskas, K.; Buzaite, O.; Vogel, F.; Jandrig, B.; Razanskas, R.; Staniulis, J.; Scherneck, S.; Krüger, D.H.; Ulrich, R. Yeast cells allow high-level expression and formation of polyomavirus-like particles. Biol. Chem. 1999, 380, 381–386. [Google Scholar] [CrossRef]
  99. Jansen, K.U.; Rosolowsky, M.; Schultz, L.D.; Markus, H.Z.; Cook, J.C.; Donnelly, J.J.; Martinez, D.; Ellis, R.W.; Shaw, A.R. Vaccination with yeast-expressed cottontail rabbit papillomavirus (CRPV) virus-like particles protects rabbits from CRPV-induced papilloma formation. Vaccine 1995, 13, 1509–1514. [Google Scholar] [CrossRef]
  100. Lowe, R.S.; Brown, D.R.; Bryan, J.T.; Cook, J.C.; George, H.A.; Hofmann, K.J.; Hurni, W.M.; Joyce, J.G.; Lehman, E.D.; Markus, H.Z.; et al. Human papillomavirus type 11 (HPV-11) neutralizing antibodies in the serum and genital mucosal secretions of African green monkeys immunized with HPV-11 virus-like particles expressed in yeast. J. Infect. Dis. 1997, 176, 1141–1145. [Google Scholar] [CrossRef]
  101. Neeper, M.P.; Hofmann, K.J.; Jansen, K.U. Expression of the major capsid protein of human papillomavirus type 11 in Saccharomyces cerevisae. Gene 1996, 180, 1–6. [Google Scholar] [CrossRef]
  102. Cook, J.C.; Joyce, J.G.; George, H.A.; Schultz, L.D.; Hurni, W.M.; Jansen, K.U.; Hepler, R.W.; Ip, C.; Lowe, R.S.; Keller, P.M.; et al. Purification of virus-like particles of recombinant human papillomavirus type 11 major capsid protein L1 from Saccharomyces cerevisiae. Protein Expr. Purif. 1999, 17, 477–484. [Google Scholar] [CrossRef]
  103. Jagadish, M.N.; Ward, C.W.; Gough, K.H.; Tulloch, P.A.; Whittaker, L.A.; Shukla, D.D. Expression of potyvirus coat protein in Escherichia coli and yeast and its assembly into virus-like particles. J. Gen. Virol. 1991, 72, 1543–1550. [Google Scholar] [CrossRef]
  104. Wang, X.W.; Sheng, W.; Zeng, Y. Formation and identification of virus-like particles of poliovirus type I. Chin. J. Exp. Clin. Virol. 2013, 27, 373–375. (In Chinese) [Google Scholar]
  105. Karpenko, L.I.; Lebedev, L.R.; Ignatyev, G.M.; Agafonov, A.P.; Poryvaeva, V.A.; Pronyaeva, T.R.; Ryabchikova, E.I.; Pokrovsky, A.G.; Ilyichev, A.A. Construction of artificial virus-like particles exposing HIV epitopes, and the study of their immunogenic properties. Vaccine 2003, 21, 386–392. [Google Scholar] [CrossRef] [PubMed]
  106. Mukherjee, S.; Thorsteinsson, M.V.; Johnston, L.B.; DePhillips, P.A.; Zlotnick, A. A quantitative description of in vitro assembly of human papillomavirus 16 virus-like particles. J. Mol. Biol. 2008, 381, 229–237. [Google Scholar] [CrossRef] [PubMed]
  107. Mach, H.; Volkin, D.B.; Troutman, R.D.; Wang, B.; Luo, Z.; Jansen, K.U.; Shi, L. Disassembly and reassembly of yeast-derived recombinant human papillomavirus virus-like particles (HPV VLPs). J. Pharm. Sci. 2006, 95, 2195–2206. [Google Scholar] [CrossRef]
  108. Gedvilaite, A.; Zvirbliene, A.; Staniulis, J.; Sasnauskas, K.; Krüger, D.H.; Ulrich, R. Segments of puumala hantavirus nucleocapsid protein inserted into chimeric polyomavirus-derived virus-like particles induce a strong immune response in mice. Viral Immunol. 2004, 17, 51–68. [Google Scholar] [CrossRef]
  109. 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]
  110. Zielonka, A.; Gedvilaite, A.; Ulrich, R.; Lüschow, D.; Sasnauskas, K.; Müller, H.; Johne, R. Generation of virus-like particles consisting of the major capsid protein VP1 of goose hemorrhagic polyomavirus and their application in serological tests. Virus Res. 2006, 120, 128–137. [Google Scholar] [CrossRef]
  111. Lawatscheck, R.; Aleksaite, E.; Schenk, J.A.; Micheel, B.; Jandrig, B.; Holland, G.; Sasnauskas, K.; Gedvilaite, A.; Ulrich, R.G. Chimeric polyomavirus-derived virus-like particles: The immunogenicity of an inserted peptide applied without adjuvant to mice depends on its insertion site and its flanking linker sequence. Viral Immunol. 2007, 20, 453–460. [Google Scholar] [CrossRef] [Green Version]
  112. Pumpens, P.; Razanskas, R.; Pushko, P.; Renhof, R.; Gusars, I.; Skrastina, D.; Ose, V.; Borisova, G.; Sominskaya, I.; Petrovskis, I.; et al. Evaluation of HBs, HBc, and frCP virus-like particles for expression of human papillomavirus 16 E7 oncoprotein epitopes. Intervirology 2002, 45, 24–32. [Google Scholar] [CrossRef]
  113. Choi, Y.R.; Kim, H.J.; Lee, J.Y.; Kang, H.A.; Kim, H.J. Chromatographically purified capsid proteins of red-spotted grouper nervous necrosis virus expressed in Saccharomyces cerevisiae form virus-like particles. Protein Expr. Purif. 2013, 89, 162–168. [Google Scholar] [CrossRef]
  114. Kwag, H.L.; Kim, H.J.; Chang, D.Y.; Kim, H.J. The production and immunogenicity of human papillomavirus type 58 virus-like particles produced in Saccharomyces cerevisiae. J. Microbiol. 2012, 50, 813–820. [Google Scholar] [CrossRef] [PubMed]
  115. Hadiji-Abbes, N.; Mihoubi, W.; Martin, M.; Karakasyan-Dia, C.; Frikha, F.; Gergely, C.; Jouenne, T.; Gargouri, A.; Mokdad-Gargouri, R. Characterization of C69R variant HBsAg: Effect on binding to anti-HBs and the structure of virus-like particles. Arch. Virol. 2015, 160, 2427–2433. [Google Scholar] [CrossRef] [PubMed]
  116. Stolt, A.; Sasnauskas, K.; Koskela, P.; Lehtinen, M.; Dillner, J. Seroepidemiology of the human polyomaviruses. J. Gen. Virol. 2003, 84, 1499–1504. [Google Scholar] [CrossRef] [PubMed]
  117. Lowin, T.; Raab, U.; Schroeder, J.; Franssila, R.; Modrow, S. Parvovirus B19 VP2-proteins produced in Saccharomyces cerevisiae: Comparison with VP2-particles produced by baculovirus-derived vectors. J. Vet. Med. B Infect. Dis. Vet. Public Health 2005, 52, 348–352. [Google Scholar] [CrossRef]
  118. Joyce, J.G.; Tung, J.S.; Przysiecki, C.T.; Cook, J.C.; Lehman, E.D.; Sands, J.A.; Jansen, K.U.; Keller, P.M. The L1 major capsid protein of human papillomavirus type 11 recombinant virus-like particles interacts with heparin and cell-surface glycosaminoglycans on human keratinocytes. J. Biol. Chem. 1999, 274, 5810–5822. [Google Scholar] [CrossRef] [Green Version]
  119. Gedvilaite, A.; Frömmel, C.; Sasnauskas, K.; Micheel, B.; Ozel, M.; Behrsing, O.; Staniulis, J.; Jandrig, B.; Scherneck, S.; Ulrich, R. Formation of immunogenic virus-like particles by inserting epitopes into surface-exposed regions of hamster polyomavirus major capsid protein. Virology 2000, 273, 21–35. [Google Scholar] [CrossRef]
  120. Fu, J.; VanDusen, W.J.; Kolodin, D.G.; O’Keefe, D.O.; Herber, W.K.; George, H.A. Continuous culture study of the expression of hepatitis B surface antigen and its self-assembly into virus-like particles in Saccharomyces cerevisiae. Biotechnol. Bioeng. 1996, 49, 578–586. [Google Scholar] [CrossRef]
  121. Towne, V.; Zhao, Q.; Brown, M.; Finnefrock, A.C. Pairwise antibody footprinting using surface plasmon resonance technology to characterize human papillomavirus type 16 virus-like particles with direct anti-HPV antibody immobilization. J. Immunol. Methods 2013, 388, 1–7. [Google Scholar] [CrossRef]
  122. Ahmad, M.; Hirz, M.; Pichler, H.; Schwab, H. Protein expression in Pichia pastoris: Recent achievements and perspectives for heterologous protein production. Appl. Microbiol. Biotechnol. 2014, 98, 5301–5317. [Google Scholar] [CrossRef] [Green Version]
  123. Barsøe, S.; Toffan, A.; Pascoli, F.; Stratmann, A.; Pretto, T.; Marsella, A.; Er-Rafik, M.; Vendramin, N.; Olesen, N.J.; Sepúlveda, D.; et al. Long-Term Protection and Serologic Response of European Sea Bass Vaccinated with a Betanodavirus Virus-Like Particle Produced in Pichia pastoris. Vaccines 2021, 9, 447. [Google Scholar] [CrossRef]
  124. Shanmugam, R.K.; Ramasamy, V.; Shukla, R.; Arora, U.; Swaminathan, S.; Khanna, N. Pichia pastoris-expressed Zika virus envelope domain III on a virus-like particle platform: Design, production and immunological evaluation. Pathog. Dis. 2019, 77, ftz026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Gupta, J.; Kaul, S.; Srivastava, A.; Kaushik, N.; Ghosh, S.; Sharma, C.; Batra, G.; Banerjee, M.; Shalimar Nayak, B.; Ranjith-Kumar, C.T.; et al. Expression, Purification and Characterization of the Hepatitis E Virus Like-Particles in the Pichia pastoris. Front. Microbiol. 2020, 11, 141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Sherry, L.; Grehan, K.; Snowden, J.S.; Knight, M.L.; Adeyemi, O.O.; Rowlands, D.J.; Stonehouse, N.J. Comparative Molecular Biology Approaches for the Production of Poliovirus Virus-Like Particles Using Pichia pastoris. mSphere 2020, 5, e00838-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Sherry, L.; Grehan, K.; Swanson, J.J.; Bahar, M.W.; Porta, C.; Fry, E.E.; Stuart, D.I.; Rowlands, D.J.; Stonehouse, N.J. Production and Characterisation of Stabilised PV-3 Virus-like Particles Using Pichia pastoris. Viruses 2022, 14, 2159. [Google Scholar] [CrossRef] [PubMed]
  128. Eto, Y.; Saubi, N.; Ferrer, P.; Joseph-Munné, J. Expression of Chimeric HPV-HIV Protein L1P18 in Pichia pastoris; Purification and Characterization of the Virus-like Particles. Pharmaceutics 2021, 13, 1967. [Google Scholar] [CrossRef]
  129. Parker, S.A.; Maloy, M.H.; Tome-Amat, J.; Bardliving, C.L.; Batt, C.A.; Lanz, K.J.; Olesberg, J.T.; Arnold, M.A. Optimization of norovirus virus-like particle production in Pichia pastoris using a real-time near-infrared bioprocess monitor. Biotechnol. Prog. 2016, 32, 518–526. [Google Scholar] [CrossRef]
  130. Pechelyulko, A.; Andreeva-Kovalevskaya, Z.; Dmitriev, D.; Lavrov, V.; Massino, Y.; Nagel, A.; Segal, O.; Sokolova, O.S.; Solonin, A.; Tarakanova, Y.; et al. A simple method to purify recombinant HCV core protein expressed in Pichia pastoris for obtaining virus-like particles and producing monoclonal antibodies. Protein Expr. Purif. 2021, 183, 105864. [Google Scholar] [CrossRef]
  131. Acosta-Rivero, N.; Aguilar, J.C.; Musacchio, A.; Falcón, V.; Viña, A.; de la Rosa, M.C.; Morales, J. Characterization of the HCV core virus-like particles produced in the methylotrophic yeast Pichia pastoris. Biochem. Biophys. Res. Commun. 2001, 287, 122–125. [Google Scholar] [CrossRef]
  132. Yang, Z.; Gao, F.; Wang, X.; Shi, L.; Zhou, Z.; Jiang, Y.; Ma, X.; Zhang, C.; Zhou, C.; Zeng, X.; et al. Development and characterization of an enterovirus 71 (EV71) virus-like particles (VLPs) vaccine produced in Pichia pastoris. Hum. Vaccin. Immunother. 2020, 16, 1602–1610. [Google Scholar] [CrossRef]
  133. Zhang, C.; Ku, Z.; Liu, Q.; Wang, X.; Chen, T.; Ye, X.; Li, D.; Jin, X.; Huang, Z. High-yield production of recombinant virus-like particles of enterovirus 71 in Pichia pastoris and their protective efficacy against oral viral challenge in mice. Vaccine 2015, 33, 2335–2341. [Google Scholar] [CrossRef]
  134. Shukla, R.; Rajpoot, R.K.; Arora, U.; Poddar, A.; Swaminathan, S.; Khanna, N. Pichia pastoris-Expressed Bivalent Virus-Like Particulate Vaccine Induces Domain III-Focused Bivalent Neutralizing Antibodies without Antibody-Dependent Enhancement in Vivo. Front. Microbiol. 2018, 8, 2644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Arora, U.; Tyagi, P.; Swaminathan, S.; Khanna, N. Virus-like particles displaying envelope domain III of dengue virus type 2 induce virus-specific antibody response in mice. Vaccine 2013, 31, 873–878. [Google Scholar] [CrossRef] [PubMed]
  136. Feng, Q.; He, Y.; Lu, J. Virus-Like Particles Produced in Pichia Pastoris Induce Protective Immune Responses against Coxsackievirus A16 in Mice. Med. Sci. Monit. 2016, 22, 3370–3382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Tomé-Amat, J.; Fleischer, L.; Parker, S.A.; Bardliving, C.L.; Batt, C.A. Secreted production of assembled Norovirus virus-like particles from Pichia pastoris. Microb. Cell Fact. 2014, 13, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. 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] [PubMed] [Green Version]
  139. Tripathi, L.; Mani, S.; Raut, R.; Poddar, A.; Tyagi, P.; Arora, U.; de Silva, A.; Swaminathan, S.; Khanna, N. Pichia pastoris-expressed dengue 3 envelope-based virus-like particles elicit predominantly domain III-focused high titer neutralizing antibodies. Front. Microbiol. 2015, 6, 1005. [Google Scholar] [CrossRef] [Green Version]
  140. Tan, B.H.; Fu, J.L.; Sugrue, R.J. Characterization of the dengue virus envelope glycoprotein expressed in Pichia pastoris. Methods Mol. Biol. 2007, 379, 163–176. [Google Scholar]
  141. Mani, S.; Tripathi, L.; Raut, R.; Tyagi, P.; Arora, U.; Barman, T.; Sood, R.; Galav, A.; Wahala, W.; de Silva, A.; et al. Pichia pastoris-expressed dengue 2 envelope forms virus-like particles without pre-membrane protein and induces high titer neutralizing antibodies. PLoS ONE 2013, 8, e64595. [Google Scholar] [CrossRef]
  142. 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] [Green Version]
  143. 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] [Green Version]
  144. Zhao, P.; Jiang, Y.; Wang, J.; Fan, H.; Cao, R. Secreted expression of Japanese encephalitis virus prME in Pichia pastoris and immunogenicity evaluation of the virus-like particles in mice. Chin. J. Biotechnol. 2017, 33, 863–874. (In Chinese) [Google Scholar]
  145. Liu, W.; Jiang, H.; Zhou, J.; Yang, X.; Tang, Y.; Fang, D.; Jiang, L. Recombinant dengue virus-like particles from Pichia pastoris: Efficient production and immunological properties. Virus Genes 2010, 40, 53–59. [Google Scholar] [CrossRef] [PubMed]
  146. Joseph, N.M.; Ho, K.L.; Tey, B.T.; Tan, C.S.; Shafee, N.; Tan, W.S. Production of the virus-like particles of nipah virus matrix protein in Pichia pastoris as diagnostic reagents. Biotechnol. Prog. 2016, 32, 1038–1045. [Google Scholar] [CrossRef] [PubMed]
  147. 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] [PubMed] [Green Version]
  148. Yun, S.M.; Jeong, Y.E.; Wang, E.; Lee, Y.J.; Han, M.G.; Park, C.; Lee, W.J.; Choi, W. Cloning and Expression of Recombinant Tick-Borne Encephalitis Virus-like Particles in Pichia pastoris. Osong Public Health Res. Perspect. 2014, 5, 274–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhou, Y.; Zhang, C.; Liu, Q.; Gong, S.; Geng, L.; Huang, Z. A virus-like particle vaccine protects mice against coxsackievirus A10 lethal infection. Antiviral Res. 2018, 152, 124–130. [Google Scholar] [CrossRef]
  150. Zahid, M.; Rinas, U. Guidelines for Small-Scale Production and Purification of Hepatitis B Surface Antigen Virus-Like Particles from Recombinant Pichia pastoris. Methods Mol. Biol. 2019, 1923, 309–322. [Google Scholar]
  151. Lünsdorf, H.; Gurramkonda, C.; Adnan, A.; Khanna, N.; Rinas, U. Virus-like particle production with yeast: Ultrastructural and immunocytochemical insights into Pichia pastoris producing high levels of the hepatitis B surface antigen. Microb. Cell Fact. 2011, 10, 48. [Google Scholar] [CrossRef] [Green Version]
  152. Hosseini, S.N.; Sarvari, T.; Bashiri, G.; Khatami, M.; Shojaosadati, S.A. Assessing virus like particles formation and r-HBsAg aggregation during large scale production of recombinant hepatitis B surface antigen from Pichia pastoris. Int. J. Biol. Macromol. 2019, 139, 697–711. [Google Scholar] [CrossRef]
  153. Gurramkonda, C.; Zahid, M.; Nemani, S.K.; Adnan, A.; Gudi, S.K.; Khanna, N.; Ebensen, T.; Lünsdorf, H.; Guzmán, C.A.; Rinas, U. Purification of hepatitis B surface antigen virus-like particles from recombinant Pichia pastoris and in vivo analysis of their immunogenic properties. J. Chromatogr. B 2013, 940, 104–111. [Google Scholar] [CrossRef]
  154. Liu, R.; Lin, Q.; Sun, Y.; Lu, X.; Qiu, Y.; Li, Y.; Guo, X. Expression, purification, and characterization of hepatitis B virus surface antigens (HBsAg) in yeast Pichia Pastoris. Appl. Biochem. Biotechnol. 2009, 158, 432–444. [Google Scholar] [CrossRef] [PubMed]
  155. Gurramkonda, C.; Adnan, A.; Gäbel, T.; Lünsdorf, H.; Ross, A.; Nemani, S.K.; Swaminathan, S.; Khanna, N.; Rinas, U. Simple high-cell density fed-batch technique for high-level recombinant protein production with Pichia pastoris: Application to intracellular production of Hepatitis B surface antigen. Microb. Cell Fact. 2009, 8, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Watelet, B.; Quibriac, M.; Rolland, D.; Gervasi, G.; Gauthier, M.; Jolivet, M.; Letourneur, O. Characterization and diagnostic potential of hepatitis B virus nucleocapsid expressed in E. coli and P. pastoris. J. Virol. Methods 2002, 99, 99–114. [Google Scholar] [CrossRef] [PubMed]
  157. Tang, Y.X.; Jiang, L.F.; Zhou, J.M.; Yin, Y.; Yang, X.M.; Liu, W.Q.; Fang, D.Y. Induction of virus-neutralizing antibodies and T cell responses by dengue virus type 1 virus-like particles prepared from Pichia pastoris. Chin. Med. J. 2012, 125, 1986–1992. [Google Scholar] [PubMed]
  158. 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]
  159. Hanumantha, R.N.; Baji, B.P.; Rajendra, L.; Sriraman, R.; Pang, Y.Y.; Schiller, J.T.; Srinivasan, V.A. Expression of codon optimized major capsid protein (L1) of human papillomavirus type 16 and 18 in Pichia pastoris; purification and characterization of the virus-like particles. Vaccine 2011, 29, 7326–7334. [Google Scholar] [CrossRef] [Green Version]
  160. Gupta, G.; Giannino, V.; Rishi, N.; Glueck, R. Immunogenicity of next-generation HPV vaccines in non-human primates: Measles-vectored HPV vaccine versus Pichia pastoris recombinant protein vaccine. Vaccine 2016, 34, 4724–4731. [Google Scholar] [CrossRef]
  161. Dewi, K.S.; Chairunnisa, S.; Swasthikawati, S.; Yuliawati Agustiyanti, D.F.; Mustopa, A.Z.; Kusharyoto, W.; Ningrum, R.A. Production of codon-optimized Human papillomavirus type 52 L1 virus-like particles in Pichia pastoris BG10 expression system. Prep. Biochem. Biotechnol. 2022, 53, 148–156. [Google Scholar] [CrossRef]
  162. Zhu, J.; Yang, K.; Liu, A.; Lu, X.; Yang, L.; Zhao, Q. Highly secretory expression of recombinant cowpea chlorotic mottle virus capsid proteins in Pichia pastoris and in-vitro encapsulation of ruthenium nanoparticles for catalysis. Protein Expr. Purif. 2020, 174, 105679. [Google Scholar] [CrossRef]
  163. Jiang, Z.; Tong, G.; Cai, B.; Xu, Y.; Lou, J. Purification and immunogenicity study of human papillomavirus 58 virus-like particles expressed in Pichia pastoris. Protein Expr. Purif. 2011, 80, 203–210. [Google Scholar] [CrossRef]
  164. 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] [Green Version]
  165. Wang, M.; Pan, Q.; Lu, Z.; Li, K.; Gao, H.; Qi, X.; Gao, Y.; Wang, X. An optimized, highly efficient, self-assembled, subvirus-like particle of infectious bursal disease virus (IBDV). Vaccine 2016, 34, 3508–3514. [Google Scholar] [CrossRef]
  166. Zhang, C.; Liu, Q.; Ku, Z.; Hu, Y.; Ye, X.; Zhang, Y.; Huang, Z. Coxsackievirus A16-like particles produced in Pichia pastoris elicit high-titer neutralizing antibodies and confer protection against lethal viral challenge in mice. Antiviral Res. 2016, 129, 47–51. [Google Scholar] [CrossRef] [PubMed]
  167. Stephen, S.L.; Beales, L.; Peyret, H.; Roe, A.; Stonehouse, N.J.; Rowlands, D.J. Recombinant Expression of Tandem-HBc Virus-Like Particles (VLPs). Methods Mol. Biol. 2018, 1776, 97–123. [Google Scholar] [PubMed]
  168. Freivalds, J.; Dislers, A.; Ose, V.; Pumpens, P.; Tars, K.; Kazaks, A. Highly efficient production of phosphorylated hepatitis B core particles in yeast Pichia pastoris. Protein Expr. Purif. 2011, 75, 218–224. [Google Scholar] [CrossRef] [PubMed]
  169. Sugrue, R.J.; Fu, J.; Howe, J.; Chan, Y.C. Expression of the dengue virus structural proteins in Pichia pastoris leads to the generation of virus-like particles. J. Gen. Virol. 1997, 78, 1861–1866. [Google Scholar] [CrossRef] [PubMed]
  170. Yazdani, R.; Shams-Bakhsh, M.; Hassani-Mehraban, A.; Arab, S.S.; Thelen, N.; Thiry, M.; Crommen, J.; Fillet, M.; Jacobs, N.; Brans, A.; et al. Production and characterization of virus-like particles of grapevine fanleaf virus presenting L2 epitope of human papillomavirus minor capsid protein. BMC Biotechnol. 2019, 19, 81. [Google Scholar] [CrossRef] [Green Version]
  171. Yan, W.W.; Cui, Z.Z.; Wang, Y.K. Expression of capsid gene of Chinese isolate of rabbit hemorrhagic disease virus in Pichia pastoris. Chin. J. Biotechnol. 2005, 21, 135–138. (In Chinese) [Google Scholar]
  172. Kazaks, A.; Lu, I.N.; Farinelle, S.; Ramirez, A.; Crescente, V.; Blaha, B.; Ogonah, O.; Mukhopadhyay, T.; de Obanos, M.P.; Krimer, A.; et al. Production and purification of chimeric HBc virus-like particles carrying influenza virus LAH domain as vaccine candidates. BMC Biotechnol. 2017, 17, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Fazlalipour, M.; Keyvani, H.; Monavari, S.H.; 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] [Green Version]
  174. Zhou, Y.; Shen, C.; Zhang, C.; Zhang, W.; Wang, L.; Lan, K.; Liu, Q.; Huang, Z. Yeast-produced recombinant virus-like particles of coxsackievirus A6 elicited protective antibodies in mice. Antiviral Res. 2016, 132, 165–169. [Google Scholar] [CrossRef] [PubMed]
  175. Sanchooli, A.; Aghaiypour, K.; Kiasari, B.A.; Samarbaf-Zadeh, A.; Ghadiri, A.; Makvandi, M. VLP Production from Recombinant L1/L2 HPV-16 Protein Expressed in Pichia pastoris. Protein Pept. Lett. 2018, 25, 783–790. [Google Scholar] [CrossRef] [PubMed]
  176. Yang, L.; Xiao, A.; Wang, H.; Zhang, X.; Zhang, Y.; Li, Y.; Wei, Y.; Liu, W.; Chen, C. A VLP-Based Vaccine Candidate Protects Mice against Japanese Encephalitis Virus Infection. Vaccines 2022, 10, 197. [Google Scholar] [CrossRef] [PubMed]
  177. Bisht, H.; Chugh, D.A.; Raje, M.; Swaminathan, S.S.; Khanna, N. Recombinant dengue virus type 2 envelope/hepatitis B surface antigen hybrid protein expressed in Pichia pastoris can function as a bivalent immunogen. J. Biotechnol. 2002, 99, 97–110. [Google Scholar] [CrossRef] [PubMed]
  178. 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] [Green Version]
  179. 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] [Green Version]
  180. Coimbra, E.C.; Gomes, F.B.; Campos, J.F.; D’arc, M.; Carvalho, J.C.; Mariz, F.C.; Jesus, A.L.; Stocco, R.C.; Beçak, W.; Freitas, A.C. Production of L1 protein from different types of HPV in Pichia pastoris using an integrative vector. Braz. J. Med. Biol. Res. 2011, 44, 1209–1214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Zhou, Y.; Fan, Y.; LaPatra, S.E.; Ma, J.; Xu, J.; Meng, Y.; Jiang, N.; Zeng, L. Protective immunity of a Pichia pastoris expressed recombinant iridovirus major capsid protein in the Chinese giant salamander, Andrias davidianus. Vaccine 2015, 33, 5662–5669. [Google Scholar] [CrossRef]
  182. Hosseini, S.N.; Javidanbardan, A.; Alizadeh Salim, B.S.; Khatami, M. Large-scale purification of recombinant hepatitis B surface antigen from Pichia pastoris with non-affinity chromatographic methods as a substitute to immunoaffinity chromatography. Prep. Biochem. Biotechnol. 2018, 48, 683–692. [Google Scholar] [CrossRef]
  183. Fernández, E.; Toledo, J.R.; Méndez, L.; González, N.; Parra, F.; Martín-Alonso, J.M.; Limonta, M.; Sánchez, K.; Cabrales, A.; Estrada, M.P.; et al. Conformational and thermal stability improvements for the large-scale production of yeast-derived rabbit hemorrhagic disease virus-like particles as multipurpose vaccine. PLoS ONE 2013, 8, e56417. [Google Scholar] [CrossRef] [Green Version]
  184. Jesus, A.L.; Mariz, F.C.; Souza, H.M.; Cordeiro, M.N.; Coimbra, E.C.; Leitão, M.C.; Nascimento, L.M.; Stocco, R.C.; Beçak, W.; Freitas, A.C. Expression of the bovine papillomavirus type 1, 2 and 4 L1 genes in the yeast Pichia pastoris. Genet. Mol. Res. 2012, 11, 2598–2607. [Google Scholar] [CrossRef] [PubMed]
  185. Bazan, S.B.; de Alencar Muniz Chaves, A.; Aires, K.A.; Cianciarullo, A.M.; Garcea, R.L.; Ho, P.L. Expression and characterization of HPV-16 L1 capsid protein in Pichia pastoris. Arch. Virol. 2009, 154, 1609–1617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Xia, M.; Farkas, T.; Jiang, X. Norovirus capsid protein expressed in yeast forms virus-like particles and stimulates systemic and mucosal immunity in mice following an oral administration of raw yeast extracts. J. Med. Virol. 2007, 79, 74–83. [Google Scholar] [CrossRef] [PubMed]
  187. Mariz, F.C.; Coimbra, E.C.; Jesus, A.L.; Nascimento, L.M.; Torres, F.A.; Freitas, A.C. Development of an IP-Free Biotechnology Platform for Constitutive Production of HPV16 L1 Capsid Protein Using the Pichia pastoris PGK1 Promoter. BioMed Res. Int. 2015, 2015, 594120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  188. Rolland, D.; Gauthier, M.; Dugua, J.M.; Fournier, C.; Delpech, L.; Watelet, B.; Letourneur, O.; Arnaud, M.; Jolivet, M. Purification of recombinant HBc antigen expressed in Escherichia coli and Pichia pastoris: Comparison of size-exclusion chromatography and ultracentrifugation. J. Chromatogr. B Biomed. Sci. Appl. 2001, 753, 51–65. [Google Scholar] [CrossRef]
  189. Fernández, E.; Toledo, J.R.; Mansur, M.; Sánchez, O.; Gil, D.F.; González-González, Y.; Lamazares, E.; Fernández, Y.; Parra, F.; Farnós, O. Secretion and assembly of calicivirus-like particles in high-cell-density yeast fermentations: Strategies based on a recombinant non-specific BPTI-Kunitz-type protease inhibitor. Appl. Microbiol. Biotechnol. 2015, 99, 3875–3886. [Google Scholar] [CrossRef]
  190. Acosta-Rivero, N.; Rodriguez, A.; Musacchio, A.; Falcón, V.; Suarez, V.M.; Martinez, G.; Guerra, I.; Paz-Lago, D.; Morera, Y.; de la Rosa, M.C.; et al. In vitro assembly into virus-like particles is an intrinsic quality of Pichia pastoris derived HCV core protein. Biochem. Biophys. Res. Commun. 2004, 325, 68–74. [Google Scholar] [CrossRef]
  191. Acosta-Rivero, N.; Falcón, V.; Alvarez, C.; Musacchio, A.; Chinea, G.; Cristina de la Rosa, M.; Rodriguez, A.; Dueñas-Carrera, S.; Tsutsumi, V.; Shibayama, M.; et al. Structured HCV nucleocapsids composed of P21 core protein assemble primary in the nucleus of Pichia pastoris yeast. Biochem. Biophys. Res. Commun. 2003, 310, 48–53. [Google Scholar] [CrossRef]
  192. Acosta-Rivero, N.; Alvarez-Obregón, J.C.; Musacchio, A.; Falcón, V.; Dueñas-Carrera, S.; Marante, J.; Menéndez, I.; Morales, J. In vitro self-assembled HCV core virus-like particles induce a strong antibody immune response in sheep. Biochem. Biophys. Res. Commun. 2002, 290, 300–304. [Google Scholar] [CrossRef]
  193. Majeau, N.; Gagné, V.; Bolduc, M.; Leclerc, D. Signal peptide peptidase promotes the formation of hepatitis C virus non-enveloped particles and is captured on the viral membrane during assembly. J. Gen. Virol. 2005, 86, 3055–3064. [Google Scholar] [CrossRef]
  194. 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]
  195. Wetzel, D.; Barbian, A.; Jenzelewski, V.; Schembecker, G.; Merz, J.; Piontek, M. Bioprocess optimization for purification of chimeric VLP displaying BVDV E2 antigens produced in yeast Hansenula polymorpha. J. Biotechnol. 2019, 306, 203–212. [Google Scholar] [CrossRef] [PubMed]
  196. Wetzel, D.; Rolf, T.; Suckow, M.; Kranz, A.; Barbian, A.; Chan, J.A.; Leitsch, J.; Weniger, M.; Jenzelewski, V.; Kouskousis, B.; et al. Establishment of a yeast based VLP platform for antigen presentation. Microb. Cell Fact. 2018, 17, 17. [Google Scholar] [CrossRef] [PubMed]
  197. Su, C.; Li, L.; Jin, Z.; Han, X.; Zhao, P.; Wang, L.; Jiang, C.; Wang, Y.; Wang, W.; Xu, D.; et al. Fermentation, purification and immunogenicity evaluation of hepatitis E virus-like particles expressed in Hansenula polymorpha. Chin. J. Biotechnol. 2017, 33, 653–663. (In Chinese) [Google Scholar]
  198. Chan, J.A.; Wetzel, D.; Reiling, L.; Miura, K.; Drew, D.R.; Gilson, P.R.; Anderson, D.A.; Richards, J.S.; Long, C.A.; Suckow, M.; et al. Malaria vaccine candidates displayed on novel virus-like particles are immunogenic and induce transmission-blocking activity. PLoS ONE 2019, 14, e0221733. [Google Scholar] [CrossRef] [Green Version]
  199. Wetzel, D.; Chan, J.A.; Suckow, M.; Barbian, A.; Weniger, M.; Jenzelewski, V.; Reiling, L.; Richards, J.S.; Anderson, D.A.; Kouskousis, B.; et al. Display of malaria transmission-blocking antigens on chimeric duck hepatitis B virus-derived virus-like particles produced in Hansenula polymorpha. PLoS ONE 2019, 14, e0221394. [Google Scholar] [CrossRef] [Green Version]
  200. McGregor, J.; Hardy, J.M.; Lay, C.S.; Boo, I.; Piontek, M.; Suckow, M.; Coulibaly, F.; Poumbourios, P.; Center, R.J.; Drummer, H.E. Virus-Like Particles Containing the E2 Core Domain of Hepatitis C Virus Generate Broadly Neutralizing Antibodies in Guinea Pigs. J. Virol. 2022, 96, e0167521. [Google Scholar] [CrossRef]
  201. Duan, J.; Yang, D.; Chen, L.; Yu, Y.; Zhou, J.; Lu, H. Efficient production of porcine circovirus virus-like particles using the nonconventional yeast Kluyveromyces marxianus. Appl. Microbiol. Biotechnol. 2019, 103, 833–842. [Google Scholar] [CrossRef]
  202. Simon, C.; Klose, T.; Herbst, S.; Han, B.G.; Sinz, A.; Glaeser, R.M.; Stubbs, M.T.; Lilie, H. Disulfide linkage and structure of highly stable yeast-derived virus-like particles of murine polyomavirus. J. Biol. Chem. 2014, 289, 10411–10418. [Google Scholar] [CrossRef] [Green Version]
  203. Yang, D.; Chen, L.; Duan, J.; Yu, Y.; Zhou, J.; Lu, H. Investigation of Kluyveromyces marxianus as a novel host for large-scale production of porcine parvovirus virus-like particles. Microb. Cell Fact. 2021, 20, 24. [Google Scholar] [CrossRef]
  204. Luu, V.T.; Moon, H.Y.; Hwang, J.Y.; Kang, B.K.; Kang, H.A. Development of recombinant Yarrowia lipolytica producing virus-like particles of a fish nervous necrosis virus. J. Microbiol. 2017, 55, 655–664. [Google Scholar] [CrossRef] [PubMed]
  205. Flores, O.; Alcaíno, J.; Fernandez-Lobato, M.; Cifuentes, V.; Baeza, M. Characterization of virus-like particles and identification of capsid proteins in Xanthophyllomyces dendrorhous. Virus Genes 2015, 50, 253–259. [Google Scholar] [CrossRef] [PubMed]
  206. Kadri, A.; Wege, C.; Jeske, H. In vivo self-assembly of TMV-like particles in yeast and bacteria for nanotechnological applications. J. Virol. Methods 2013, 189, 328–340. [Google Scholar] [CrossRef] [PubMed]
  207. Wildt, S.; Gerngross, T.U. The humanization of N-glycosylation pathways in yeast. Nat. Rev. Microbiol. 2005, 3, 119–128. [Google Scholar] [CrossRef]
  208. Conde, R.; Cueva, R.; Pablo, G.; Polaina, J.; Larriba, G. A search for hyperglycosylation signals in yeast glycoproteins. J. Biol. Chem. 2004, 279, 43789–43798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Dowling, W.; Thompson, E.; Badger, C.; Mellquist, J.L.; Garrison, A.R.; Smith, J.M.; Paragas, J.; Hogan, R.J.; Schmaljohn, C. Influences of glycosylation on antigenicity, immunogenicity, and protective efficacy of Ebola virus GP DNA vaccines. J. Virol. 2007, 81, 1821–1837. [Google Scholar] [CrossRef] [Green Version]
  210. Sakuragi, S.; Goto, T.; Sano, K.; Morikawa, Y. HIV type 1 Gag virus-like particle budding from spheroplasts of Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA 2002, 99, 7956–7961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Cervera, L.; Gutiérrez-Granados, S.; Martínez, M.; Blanco, J.; Gòdia, F.; Segura, M.M. Generation of HIV-1 Gag VLPs by transient transfection of HEK 293 suspension cell cultures using an optimized animal-derived component free medium. J. Biotechnol. 2013, 166, 152–165. [Google Scholar] [CrossRef]
  212. Fuenmayor, J.; Gòdia, F.; Cervera, L. Production of virus-like particles for vaccines. N. Biotechnol. 2017, 39, 174–180. [Google Scholar] [CrossRef]
  213. Chen, Y.; Zhang, Y.; Quan, C.; Luo, J.; Yang, Y.; Yu, M.; Kong, Y.; Ma, G.; Su, Z. Aggregation and antigenicity of virus like particle in salt solution–A case study with hepatitis B surface antigen. Vaccine 2015, 33, 4300–4306. [Google Scholar] [CrossRef]
  214. Lang, R.; Winter, G.; Vogt, L.; Zurcher, A.; Dorigo, B.; Schimmele, B. Rational design of a stable, freeze-dried virus-like particle-based vaccine formulation. Drug Dev. Ind. Pharm. 2009, 35, 83–97. [Google Scholar] [CrossRef]
  215. Jezek, J.; Chen, D.; Watson, L.; Crawford, J.; Perkins, S.; Tyagi, A.; Jones-Braun, L. A heat-stable hepatitis B vaccine formulation. Hum. Vaccines 2009, 5, 529–535. [Google Scholar] [CrossRef] [PubMed]
  216. Kissmann, J.; Ausar, S.F.; Foubert, T.R.; Brock, J.; Switzer, M.H.; Detzi, E.J.; Vedvick, T.S.; Middaugh, C.R. Physical stabilization of Norwalk virus-like particles. J. Pharm. Sci. 2008, 97, 4208–4218. [Google Scholar] [CrossRef] [PubMed]
  217. Tsoka, S.; Holwill, I.; Hoare, M. Virus-like particle analysis in yeast homogenate using a laser light-scattering assay. Biotechnol. Bioeng. 1999, 63, 290–297. [Google Scholar] [CrossRef]
  218. Shi, L.; Sanyal, G.; Ni, A.; Luo, Z.; Doshna, S.; Wang, B.; Graham, T.L.; Wang, N.; Volkin, D.B. Stabilization of human papillomavirus virus-like particles by non-ionic surfactants. J. Pharm. Sci. 2005, 94, 1538–1551. [Google Scholar] [CrossRef]
  219. Ding, Y.; Chuan, Y.P.; He, L.; Middelberg, A.P. Modeling the competition between aggregation and self-assembly during virus-like particle processing. Biotechnol. Bioeng. 2010, 107, 550–560. [Google Scholar] [CrossRef] [PubMed]
  220. Huang, Y.; Bi, J.; Zhou, W.; Li, Y.; Wang, Y.; Ma, G.; Su, Z. Improving recovery of recombinant hepatitis B virus surface antigen by ion-exchange chromatographic supports with low ligand density. Process Biochem. 2006, 41, 2320–2326. [Google Scholar] [CrossRef]
  221. Meingast, C.; Heldt, C.L. Arginine-enveloped virus inactivation and potential mechanisms. Biotechnol. Prog. 2020, 36, e2931. [Google Scholar] [CrossRef]
  222. Kissmann, J.; Joshi, S.B.; Haynes, J.R.; Dokken, L.; Richardson, C.; Middaugh, C.R. H1N1 influenza virus-like particles: Physical degradation pathways and identification of stabilizers. J. Pharm. Sci. 2011, 100, 634–645. [Google Scholar] [CrossRef]
  223. Hashemi, K.; Ghahramani Seno, M.M.; Ahmadian, M.R.; Malaekeh-Nikouei, B.; Bassami, M.R.; Dehghani, H.; Afkhami-Goli, A. Optimizing the synthesis and purification of MS2 virus like particles. Sci. Rep. 2021, 11, 19851. [Google Scholar] [CrossRef]
  224. Galula, J.U.; Chang, G.J.; Chao, D.Y. Production and Purification of Dengue Virus-like Particles from COS-1 Cells. Bio. Protoc. 2019, 9, e3280. [Google Scholar] [CrossRef] [PubMed]
  225. Pease, L.F., 3rd; Lipin, D.I.; Tsai, D.H.; Zachariah, M.R.; Lua, L.H.; Tarlov, M.J.; Middelberg, A.P. Quantitative characterization of virus-like particles by asymmetrical flow field flow fractionation, electrospray differential mobility analysis, and transmission electron microscopy. Biotechnol Bioeng. 2009, 102, 845–855. [Google Scholar] [CrossRef] [PubMed]
  226. Lua, L.H.L.; Connors, N.K.; Sainsbury, F.; Chuan, Y.P.; Wibowo, N.; Middelberg, A.P.J. Bioengineering Virus-like Particles as Vaccines. Biotechnol. Bioeng. 2014, 111, 425–440. [Google Scholar] [CrossRef] [PubMed]
  227. Zhao, Q.; Modis, Y.; High, K.; Towne, V.; Meng, Y.; Wang, Y.; Alexandroff, J.; Brown, M.; Carragher, B.; Potter, C.S.; et al. Disassembly and reassembly of human papillomavirus virus-like particles produces more virion-like antibody reactivity. Virol. J. 2012, 9, 52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Syed, Y.Y. RTS, S/AS01 malaria vaccine (Mosquirix®): A profile of its use. Drugs Ther. Perspect. 2022, 38, 373–381. [Google Scholar] [CrossRef] [PubMed]
  229. Ashley, C.E.; Carnes, E.C.; Phillips, G.K.; Durfee, P.N.; Buley, M.D.; Lino, C.A.; Padilla, D.P.; Phillips, B.; Carter, M.B.; Willman, C.L.; et al. Cell-specific delivery of diverse cargos by bacteriophage MS2 virus-like particles. ACS Nano 2011, 5, 5729–5745. [Google Scholar] [CrossRef] [Green Version]
  230. Yacoby, I.; Shamis, M.; Bar, H.; Shabat, D.; Benhar, I. Targeting antibacterial agents by using drug-carrying filamentous bacteriophages. Antimicrob. Agents Chemother. 2006, 50, 2087–2097. [Google Scholar] [CrossRef] [Green Version]
  231. Crooke, S.N.; Schimer, J.; Raji, I.; Wu, B.; Oyelere, A.K.; Finn, M.G. Lung Tissue Delivery of Virus-Like Particles Mediated by Macrolide Antibiotics. Mol. Pharm. 2019, 16, 2947–2955. [Google Scholar] [CrossRef]
  232. Legendre, D.; Fastrez, J. Production in Saccharomyces cerevisiae of MS2 virus-like particles packaging functional heterologous mRNAs. J. Biotechnol. 2005, 117, 183–194. [Google Scholar] [CrossRef]
  233. Sun, S.; Li, W.; Sun, Y.; Pan, Y.; Li, J. A new RNA vaccine platform based on MS2 virus-like particles produced in Saccharomyces cerevisiae. Biochem. Biophys. Res. Commun. 2011, 407, 124–128. [Google Scholar] [CrossRef]
  234. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Tornesello, A.L.; Tagliamonte, M.; Buonaguro, F.M.; Tornesello, M.L.; Buonaguro, L. Virus-like Particles as Preventive and Therapeutic Cancer Vaccines. Vaccines 2022, 10, 227. [Google Scholar] [CrossRef] [PubMed]
  236. Fei, W.; Shui, G.; Zhang, Y.; Krahmer, N.; Ferguson, C.; Kapterian, T.S.; Lin, R.C.; Dawes, I.W.; Brown, A.J.; Li, P.; et al. A role for phosphatidic acid in the formation of “supersized” lipid droplets. PLoS Genet. 2011, 7, e1002201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic showing the different strategies of anti-viral vaccine development. The figure shows both conventional (attenuated and killed virus) and newer methods of vaccine development (including subunit, nucleic acid, and VLP-based vaccines).
Figure 1. Schematic showing the different strategies of anti-viral vaccine development. The figure shows both conventional (attenuated and killed virus) and newer methods of vaccine development (including subunit, nucleic acid, and VLP-based vaccines).
Vaccines 11 00479 g001
Figure 2. Schematic showing the different applications of VLPs. Several studies have already shown proof of concept for applications like vaccines, drug delivery, and photo imaging. We propose the possible use of VLPs to study virus-host interaction or internalization.
Figure 2. Schematic showing the different applications of VLPs. Several studies have already shown proof of concept for applications like vaccines, drug delivery, and photo imaging. We propose the possible use of VLPs to study virus-host interaction or internalization.
Vaccines 11 00479 g002
Figure 3. Schematic showing different systems used as hosts for expressing and purifying VLPs. Note: Saccharomyces cerevisiae and Komagataella phaffii (in red font) remain the most commonly used yeast species. In the case of in vitro production of VLPs, one can use the protein translational machinery of either bacteria (prokaryotes) or yeast (eukaryotes).
Figure 3. Schematic showing different systems used as hosts for expressing and purifying VLPs. Note: Saccharomyces cerevisiae and Komagataella phaffii (in red font) remain the most commonly used yeast species. In the case of in vitro production of VLPs, one can use the protein translational machinery of either bacteria (prokaryotes) or yeast (eukaryotes).
Vaccines 11 00479 g003
Table 1. A comparison of the different model systems used as a host for the production of VLPs [22,54].
Table 1. A comparison of the different model systems used as a host for the production of VLPs [22,54].
FeatureBacteriaInsect CellsMammalian CellsPlantYeast
Production costLowHighHighModerateLow
Growth mediaSimpleComplexComplexSimpleSimple
GrowthFastSlowSlowVery slowFast
Growth durationVery smallSmallSmallLongSmall
Indoor/OutdoorIndoorIndoorIndoorOutdoor/polyhouseIndoor
Scale-upEasyVery difficultVery difficultDifficultEasy
SecretionNoYesYesNAYes
Enveloped/non-envelopedNon-envelopedEnveloped/Non-envelopedEnveloped/Non-envelopedNANon-enveloped/Enveloped possible
Speed of transformant screeningVery fastSlowSlowVery slowFast
Effect of seasonal variationsNoNoNoYesNo
NA: clear information not available.
Table 2. Studies in which S. cerevisiae yeast was used to generate VLPs.
Table 2. Studies in which S. cerevisiae yeast was used to generate VLPs.
S. NoProtein AntigenVirusProtein
Localization
PromoterReferences
1VP2Human parvovirus 4ICHybrid GAL10-PYK1promoter[62]
2Capsid proteinHepatitis E virusICGAL promoter[63]
3Capsid proteinPorcine circovirus type 2ECGPD, TEF2[64]
4Nucleocapsid proteinSendai virusICGAL7[65]
5VP2, VP1Human bocavirusesIC [66]
6Surface antigenHepatitis B virusICGAL[67,68,69]
7p55(gag)HIV-1EC [70]
8VP1Human polyomavirusesICGAL[71]
9L1HPV 16ICGAL10[72,73]
10Nucleocapsid proteinTioman virusICGAL10[74]
11L-HDAg and surface antigenHepatitis delta virusICGAD[75]
12Capsid proteinPorcine circovirus type 2ICGAL10[76]
13Capsid proteinEnterovirus 71ICGAL10[77]
14VP1Human and non-human polyomavirusesICGAL[78]
15Capsid proteinAdeno-associated virusICGAL1[79]
16Nucleocapsid proteinHuman parainfluenza virus 4ICGAL7[80]
17 *Coat proteinCacteriophage Qbeta virusICGAL[81]
18Capsid proteinNervous necrosis virusICGAL[82]
19VP1,2Parvovirus B19ICADH2/GAPDH[83,84]
20VP1Bird polyomavirusesICGAL[85]
21P1Enterovirus 71 and Coxsackievirus A16ICGAL1[86]
22Capsid protein Porcine circovirus type 2ICGAL1[87]
23VP2,6,7RotavirusICPGK1, TEF1[88,89]
24VP2Human parvovirus 4ICGAL1-10[90]
25Nucleocapsid proteinHuman parainfluenza virus 2ICGAL[91]
26Nucleocapsid proteinMenangle virusICGAL7[92]
27GagHIV-1ICGAP[93]
28VP2Porcine parvovirusICGAL1-10 [94]
29P1, CD3Coxsackievirus A16ICGAL1[95]
30Capsid proteinPorcine circovirus type 2ICGAL[96]
31VP1,2Hepatitis B/PolyomavirusICGAL7[97]
32VP1Hamster polyomavirusIC [98]
33L1/L1 + L2Cottontail rabbit papillomavirusICGAL1-10 [99]
34L1HPV 11ICGAL[100,101,102]
35Coat proteinPotyvirus (Johnsongrass mosaic virus)ICADC1[103]
36P1, CD3Poliovirus type IIC [104]
37 HIV-1IC [105]
38L1HPV 16ICGAL[106]
39L1HPV 6,11 16ICGAL[107]
40VP1 with Puumala hantavirus nucleocapsid protein segmentsHamster polyomavirus ICHybrid GAL10-PYK1[108]
41M proteinHepatitis B virusICGAL10/CYC1[109]
42VP1,2Goose hemorrhagic polyomavirusICGAL[110]
43CEA/VP1Hamster polyomavirusICGAL[111]
44E7 oncoprotein of HPV16Hepatitis B virusIC [112]
45Capsid proteinRed-spotted grouper nervous necrosis virusICGAL10[113]
46L1HPV 58ICGAL10[114]
47C69R variant of surface antigenHepatitis B virusICGAL10/CYC1[115]
48VP1Human polyomavirus 2ICGAL[116]
49VP2Parvovirus B19ICGAL1[117]
50L1HPV 11ICGAL110-11[118]
51VP1 with pre-S1 region of the Hepatitis B virusHamster polyomavirusICGAL[119]
52Surface antigenHepatitis B virusICGAL10[120]
53L1HPV 16IC [121]
* Additionally, expressed in K. phaffii; IC: intracellular; EC: extracellular.
Table 4. Studies where H. polymorpha yeast was used to generate VLPs.
Table 4. Studies where H. polymorpha yeast was used to generate VLPs.
S. NoProtein AntigenDiseaseSecretion
Localization
PromoterReferences
1Porcine circovirus type 2b capsid proteinPostweaning multisystemic wasting disease (PMWS) in pigsICMOX (methanol)[194]
2Bovine viral diarrhea virus (BVDV) glycoprotein E2Bovine diarrheaICFMD (formate dehydrogenase) [195]
3Membrane integral small surface protein (dS) of the duck Hepatitis B virusBovine diarrheaICMOX (methanol)[196]
4Surface antigenHepatitis E IC [197]
5Sporozoite antigenMalaria IC [198]
6Sporozoite antigenMalariaICMOX (methanol)[199]
7Glycoprotein E2 ectodomain Hepatitis CEC [200]
IC: intracellular; EC: extracellular.
Table 5. Studies where yeast species (other than those in Table 2, Table 3 and Table 4) were used to generate VLPs.
Table 5. Studies where yeast species (other than those in Table 2, Table 3 and Table 4) were used to generate VLPs.
S. NoProtein AntigenDiseaseSecretion
Localization
Yeast SpeciesReferences
1Capsid protein of porcine circovirusPostweaning multisystemic wasting syndrome (PMWS) and porcine circovirus diseases (PCVDs)ICKluyveromyces marxianus[201]
2VP1 of murine polyomavirusCancerICKluyveromyces lactis[202]
3VP2 of porcine parvovirusEmbryonic and fetal loss, death, and mummificationICKluyveromyces marxianus[203]
4Capsid protein of red-spotted grouper nervous necrosis virusViral encephalopathy and
retinopathy
ICYarrowia lipolytica[204]
5Totivirus capsid proteinNAICXanthophyllomyces dendrorhous[205]
6Tobacco mosaic virus coat proteinMosaic-like mottling and discoloration on the leavesICSchizosaccharomyces pombe[206]
IC: intracellular; EC: extracellular. NA: not applicable as no disease is reported where Totivirus is responsible.
Table 6. VLP-based vaccines expressed in yeast that are either approved by the FDA for commercial use or are undergoing clinical trials.
Table 6. VLP-based vaccines expressed in yeast that are either approved by the FDA for commercial use or are undergoing clinical trials.
Trade NameInfectious
Agent
DiseaseCompanyAntigenStatus
Gardasil®HPVCervical carcinomaMerckL1Approved
Gardasil9®HPVCervical carcinomaMerckL1Approved
Mosquirix™P. falciparumMalariaUniv. of Rochester AVEG, Rochester, New York, United StatesP. falciparum circumsporozoite protein fused to the Hepatitis B surface antigenApproved
NAHIVAcquired immune deficiency syndrome HIV p17/p24: Ty-VLPClinical trial Phase 1(Clinical trial No.: NCT00001053) [43]
Hepavax-GeneHBVHepatocellular carcinomaCrucell (Dusseldorf-Germany)SHBs, MHBsLicensed *
FendrixHBVHepatocellular carcinomaGSK (Belgium)SHBsLicensed
Heplisav-BHBVHepatocellular carcinomaDynavax (Oakland-USA)SHBsLicensed *
EngerixHBVHepatocellular carcinomaGSK (UK)SHBsLicensed
Recombivax HB (H-B-Vac®-II)HBVHepatocellular carcinomaMerck Vaccine (Canada)SHBsLicensed
NA: Not Available; HPV: Human Papillomavirus [22]. * H. polymorpha yeast was used as the expression host, while in the rest of the cases, S. cerevisiae was used. HBV: Hepatitis B virus.
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

Srivastava, V.; Nand, K.N.; Ahmad, A.; Kumar, R. Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery. Vaccines 2023, 11, 479. https://doi.org/10.3390/vaccines11020479

AMA Style

Srivastava V, Nand KN, Ahmad A, Kumar R. Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery. Vaccines. 2023; 11(2):479. https://doi.org/10.3390/vaccines11020479

Chicago/Turabian Style

Srivastava, Vartika, Kripa N. Nand, Aijaz Ahmad, and Ravinder Kumar. 2023. "Yeast-Based Virus-like Particles as an Emerging Platform for Vaccine Development and Delivery" Vaccines 11, no. 2: 479. https://doi.org/10.3390/vaccines11020479

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