We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
ReviewOpen Accesscc iconby icon

Virus-like particles derived from bacteriophage MS2 as antigen scaffolds and RNA protective shells

    Antonina Naskalska

    *Author for correspondence:

    E-mail Address: antonina.naskalska@uj.edu.pl

    Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, 30-392, Poland

    &
    Jonathan Gardiner Heddle

    **Author for correspondence:

    E-mail Address: jonathan.heddle@uj.edu.pl

    Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, 30-392, Poland

    Department of Biosciences, Durham University, Durham, DH1 3LE, UK

    Published Online:https://doi.org/10.2217/nnm-2023-0362

    Abstract

    The versatile potential of bacteriophage MS2-derived virus-like particles (VLPs) in medical biotechnology has been extensively studied during the last 30 years. Since the first reports showing that MS2 VLPs can be produced at high yield and relatively easily engineered, numerous applications have been proposed. Particular effort has been spent in developing MS2 VLPs as protective capsules and delivery platforms for diverse molecules, such as chemical compounds, proteins and nucleic acids. Among these, two are particularly noteworthy: as scaffolds displaying heterologous epitopes for vaccine development and as capsids for encapsulation of foreign RNA. In this review, we summarize the progress in developing MS2 VLPs for these two areas.

    Plain language summary

    Hollow, nanosized protein particles have many potential uses. If they can be appropriately engineered, they may for example be able to carry therapeutic cargoes to diseased cells or be used as a vaccine where appropriate antigens are mounted on their external surface. Many viruses offer a ready-made protein particle, the capsid, which can be made hollow by exclusion of the viral genetic material. MS2 is a virus that targets bacteria – a bacteriophage – which is well characterized and has been developed over many years for a number of applications. It has particular promise for development as a vaccine and for RNA delivery, both of which are reviewed here.

    Graphical abstract

    Bacteriophage MS2 has features and properties that make it attractive for a range of biotechnological and biomedical applications. Figure created with Biorender.com.

    Viruses effectively deliver nucleic acid materials to cells. For this reason, replacing native genetic material with therapeutic nucleic acids or other cargoes has long been attractive and considerable research has been carried out in the area [1–3]. Bacteriophages (viruses that target bacteria) are of particular interest, being nonpathogenic for humans. Attractive features of such virus-based systems include the fact that heterologous DNAs and RNAs can be relatively easily incorporated into the bacteriophage genome and that peptide display on virus-like particles (VLPs) derived from bacteriophages is highly feasible meaning that these vehicles can be ligand-targeted to a specific cell type. In addition, available bacteriophages cover a wide range of sizes and shapes which may give different biodistribution properties, and they are typically easy to handle with good stability and easy to produce at large scales.

    MS2 is an icosahedral ssRNA bacteriophage belonging to the Leviviridae family. Its capsid, measuring ∼26 nm in diameter, consists of 178 copies of coat protein (CP) and one copy of the maturation protein (A protein or maturase). These are encoded in the ∼3.6 kb RNA genome which also encodes a replicase, and lysis protein. During virus assembly the CP dimerizes and binds to a specific viral RNA hairpin (called the pac site). However, when the CP is expressed from a plasmid in a bacterial or yeast host, in absence of other viral elements, it self-assembles into empty capsids called VLPs. Analogous to the native MS2 virion, formation of the MS2 VLP involves dimerization of the CP and formation of an unstructured loop connecting the F and G β-strands of each subunit (known as the FG loop) [4]. Depending on the conformation of the FG loop, the CP monomer can exist in three conformations: A, B and C. Likewise, the resulting dimers adopt two forms: asymmetric (called A/B) or symmetric (called C/C) as depicted in Figure 1. Of note, MS2 VLPs are composed of 90 CP dimers (60 A/B dimers and 30 C/C dimers), thus compensating the lack of maturase protein present in the native virion.

    Figure 1. Structure of MS2 bacteriophage.

    (A) Crystal structure of MS2 (PDB 2MS2) [5]. CPs A, B and C are colored lime green, dark pink and coral blue, respectively. (B) Schematic representation of the structure of MS2 showing that CP forms dimers adopting two conformations: called A/B and C/C as indicated, adapted from Biela et al. published under a CC BY 4.0 license ( http://creativecommons.org/licenses/by/4.0/) [6]. (C & D) Close-up orthogonal views of the structures of two dimer types represented as ribbons and colored as in panel (A).

    CP: Coat protein; PDB: Protein Data Bank.

    Since the initial description of MS2 VLPs [7] they have been extensively characterized and much is now understood of the mechanisms governing the assembly process, the flexibility of the particle in terms of supporting structural modifications and possibility to exploit its internal cavity. Purification protocols have also been developed and refined. Many of these studies were pioneered over several decades by the groups of Peabody [4,8–12] and Stockley [13–19]. They have enabled further development of MS2 VLPs for the applications described below. Here we identify and review the development of MS2 VLPs as scaffolds displaying antigenic peptides for vaccine development and as capsids for encapsulation of foreign RNA.

    MS2 VLP as scaffolds displaying antigenic peptides

    VLPs are known to be effective immunogens, a property that is exploited in a number of successful VLP-based vaccines. The most established examples include the human papillomavirus (HPV) vaccine and the hepatitis B vaccine [20]. The immunogenic efficacy of VLPs appears to be related to their morphological properties such as size, shape and the high-density display of multiple copies of antigens. This results in efficient uptake by antigen-presenting cells and subsequent stimulation of B lymphocytes for antibody production. Additionally, unlike other exogenous antigens, VLPs are cross-presented in association with both MHC class I and class II despite the absence of infection or intracellular replication. Consequently, they are highly effective at stimulating CD4+ T-helper and CD8+ cytotoxic T-lymphocyte responses. Finally, thanks to pathogen-associated molecular patterns originating from producer cells, VLPs can activate the innate immune system too. Because of this, VLPs may be considered as self-boosting antigen carriers and therefore may serve as ideal scaffolds displaying poorly immunogenic antigens [21]. Such chimeric VLPs are also an attractive option for multivalent vaccines, which display several different epitopes on one particle, raising the potential for ‘universal’ vaccines against multiple viral strains [22,23].

    First attempts to incorporate immunogenic epitopes into MS2 VLPs were reported by Mastico and colleagues [15] who reasoned that the most distal loop present at the particle surface could accommodate foreign peptides. This loop corresponds to the N-terminal β-hairpin of the CP and is also referred to as the AB loop [24]. Using a genetic fusion approach, Mastico et al. were able to insert several viral and nonviral peptides of 8–24 amino acids length. Some variants of the produced chimeric MS2 VLPs were soluble and could be further purified and used to immunize mice. Effective production of high titer antibodies specific to the peptides displayed on the MS2 VLP was obtained, thereby providing a proof-of-concept for further development of such a system. This strategy was subsequently used to produce MS2 VLPs displaying malaria epitopes [25]. Specifically, an epitope from the immunodominant liver-stage antigen-1 of the malaria parasite Plasmodium falciparum was incorporated into MS2 VLPs. The ability to induce humoral and cellular-specific immune responses by this vaccine candidate was demonstrated in mice models. However, based on subsequent findings related to Plasmodium evasion of human immune response, other malaria antigens were targeted [26]. Eventually, after many years of efforts, an effective malaria vaccine, based on a VLP (derived from hepatitis B virus) displaying another antigen (RTS,S) has been developed and licensed (Mosquirix™, GSK, London, UK).

    Structural studies of MS2 VLP have shown that genetic fusion of the two identical CP monomers results in the expression of properly folded CP single-chain dimers [10]. Moreover, it was found that this modification protects the protein against the destabilizing effects of amino acid substitutions and chemical denaturants, and thus stabilizes the resulting VLPs [12]. It is worth noting, however, that only one insertion of a foreign peptide into the single-chain dimer is tolerated if proper VLP assembly is to be achieved. Thus, the display density on the VLP is consequently reduced by half (from 180 copies to 90). Using this approach Peabody and colleagues managed to incorporate 6, 8 and 10 amino acid peptides originating from an HIV antigen into MS2 VLPs [27].

    While MS2 has been investigated, the general field of VLP-based vaccine development has experienced great success with the approval of VLP vaccines protecting from infection of HPVs (Gardasil®, Merck & Co. [NJ, USA]), Cervarix® (GSK) [28]. These VLPs are made of HPV major capsid protein L1 and are protective against several of the most common serotypes of the virus. However, the L1 protein is not conserved among all existing serotypes and thus L1-based vaccines do not confer enough cross-protection. On the other hand, the minor HPV capsid protein – L2 – is highly conserved among HPV types and therefore of interest in developing a broadly protective HPV vaccine. Unfortunately, the L2 protein, unlike L1, cannot form VLPs and as such must be displayed on carriers to ensure sufficient immunogenicity. Production of an MS2 VLP displaying HPV L2 antigens was first reported in 2012 by Tumban et al. [29] and further improved by the same group [30–32]. Remarkably, Tumban and coauthors chose a different location for epitope insertion: the gene coding for L2 peptide was fused to the N-terminus of the CP single-chain dimer. Interestingly, they observed that this location offered better cross-protection against diverse HPV serotypes [33]. They also tested insertions ranging from 12 to 27 amino acids, and created bivalent MS2 VLPs displaying 17–36 and/or 69–86 HPV L2 peptides, further increasing cross-reactivity [30,34]. Finally, they developed a formulation to enhance vaccine stability and clinical applicability and refined its optimal dose [35]. The efficacy of all produced MS2 HPV vaccines was validated in challenge studies in mice.

    In 2015, the production of MS2 VLP-carrying epitopes for foot-and-mouth disease virus (FMDV) was reported [36]. Here, a 19 amino acid peptide (epitope peptide 141–160) was inserted into the AB loop of the single-chain dimer of CP. Proper assembly of the chimeric VLPs was demonstrated, but also challenge tests on guinea pigs and swine were carried out, showing that the vaccine could partially protect animals against FMDV. This vaccine candidate was improved 3 years later by elongating the immunodominant epitope with a motif shown to be essential in inducing FMDV neutralizing antibodies (epitope peptide 131–160) [37]. The authors of these studies not only demonstrated that their preparation could efficiently enhance antibody levels and cellular immune response compared with a commercialized synthetic peptide vaccine but also provided evidence that MS2 VLP can be used in larger mammals.

    When Zika virus (ZIKV) emerged, a chimeric MS2 VLP displaying viral peptides was constructed. Six different ZIKV envelope protein fragments (up to 21 amino acids), theoretically predicted to be B-cell linear epitopes, were inserted into the N-terminus of the single-chain dimer of MS2 CP [38]. Mice immunized with a mixture of VLPs displaying ZIKV epitopes elicited anti-ZIKV antibodies. Although immunized mice were not protected against a high-challenge dose of ZIKV, their sera neutralized a low dose of ZIKV in vitro. The authors hypothesized that this could be because the epitopes displayed in MS2 VLP were linear and not conformational (as in the native virus). Similar observations were reported by Mogus and coauthors who displayed the linear epitope from HIV fusion peptide 8 on MS2 VLP via N-terminal fusion to the CP single-chain dimer. Mice immunized with this vaccine candidate exhibited high titers of fusion peptide 8-specific antibodies, albeit not correlating with neutralization or protection [39]. These findings perhaps indicate that for complex antigens requiring additional modifications (i.e., quaternary structure) genetic fusion to CP and subsequent expression in bacteria is not the optimal strategy. This can, however, be overcome by post-assembly attachment of such complex antigens. One example is the work showing successful tethering of biotinylated SARS-CoV-2 spike protein antigen to MS2 VLPs coated with streptavidin [40].

    The most recent MS2 VLP-based vaccine candidate is the one displaying a Chlamydia trichomatis conserved epitope. The 9–10 amino acid peptides were inserted into the AB loop and immunization resulted in protection against a vaginal Chlamydia challenge in mice [41].

    All published work cited in the section above (summarized in Table 1) readily shows that MS2 VLPs can act as universal antigen scaffolds with promising immunological potential. Also, taking into consideration the number of studies in vivo, they seem to exhibit a good safety profile – a feature considered to be essential in the field of vaccine development.

    Table 1. MS2 as scaffolds for foreign peptide display.
    TargetInsertion positionPeptide length (amino acid)Immunogenicity testingRef.
    Influenza virus, Plasmodium falciparum, human papillomavirus, HIVAB loop8–24Mice[15]
    Plasmodium falciparumAB loop24Mice[25]
    HIVAB loop6, 8, 10Mice[27]
    Human papillomavirusN-terminal12–27Mice[29]
    Foot-and-mouth diseaseAB loop19, 29Guinea pigs, swine[36]
    Zika virusN-terminal21Mice[38]
    HIVN-terminal8Mice[39]
    Chlamydia trichomatisAB loop9–10Mice[41]

    In the research area combining vaccine development and MS2 VLPs, there is one more interesting application: biopanning – a method to evaluate antigens in terms of their reactivity with antibodies. In general, this assay takes advantage of the fact the MS2 VLPs encapsidate the nucleic acid that encodes the CP and its guest peptide (as explained in the next section). This means that MS2 VLPs displaying a peptide library can be affinity-selected by interaction with the specific antibody (i.e., from the patient's serum) and its genetic sequences can be recovered and amplified by reverse transcription and PCR. As a consequence, this method allows researchers to pan an antigen in order to identify targets of antibody response or the opposite: to map the antibody repertoire against a defined antigen. Using this approach the specific neutralization-sensitive epitope RH5 [42] and the conformational epitope within blood-stage protein AMA1, both from P. falciparum, were identified [43]. Similarly, dengue virus epitopes recognized by antibodies elicited during acute infection were identified [44]. Importantly, the MS2 VLP can integrate the epitope identification and immunization functions into a single platform as products of affinity selection can be directly used as vaccines.

    MS2 VLP as capsids for encapsulation of foreign RNA

    As already mentioned, in MS2 bacteriophage the packaging of RNA molecules is driven by the dimerization of CPs followed by a specific interaction with a stem-loop structure called the pac site. This stem loop is located at the 5′ of the replicase gene and its binding to CP triggers encapsidation during the assembly process of the MS2 virion, but also acts as a repressor for bacteriophage replication [7]. Interestingly, when the pac site sequence (19 nucleotides [nt]) is fused to a foreign sequence of interest, interaction with the MS2 CP expressed within the same cell is retained and thus packaging of the respective RNA molecule into MS2 VLP can be achieved [8]. Of note, the maturase also plays a role in the process: in the MS2 bacteriophage it interacts specifically with viral RNA [45] and it is assumed that it may facilitate the packaging of foreign RNA into MS2 VLPs. Since the first report describing this packaging strategy in MS2 VLPs, its efficiency has been improved by several modifications of the expression system, as discussed below.

    Armored RNA

    After first demonstration that foreign RNA can be encapsidated inside MS2 VLP [8], the technology was named armored RNA (AR) and has been further developed and patented by du Bois and Pasloske [46]. These authors constructed a one-plasmid-driven packaging system. In this single-plasmid approach, the target RNA is fused to one copy of the pac site sequence and expressed from the same transcript as the MS2 CP and the maturase (Figure 2). It was claimed that such a design results in the optimal ratio of recombinant RNA to MS2 CP and thus avoids the nonspecific packaging of bacterial RNA, which was the major limitation of the initial system described by Pickett and Peabody. DuBois and Pasloske assumed that when CP is being translated from the same RNA that is to be packaged, its expression is regulated because once the concentration of CP is sufficiently high, it encapsidates the RNA from which it is being translated and thus prevents any further CP from being translated from that RNA [47]. Using this approach Pasloske produced AR containing the 172-base consensus sequence from the HIV type 1 gag gene and a consensus 412-base sequence from the 5′-non-coding region/core region of hepatitis C virus subtype 2b [46].

    Figure 2. Comparison of expression systems used to encapsulate foreign RNA in MS2 virus-like particles.

    In the single-plasmid system (top) the maturase, coat protein and RNA sequence to be packaged are encoded on one transcript. Alternatively, maturase and coat protein are encoded on one transcript and the RNA sequence on another, both in the same plasmid. In the two-plasmid system (bottom) the maturase, coat protein and RNA sequence to be packaged are encoded on two separate transcripts, from two different plasmids. Figure created with Biorender.com.

    VLP: Virus-like particle.

    The AR is not only protected from ubiquitous nucleases, but also the concentration of the packaged RNA is controllable, which means that such particles can be used as standards (internal controls [ICs]) for detection or quantification of the corresponding native viruses. Two major advantages of using IC AR are that, contrary to naked RNA, they can be stored for several months at -20°C without losing integrity; and that the AR is processed identically as the tested sample during all steps of the quantification assay: it has to be extracted from the viral particle, reverse-transcribed and then PCR-amplified. As a consequence, using AR as IC allows for more accurate quantification of viral loads in tested samples and thus improved reliability of diagnostic assays. After being patented, the IC AR prepared in MS2 VLPs technology was further commercialized by Ambion Inc. (later Asuragen, TX, USA). In the next years, several different groups described the use of similarly produced enterovirus, HCV, hantavirus and a Crimean–Congo hemorrhagic fever virus AR in real time-PCR diagnosis of clinical specimens [48–51]. Remarkably, a smart improvement of this technology has been introduced by Cheng and colleagues who incorporated a histidine-tag (His-tag) into single-chain dimer CP thus simplifying the purification of particles using His-tag affinity [52].

    It is worth noting that in all examples cited above, the length of RNA packaged in MS2 VLPs is less than 500 bases. Although most RT-PCR assays do not target RNA sequences longer than 500 bases, there are some advantages if longer target RNA sequences are packaged. For instance, in PCRs in which several primer pairs are used for the detection of different regions in the target gene. In addition, if long RNA sequences could be packaged, a single chimeric AR could serve as a standard for a variety of different assays designed for a one-step detection of multiple viruses. To meet this need Wei and coauthors proposed two modifications in the AR technology design that significantly improved the MS2 particle loading capacity [53]. First, they returned to the original idea of using a two-plasmid coexpression system in which the CP and the maturase were expressed from one plasmid while the target RNA containing the pac site was produced by a second plasmid (Figure 2). In order to ensure the optimal ratio of CP and the RNA to be produced, these two plasmids should have equivalent copy numbers. Additionally, plasmids should encode resistance to two different antibiotics, to be stably maintained together in the same bacterial host [53]. Second, they used a variant of the pac site in which uridine at position 5 was replaced by cytosine (called C5 variant), as it has been previously shown that this C5 variant exhibits 6–50-fold increased affinity to the CP [54]. Thanks to these two modifications Wei and colleagues managed to produce the AR comprising a 2248 nt foreign RNA sequence, which includes three SARS-CoV-1 fragments, one HCV fragment and two H5N1 fragments [53]. They also validated the obtained AR as IC for qualitative or quantitative detection by RT-PCR of the respective viruses. Next, the same group demonstrated that adding a single repetition of the pac site sequence (both wild type and C5 variant) in the one-plasmid expression system resulted in increased RNA packaging (from 1.2 kb to 1.8 kb) [55]. Finally, these authors improved the packaging efficiency even further by using a one-plasmid double expression plasmid, where CP and maturase are expressed from the first cloning site of pACYC Duet vector and target RNA with three copies of pac site sequence – from the second cloning site (Figure 2) [56]. In this and subsequently published work, long RNA fragments (up to 3 kb) were packaged in MS2 VLPs (Table 2) [56–59]. Interestingly, thanks to the mentioned modifications, Wei and coauthors exceeded the theoretical packaging limit of approximately 2.0 kb (taking into consideration that MS2 bacteriophage RNA genome is 3.6 kb and the sequence encoding the CP, the maturase and the pac site is 1.7 kb). They hypothesized that this particular setting (one-plasmid double expression and presence of doubled or tripled pac site) might benefit from an optimal ratio of CP/RNA and a cooperative binding of RNA to CP.

    Table 2. MS2 virus-like particles as capsids for armored RNA.
    TargetExpression systemSequence lengthRef.
    HIVOne-plasmid, 1 wt pac site172 nt[46]
    EnteroUp to 500 nt[50]
    HCV[51]
    HCV[60]
    Entero[61]
    HCV[62]
    Lassa virus[63]
    SARS-CoV, H5N1, HCVTwo-plasmid
    1× C5 variant pac site
    2248 nt[53]
    SARS-CoV, HCVOne-plasmid
    2× C5 variant pac site
    1891 nt[55]
    HIVOne-plasmid, double expression
    3× C5 variant pac site
    3034[56]
    Measles virus385[57]
    MERS1002 (1629 + 394)[58]
    SARS-Cov-22908[59]

    HCV: Hepatitis C virus; MERS: Middle East Respiratory Syndrome; nt: Nucleotide.

    Therapeutic RNA

    Therapeutic RNA can be classified into ncRNA, sRNA and coding mRNA. The former is further differentiated into siRNA, which is double stranded and inhibits expression of one specific mRNA, or miRNA (miR), which is single stranded and acts by binding to the 3′-untranslated region of a target mRNA, causing its degradation or translational suppression. By contrast, therapeutic mRNA is meant to be translated and expressed to produce a protein of interest. In all cases, the therapeutic RNA needs to be delivered to the living cells. MS2 VLPs, being natural protective shells for the phage RNA genome, seem to be the ideal tool for this purpose. Indeed, both sRNA and mRNA have been successfully encapsulated in MS2 VLP. While siRNA and miR can be relatively easily packaged, using the same strategy as for generation of the AR, efficient loading of functional mRNA remains more challenging.

    The first report describing attempts to encapsulate miR in MS2 VLPs used the two-plasmid method and targeted a 385 nt sequence of the 5′-untranslated region of the HCV genome [64]. Even though such preparations were successfully delivered to Huh-7 cells, it is not clear how efficient the encapsulation itself was (the published work shows a limited effect on translation inhibition). In the subsequent years, MS2-encapsulated miR-146 [65,66] and miR-122 [67,68] have been reported, targeting, respectively, systemic lupus erythromatosis autoantibodies, osteoclast precursors and hepatocellular carcinoma. In these studies, a one-plasmid system with double pac site (C variant) was used for encapsulation. Importantly, the obtained constructs were not only successfully delivered to cells (in vitro and in vivo) by means of cell-penetrating peptides tethered to the external surface of MS2 VLP, but also demonstrated substantial translation inhibition. Similarly to AR, MS2 VLPs harboring miR can be affinity-purified using His-tag displayed on their surface, as shown by Mikel and coauthors, who adopted this idea for one-plasmid double-expression strategy [69].

    A different approach was undertaken by Ashley and colleagues [70] who encapsulated siRNA (targeting CCNA2 gene): they chemically disassembled MS2 VLPs by treating them with acetic acid and then reassembled them by restoring the neutral pH, in presence of siRNA of interest. The yield of this method was 85 molecules of siRNA per one MS2 VLP (as claimed by the authors). They also demonstrated the functional efficacy of cargo-loaded and cell-penetrating peptide-decorated particles delivered to cells in culture.

    In the case of mRNA to be expressed in mammalian cells, two important sequence modifications are essential: capping the 5′-termini and polyadenylation of the 3′-termini. As neither of them can be conferred by bacteria, a eukaryotic expression system must be used for simultaneous mRNA production and encapsulation into MS2 VLP. For this, a one-plasmid double expression can be used in Saccharomyces cerevisiae. The feasibility of this strategy was first demonstrated by Legendre and Fastrez, who incorporated mRNA coding for human growth hormone (896 nt) into MS2 VLPs [71]. However, these authors confirmed the functionality of their construct only indirectly: by showing that mRNAs extracted from such MS2 preparations can be efficiently translated in cell culture. Eventually, another group managed to package mRNA coding for a prostate cancer antigen (1544 nt) into MS2 VLPs and showed that the resulting antigen is properly expressed by macrophages (following phagocytosis) [72]. Moreover, they demonstrated that the expressed antigen induces the expected immune response in a mice model, thereby providing a proof-of-concept for an MS2 VLP-based therapeutic vaccine.

    Recently, an interesting example of chimeric MS2-alpharetrovirus Gag VLPs for encapsulation of the RNA components of the CRISPR-Cas system has been reported [73]. Namely, the authors of this work managed to package not only mRNA coding for the Cas enzyme but also sgRNA in chimeric particles. Furthermore, with such preparations, they were able to edit targeted genes in human primary cells. Impressively, they even showed that co-packaging of multiple sgRNAs into their chimeric VLPs allowed for the knockout of up to three genes.

    Examples of therapeutic RNA packaged into MS2 VLPs are gathered in Table 3.

    Table 3. MS2 virus-like particles as capsids for therapeutic RNA.
    RNA typeTargetExpression systemCell delivery strategyRef.
    siRNACyclinDisassembly/reassembly (no pac site)SP94 peptide
    HCC
    [70]
    miRNAHCV IRESTwo-plasmid
    pac site C-variant
    TAT peptide
    Huh-7 cells
    [64]
    miR-146a (SLE)One-plasmid
    pac site C-variant
    TAT peptide
    HeLa, the human HCC cell line HepG2 and Huh-7 cells
    [65]
    [74]
    [66]
    miR-122 (SLE, osteoporosis)One-plasmid
    pac site C-variant
    TAT peptide
    HCC Hep3B, HepG2 and Huh7 cells and Hep3B
    [67]
    mRNAHuman growth hormone (896 bp)One-plasmid (yeast)
    1× wt pac site
    None[71]
    HIV-1 Gag mRNAs (1544 bp)One-plasmid (yeast)
    pac site C variant
    Intramuscular vaccination (mice)[72]
    Prostate acid phosphatase (1062 bp)One-plasmid,
    pac site C variant
    Phagocytosis (murine macrophages) Intravenous vaccination (mice)[75]
    sgRNA (CRISPR)SpCas9 mRNA and sgRNA targeting TP53/Trp53 genesAdopted to GagMS2 particlesFibroblasts, hepatocytes, and cord-blood-derived stem and progenitor cells[73]

    HCC: Hepatocellular carcinoma; IRES: Internal ribosome entry site; SLE: Systemic lupus erythematosus.

    In summary, packaging strategies of RNA into MS2 VLPs can be differentiated into ‘in vitro’ (when already formed MS2 VLPs are disassembled and then reassembled in the presence of RNA of interest) and ‘in vivo’ (when RNA packaging occurs in the producing cell [bacterial or yeast] simultaneously with VLP assembly). Packaging of foreign RNA is feasible using several expression settings, as depicted in Figure 3. The encapsulated RNA can be further classified depending on its type (sncRNA or mRNA) and resulting application: as therapeutics (and potentially vaccine) or controls in diagnostics (Figure 3).

    Figure 3. Reported developments in the field of RNA packaging into MS2 virus-like particles.

    Figure created with Biorender.com.

    VLP: Virus-like particle.

    Conclusion

    During the last three decades, MS2 VLPs have emerged as a versatile tool for multiple applications. Due to their relative flexibility in terms of both capsid and internal cargo modifications, these particles can potentially act as universal vaccines combining mRNA packaging, antigen display and decoration with targeting molecules. This ease of functionalization, together with the good safety profile and cost–effectiveness, also makes them attractive candidates for the burgeoning field of RNA therapeutics. It is worth noting that the two areas discussed in this review (scaffolds for foreign peptides and protective shells for heterologous RNA) are not exhaustive: MS2 VLPs are also used as vehicles for chemical agents [70,76], proteins and gold nanoparticles [77], and oligodeoxynucleotides [78]. Another important field of MS2 application, which takes advantage of its controllable affinity to RNA, is in-cell visualization of endogenous RNA [79]. With further development, MS2 VLPs have the potential to play a crucial role in the advancement of several biotechnological and clinical applications.

    Future perspective

    MS2 is well-characterized and has already found practical application as a diagnostic in the form of AR. The most exciting areas for future development are in vaccines and RNA-delivery systems.

    For future vaccine development, an obstacle is that approaches to date have involved the insertion of relatively short antigenic peptides into an external loop. A superior immune response may be generated if epitopes consisting of larger, folded protein domains could be displayed. This may be addressed by post-assembly attachment of such complex antigens (using adapter systems such as SpyTag/SpyCatcher, sortase, biotin, etc.,) – examples of which are mentioned in the text. Another important consideration for use of recombinant VLPs as vaccines is the risk of contamination with bacterial toxins (lipopolysaccharide and others). This could be potentially avoided by adopting bacteria-free production systems. However, it is also worth mentioning that VLPs from phages are in general considered as safer than adenovirus-derived vehicles for instance, as in Lam and Steinmetz [80].

    For progress in RNA delivery, particularly mRNA, the challenge is to find a method to efficiently disassemble and reassemble the CPs to achieve a high yield of cargo loading. This also will require significant reengineering of the protein. One step toward this goal has been achieved with the demonstration of MS2 VLPs engineered to form larger particles with greater capacity [6]. Another approach is production in a eukaryotic host in order to bypass problems associated with assembly/disassembly. This could allow in-cell encapsulation of mRNA containing all necessary elements and most likely will be more efficient in terms of packaging.

    To effectively reengineer the CP to endow it with required new properties, computational and artificial intelligence-based approaches will be invaluable. These could include, for example, Rosetta Protein Design [81], ProteinMPNN [82], AlphaFold [83] and RFdiffusion [84]. These tools, some of them very recent developments, will provide exciting new possibilities for developing a new generation of MS2-based VLPs.

    Executive summary

    MS2 virus-like particles as scaffolds displaying antigenic peptides

    • A number of virus-like particles (VLPs) are known to be effective immunogens and in some cases have been developed into clinically approved vaccines.

    • MS2 is an icosahedral ssRNA bacteriophage around 26 nm in diameter.

    • Immunogenic epitopes can be genetically engineered into the externally displayed AB loop of the capsid proteins.

    • MS2 VLPs displaying such epitopes have been shown to be capable of eliciting an immune response in mice.

    • Genetic fusion of identical coat protein monomers to form fused dimers stabilizes the resulting VLP but only one A/B loop of the resulting dimer can tolerate an insertion if stability is to be maintained.

    • Proof-of-principle studies have been carried out for MS2 modified to act as vaccines against HPV, foot-and-mouth virus and Chlamydia trichomatis.

    • The ability of MS2 VLPs to encapsidate their own encoding nucleic acid means that CPs with fused peptide can be used to ‘biopan’ a peptide library for binding to targets (e.g., antibodies) wherein the sequence encoding binding peptides can be easily recovered.

    MS2 VLPs as capsids for encapsulation of foreign RNA

    • MS2 encapsidates its own RNA by binding to the ‘pac site’, a stem-loop structure.

    • MS2 VLPs can encapsidate foreign RNA sequences by fusion to a pac site sequence.

    • Fusion of target RNA to a copy of the pac site sequence with expression from the same transcript as the MS2 CP and the maturase avoids nonspecific RNA packing. The RNA is protected and stabilized and is known as armored RNA.

    • Armored RNA can be used as internal control standards in real-time-PCR.

    • Armored RNAs able to protect longer RNA sequences were developed through modification of expression vector strategy and pac site.

    • Both in vivo and in vitro RNA packaging approaches have been developed.

    • Foreign sRNAs and mRNA can be encapsidated within MS2 VLPs.

    • MS2 VLPs decorated with cell-penetrating peptides can deliver miRNAs to cells.

    • mRNA, being longer, is more challenging to package but this has been demonstrated.

    Financial disclosure

    A Naskalska acknowledges the support of grant no. UMO-2020/37/B/NZ6/03878 awarded by the National Science Centre (NCN) Poland. JG Heddle was funded by a Leverhulme International Professorship awarded by the Leverhulme Trust. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    The authors declare the following competing financial interest: JG Heddle is the founder of and holds equity in nCage Therapeutics LLC, which aims to commercialize protein cages for therapeutic applications. The authors have no other competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Creative Commons Attribution 4.0 License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Foglizzo V, Marchio S. Bacteriophages as therapeutic and diagnostic vehicles in cancer. Pharmaceuticals (Basel) 14(2), 161 (2021).
    • 2. Karimi M, Mirshekari H, Moosavi Basri SM, Bahrami S, Moghoofei M, Hamblin MR. Bacteriophages and phage-inspired nanocarriers for targeted delivery of therapeutic cargos. Adv. Drug Deliv. Rev. 106(Pt A), 45–62 (2016).
    • 3. van Kan-Davelaar HE, van Hest JC, Cornelissen JJ, Koay MS. Using viruses as nanomedicines. Br. J. Pharmacol. 171(17), 4001–4009 (2014).
    • 4. Peabody DS, Lim F. Complementation of RNA binding site mutations in MS2 coat protein heterodimers. Nucleic Acids Res. 24(12), 2352–2359 (1996).
    • 5. Golmohammadi R, Valegår K, Fridborg K, LiljasL. The refined structure of bacteriophage MS2 at 2· 8 Å resolution. J. Mol. Biol. 234(3), 620–639 (1993).
    • 6. Biela AP, Naskalska A, Fatehi F, Twarock R, Heddle JG. Programmable polymorphism of a virus-like particle. Commun. Mater. 3, 7 (2022).
    • 7. Peabody DS. Translational repression by bacteriophage MS2 coat protein expressed from a plasmid. A system for genetic analysis of a protein–RNA interaction. J. Biol. Chem. 265(10), 5684–5689 (1990).
    • 8. Pickett GG, Peabody DS. Encapsidation of heterologous RNAs by bacteriophage MS2 coat protein. Nucleic Acids Res. 21(19), 4621–4626 (1993). • Breakthrough demonstration that foreign RNA can be encapsidated in MS2, leading to the development of armored RNA.
    • 9. Peabody DS. The RNA binding site of bacteriophage MS2 coat protein. EMBO J. 12(2), 595–600 (1993).
    • 10. Ni CZ, Syed R, Kodandapani R, Wickersham J, Peabody DS, Ely KR. Crystal structure of the MS2 coat protein dimer: implications for RNA binding and virus assembly. Structure 3(3), 255–263 (1995).
    • 11. Peabody DS. Role of the coat protein–RNA interaction in the life cycle of bacteriophage MS2. Mol. Gen. Genet. 254(4), 358–364 (1997).
    • 12. Peabody DS. Subunit fusion confers tolerance to peptide insertions in a virus coat protein. Arch. Biochem. Biophys. 347(1), 85–92 (1997). • Important finding having implications for further peptide insertion incorporation into the coat protein.
    • 13. Talbot SJ, Goodman S, Bates SR, Fishwick CW, Stockley PG. Use of synthetic oligoribonucleotides to probe RNA–protein interactions in the MS2 translational operator complex. Nucleic Acids Res. 18(12), 3521–3528 (1990).
    • 14. Goodman ST, Talbot SJ, Haneef I, Tewary HK, Fishwick CW, Stockley PG. Synthetic RNA in the study of RNA–protein interactions of MS2 bacteriophage. Nucleic Acids Symp. Ser. (24), 293 (1991).
    • 15. Mastico RA, Talbot SJ, Stockley PG. Multiple presentation of foreign peptides on the surface of an RNA-free spherical bacteriophage capsid. J. Gen. Virol. 74(Pt 4), 541–548 (1993). • Foreign peptides incorporated into MS2 virus-like particle (VLP) for the first time.
    • 16. Valegård K, Murray JB, Stockley PG, Stonehouse NJ, Liljas L. Crystal structure of an RNA bacteriophage coat protein–operator complex. Nature 371(6498), 623–626 (1994).
    • 17. Stockley PG, Stonehouse NJ, Murray JB et al. Probing sequence-specific RNA recognition by the bacteriophage MS2 coat protein. Nucleic Acids Res. 23(13), 2512–2518 (1995).
    • 18. Stonehouse NJ, Scott DJ, Fonseca S et al. Molecular interactions in the RNA bacteriophage MS2. Biochem. Soc. Trans. 24(3), S412 (1996).
    • 19. Stonehouse NJ, Valegård K, Golmohammadi R et al. Crystal structures of MS2 capsids with mutations in the subunit FG loop. J. Mol. Biol. 256(2), 330–339 (1996).
    • 20. Mohsen MO, Zha L, Cabral-Miranda G, Bachmann MF. Major findings and recent advances in virus-like particle (VLP)-based vaccines. Semin. Immunol. 34, 123–132 (2017).
    • 21. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat. Rev. Immunol. 10(11), 787–796 (2010).
    • 22. Cohen AA, van Doremalen N, Greaney AJ et al. Mosaic RBD nanoparticles protect against challenge by diverse sarbecoviruses in animal models. Science 377(6606), eabq0839 (2022).
    • 23. Zhang X, Wu S, Liu J et al. A mosaic nanoparticle vaccine elicits potent mucosal immune response with significant cross-protection activity against multiple SARS-CoV-2 sublineages. Adv. Sci. (Weinh.) 10(27), e2301034 (2023).
    • 24. Valegård K, Liljas L, Fridborg K, Unge T. The three-dimensional structure of the bacterial virus MS2. Nature 345(6270), 36–41 (1990).
    • 25. Heal KG, Hill HR, Stockley PG, Hollingdale MR, Taylor-Robinson AW. Expression and immunogenicity of a liver stage malaria epitope presented as a foreign peptide on the surface of RNA-free MS2 bacteriophage capsids. Vaccine 18(3–4), 251–258 (1999).
    • 26. El-Moamly AA, El-Sweify MA. Malaria vaccines: the 60-year journey of hope and final success – lessons learned and future prospects. Trop. Med. Health 51(1), 29 (2023).
    • 27. Peabody DS, Manifold-Wheeler B, Medford A, Jordan SK, do Carmo Caldeira J, Chackerian B. Immunogenic display of diverse peptides on virus-like particles of RNA phage MS2. J. Mol. Biol. 380(1), 252–263 (2008).
    • 28. CDC. www.cdc.gov/std/HPV/STDFact-HPV.htm
    • 29. Tumban E, Peabody J, Tyler M, Peabody DS, Chackerian B. VLPs displaying a single L2 epitope induce broadly cross-neutralizing antibodies against human papillomavirus. PLOS ONE 7(11), e49751 (2012).
    • 30. Zhai L, Peabody J, Pang YS, Schiller J, Chackerian B, Tumban E. A novel candidate HPV vaccine: MS2 phage VLP displaying a tandem HPV L2 peptide offers similar protection in mice to Gardasil-9. Antiviral Res. 147, 116–123 (2017).
    • 31. Tyler M, Tumban E, Dziduszko A, Ozbun MA, Peabody DS, Chackerian B. Immunization with a consensus epitope from human papillomavirus L2 induces antibodies that are broadly neutralizing. Vaccine 32(34), 4267–4274 (2014).
    • 32. Tumban E, Peabody J, Peabody DS, Chackerian B. A universal virus-like particle-based vaccine for human papillomavirus: longevity of protection and role of endogenous and exogenous adjuvants. Vaccine 31(41), 4647–4654 (2013).
    • 33. Yadav R, Zhai L, Tumban E. Virus-like particle-based L2 vaccines against HPVs: where are we today? Viruses 12(1), 18 (2019).
    • 34. Zhai L, Yadav R, Kunda NK et al. Oral immunization with bacteriophage MS2-L2 VLPs protects against oral and genital infection with multiple HPV types associated with head and neck cancers and cervical cancer. Antiviral Res. 166, 56–65 (2019).
    • 35. Hunter Z, Tumban E, Dziduszko A, Chackerian B. Aerosol delivery of virus-like particles to the genital tract induces local and systemic antibody responses. Vaccine 29(28), 4584–4592 (2011).
    • 36. Dong YM, Zhang GG, Huang XJ, Chen L, Chen HT. Promising MS2 mediated virus-like particle vaccine against foot-and-mouth disease. Antiviral Res. 117, 39–43 (2015).
    • 37. Wang G, Liu Y, Feng H et al. Immunogenicity evaluation of MS2 phage-mediated chimeric nanoparticle displaying an immunodominant B cell epitope of foot-and-mouth disease virus. PeerJ 6, e4823 (2018).
    • 38. Basu R, Zhai L, Contreras A, Tumban E. Immunization with phage virus-like particles displaying Zika virus potential B-cell epitopes neutralizes Zika virus infection of monkey kidney cells. Vaccine 36(10), 1256–1264 (2018).
    • 39. Mogus AT, Liu L, Jia M et al. Virus-like particle based vaccines elicit neutralizing antibodies against the HIV-1 fusion peptide. Vaccines (Basel) 8(4), 765 (2020).
    • 40. Chiba S, Frey SJ, Halfmann PJ et al. Multivalent nanoparticle-based vaccines protect hamsters against SARS-CoV-2 after a single immunization. Commun. Biol. 4(1), 597 (2021).
    • 41. Collar AL, Linville AC, Core SB, Frietze KM. Epitope-based vaccines against the Chlamydia trachomatis major outer membrane protein variable domain 4 elicit protection in mice. Vaccines (Basel) 10(6), 875 (2022).
    • 42. Ord RL, Caldeira JC, Rodriguez M et al. A malaria vaccine candidate based on an epitope of the Plasmodium falciparum RH5 protein. Malar. J. 13, 326 (2014).
    • 43. Crossey E, Frietze K, Narum DL, Peabody DS, Chackerian B. Identification of an immunogenic mimic of a conserved epitope on the Plasmodium falciparum blood stage antigen AMA1 using virus-like particle (VLP) peptide display. PLOS ONE 10(7), e0132560 (2015).
    • 44. Frietze KM, Pascale JM, Moreno B, Chackerian B, Peabody DS. Pathogen-specific deep sequence-coupled biopanning: a method for surveying human antibody responses. PLOS ONE 12(2), e0171511 (2017). • Validation of the proof-of-concept that MS2 phage can be used for biopanning.
    • 45. Shiba T, Suzuki Y. Localization of A protein in the RNA–A protein complex of RNA phage MS2. Biochim. Biophys. Acta 654(2), 249–255 (1981).
    • 46. Pasloske BL, Walkerpeach CR, Obermoeller RD, Winkler M, DuBois DB. Armored RNA technology for production of ribonuclease-resistant viral RNA controls and standards. J. Clin. Microbiol. 36(12), 3590–3594 (1998). •• First demonstration that MS2 VLP can have find application in biotechnology. Representing a milestone in viral diagnostics, improving the accuracy of virus quantification.
    • 47. DuBois DB, Winkler MM, Pasloske BL. Ribonuclease Resistant Viral RNA Standards. Cenetron Diagnostics LLC Applied Biosystems LLC Asuragen Inc. US5677124A (1997).
    • 48. Rolfe KJ, Parmar S, Mururi D et al. An internally controlled, one-step, real-time RT-PCR assay for norovirus detection and genogrouping. J. Clin. Virol. 39(4), 318–321 (2007).
    • 49. Ninove L, Nougairede A, Gazin C et al. RNA and DNA bacteriophages as molecular diagnosis controls in clinical virology: a comprehensive study of more than 45,000 routine PCR tests. PLOS ONE 6(2), e16142 (2011).
    • 50. Beld M, Minnaar R, Weel J et al. Highly sensitive assay for detection of enterovirus in clinical specimens by reverse transcription-PCR with an armored RNA internal control. J. Clin. Microbiol. 42(7), 3059–3064 (2004).
    • 51. Meng S, Li J. A novel duplex real-time reverse transcriptase-polymerase chain reaction assay for the detection of hepatitis C viral RNA with armored RNA as internal control. Virol. J. 7, 117 (2010).
    • 52. Cheng Y, Niu J, Zhang Y, Huang J, Li Q. Preparation of His-tagged armored RNA phage particles as a control for real-time reverse transcription-PCR detection of severe acute respiratory syndrome coronavirus. J. Clin. Microbiol. 44(10), 3557–3561 (2006).
    • 53. Wei Y, Yang C, Wei B et al. RNase-resistant virus-like particles containing long chimeric RNA sequences produced by two-plasmid coexpression system. J. Clin. Microbiol. 46(5), 1734–1740 (2008).
    • 54. Lowary PT, Uhlenbeck OC. An RNA mutation that increases the affinity of an RNA–protein interaction. Nucleic Acids Res. 15(24), 10483–10493 (1987).
    • 55. Wei B, Wei Y, Zhang K et al. Construction of armored RNA containing long-size chimeric RNA by increasing the number and affinity of the pac site in exogenous RNA and sequence coding coat protein of the MS2 bacteriophage. Intervirology 51(2), 144–150 (2008).
    • 56. Zhan S, Li J, Xu R, Wang L, Zhang K, Zhang R. Armored long RNA controls or standards for branched DNA assay for detection of human immunodeficiency virus type 1. J. Clin. Microbiol. 47(8), 2571–2576 (2009).
    • 57. Zhang D, Sun Y, Jia T et al. External quality assessment for the detection of measles virus by reverse transcription-PCR using armored RNA. PLOS ONE 10(8), e0134681 (2015).
    • 58. Zhang L, Hao M, Zhang K et al. External quality assessment for the molecular detection of MERS-CoV in China. J. Clin. Virol. 75, 5–9 (2016).
    • 59. Wang Z, Chen Y, Yang J et al. External quality assessment for molecular detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in clinical laboratories. J. Mol. Diagn. 23(1), 19–28 (2021).
    • 60. Felder E, Wölfel R. Development of a versatile and stable internal control system for RT-qPCR assays. J. Virol. Methods 208, 33–40 (2014).
    • 61. Mikel P, Vasickova P, Tesarik R et al. Preparation of MS2 phage-like particles and their use as potential process control viruses for detection and quantification of enteric RNA viruses in different matrices. Front. Microbiol. 7, 1911 (2016).
    • 62. Zambenedetti MR, Pavoni DP, Dallabona AC et al. Internal control for real-time polymerase chain reaction based on MS2 bacteriophage for RNA viruses diagnostics. Mem. Inst. Oswaldo Cruz 112(5), 339–347 (2017).
    • 63. Dedkov VG, Magassouba N, Safonova MV et al. Development and evaluation of a one-step quantitative RT-PCR assay for detection of Lassa virus. J. Virol. Methods 271, 113674 (2019).
    • 64. Wei B, Wei Y, Zhang K et al. Development of an antisense RNA delivery system using conjugates of the MS2 bacteriophage capsids and HIV-1 TAT cell-penetrating peptide. Biomed. Pharmacother. 63(4), 313–318 (2009).
    • 65. Pan Y, Zhang Y, Jia T, Zhang K, Li J, Wang L. Development of a microRNA delivery system based on bacteriophage MS2 virus-like particles. FEBS J. 279(7), 1198–1208 (2012).
    • 66. Yao Y, Jia T, Pan Y et al. Using a novel microRNA delivery system to inhibit osteoclastogenesis. Int. J. Mol. Sci. 16(4), 8337–8350 (2015).
    • 67. Wang G, Jia T, Xu X et al. Novel miR-122 delivery system based on MS2 virus like particle surface displaying cell-penetrating peptide TAT for hepatocellular carcinoma. Oncotarget 7(37), 59402–59416 (2016).
    • 68. Zhang J, Li D, Zhang R, Peng R, Li J. Delivery of microRNA-21-sponge and pre-microRNA-122 by MS2 virus-like particles to therapeutically target hepatocellular carcinoma cells. Exp. Biol. Med. (Maywood) 246(23), 2463–2472 (2021).
    • 69. Mikel P, Vasickova P, Kralik P. One-plasmid double-expression His-tag system for rapid production and easy purification of MS2 phage-like particles. Sci. Rep. 7(1), 17501 (2017).
    • 70. Ashley CE, Carnes EC, Phillips GK et al. Cell-specific delivery of diverse cargos by bacteriophage MS2 virus-like particles. ACS Nano 5(7), 5729–5745 (2011). • Comprehensive study demonstrating that MS2 VLP is a versatile delivery vehicle.
    • 71. Legendre D, Fastrez J. Production in Saccharomyces cerevisiae of MS2 virus-like particles packaging functional heterologous mRNAs. J. Biotechnol. 117(2), 183–194 (2005). • First attempt to package eukaryotic mRNA in MS2 VLPs – the first demonstration that MS2 VLPs can be produced in yeast.
    • 72. 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. 407(1), 124–128 (2011).
    • 73. Baron Y, Sens J, Lange L et al. Improved alpharetrovirus-based Gag.MS2 particles for efficient and transient delivery of CRISPR-Cas9 into target cells. Mol. Ther. Nucleic Acids 27, 810–823 (2022).
    • 74. Pan Y, Jia T, Zhang Y et al. MS2 VLP-based delivery of microRNA-146a inhibits autoantibody production in lupus-prone mice. Int. J. Nanomed. 7, 5957–5967 (2012).
    • 75. Li J, Sun Y, Jia T, Zhang R, Zhang K, Wang L. Messenger RNA vaccine based on recombinant MS2 virus-like particles against prostate cancer. Int. J. Cancer 134(7), 1683–1694 (2014).
    • 76. Aanei IL, Francis MB. Dual surface modification of genome-free MS2 capsids for delivery applications. Methods Mol. Biol. 1776, 629–642 (2018).
    • 77. Aanei IL, Glasgow JE, Capehart SL, Francis MB. Encapsulation of negatively charged cargo in MS2 viral capsids. Methods Mol. Biol. 1776, 303–317 (2018).
    • 78. Wu M, Sherwin T, Brown WL, Stockley PG. Delivery of antisense oligonucleotides to leukemia cells by RNA bacteriophage capsids. Nanomedicine 1(1), 67–76 (2005).
    • 79. Dictenberg J. Genetic encoding of fluorescent RNA ensures a bright future for visualizing nucleic acid dynamics. Trends Biotechnol. 30(120), 621–626 (2012).
    • 80. Lam P, Steinmetz NF. Plant viral and bacteriophage delivery of nucleic acid therapeutics. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 10(1), e1487 (2018).
    • 81. Leman JK, Weitzner BD, Lewis SM et al. Macromolecular modeling and design in Rosetta: recent methods and frameworks. Nat. Methods 2020;17:665–680.
    • 82. Dauparas J, Anishchenko I, Bennett N et al. Robust deep learning-based protein sequence design using Protein MPNN. Science 378(6615), 49–56 (2022).
    • 83. Jumper J, Evans R, Pritzel A et al. Highly accurate protein structure prediction with AlphaFold. Nature 596(7873), 583–589 (2021).
    • 84. Watson JL, Juergens D, Bennett NR et al. De novo design of protein structure and function with RF diffusion. Nature 620(7976), 1089–1100 (2023).