Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2015 Oct 10;99(24):10415–10432. doi: 10.1007/s00253-015-7000-8

The application of virus-like particles as vaccines and biological vehicles

Dan Yan 1, Yan-Quan Wei 1, Hui-Chen Guo 1, Shi-Qi Sun 1,
PMCID: PMC7080154  PMID: 26454868

Abstract

Virus-like particles (VLPs) can be spontaneously self-assembled by viral structural proteins under appropriate conditions in vitro while excluding the genetic material and potential replication probability. In addition, VLPs possess several features including can be rapidly produced in large quantities through existing expression systems, highly resembling native viruses in terms of conformation and appearance, and displaying repeated cluster of epitopes. Their capsids can be modified via genetic insertion or chemical conjugation which facilitating the multivalent display of a homologous or heterogeneous epitope antigen. Therefore, VLPs are considered as a safe and effective candidate of prophylactic and therapeutic vaccines. VLPs, with a diameter of approximately 20 to 150 nm, also have the characteristics of nanometer materials, such as large surface area, surface-accessible amino acids with reactive moieties (e.g., lysine and glutamic acid residues), inerratic spatial structure, and good biocompatibility. Therefore, assembled VLPs have great potential as a delivery system for specifically carrying a variety of materials. This review summarized recent researches on VLP development as vaccines and biological vehicles, which demonstrated the advantages and potential of VLPs in disease control and prevention and diagnosis. Then, the prospect of VLP biology application in the future is discussed as well.

Keywords: Virus-like particles, VLPs, Vaccine, Drug delivery, Diagnostic technology

Introduction

Virus-like particles (VLPs) are composed of one or more structural proteins/capsid proteins of viruses by self-assembling into a particular spatial conformation. In terms of appearance, VLPs are very similar to a live virus without genetic components (Chroboczek et al. 2014; Kushnir et al. 2012). The high density of the epitopes on its surface can be recognized and presented to the immune system by antigen-presenting cells (APC), thus stimulating humoral and cellular immunity effectively through similar pathways as the original pathogens do (Keller et al. 2010). At the same time, none of the viral genetic materials will participate in the formation process of VLPs, which means that there is no risk of viral replication or proliferation. Therefore, VLPs are considered as one of the safest candidate vaccines.

In the 1960s, some empty viral particles without nucleic acid were identified as the capsid protein of hepatic B virus (Blumberg et al. 1965). This finding was considered as the first recorded instance of the natural existence of VLPs. Subsequently, hepatic B virus VLPs were detected that they can induce the host immune responses to eliminate the invasion of the authentic hepatic virus (Bayer et al. 1968). The phenomenon is a clue to understand the relationship between the VLPs and the host immune system. With the progress in genetic engineering technology, the expression and purification of the major capsid protein of human papillomavirus (HPV) was achieved easily in vitro through experiments (Hagensee et al. 1993; Kirnbauer et al. 1992; Li et al. 1997). Thus, models for understanding the assembly of VLPs in vitro were obtained (Brady and Consigli 1978; Li et al. 1997). In the 1980s, the antigenicity and immunogenicity mechanism of HBsAg VLPs were interpreted. Hence, VLP-based vaccines gained further attention from researchers (Edman et al. 1981; McAleer et al. 1984; Valenzuela et al. 1982).

The application of VLPs in vaccines is just one of its major biological applications. As a nanoscale material, VLPs have similar characteristics as nanomaterials and have the ability to mediate more biomedical functions through biotechnological methods. Therefore, VLPs, a new biological tool, have a significant function in drug delivery, genetic therapy, cellular targeting, and cancer treatment. This article will discuss the research development of VLPs in its biomedical application.

VLP-based vaccines

The use of vaccines is one of the most effective strategies in the prevention against pathogenic infection. Since the smallpox vaccination performed by Edward Jenner in the eighteenth century, which was the birth of the concept of vaccines, various inactivated or attenuated vaccines for both human and animals have emerged. In fighting against infectious diseases, these vaccines have made significant contributions, particularly in preventing and eliminating poliovirus, measles, mumps, rubella, influenza, and hepatitis A (Amanna and Slifka 2009; Lua et al. 2014; Plotkin 2005). However, several deficiencies still exist in the use of inactivated and attenuated vaccines. For example, there is a security issue that attenuated vaccines may enhance pathogenicity by reverting to a wild-type phenotype in vaccined individuals, although effective immune responses can be induced by such vaccines (Burns et al. 2014; Esteves 1988; Fachinger et al. 2008; Horaud 1993; Sanders et al. 2015; Wang et al. 2014). Inactivated vaccines cannot replicate in vivo after inoculation; however, acquiring full protection after a single immunization by such vaccines will be difficult (Bright et al. 2007). In addition, there is an urgent practical need to find a candidate vaccine that could be produced in an efficient, scalable, and inexpensive way and with a high degree of safety for some viruses that are difficult to culture in vitro (Chackerian 2007; Moon et al. 2014).

However, VLPs, mimicking the organization and conformation of authentic native viruses but lacking the viral genome, have been used as immunogenic molecules in several recombinant vaccines in the last few years and even used as a therapeutic vaccine to induce the production of specific antibodies against endogenous molecules with a preponderant role in chronic diseases (Andrade et al. 2013; Roldao et al. 2010; Speiser et al. 2010; Spohn et al. 2008). Some VLP vaccines have been licensed and commercialized, and a large number of VLP-based vaccine candidates have also been undergoing clinical evaluation (see Table 1); therefore, VLPs have provided delivery systems that combine good safety profiles with strong immunogenicity and constituted a safe alternative to inactivated vaccines or attenuated vaccines.

Table 1.

List of partial virus-like particle (VLP) vaccines derived from different viruses

Family Virus Composition Expression system Ref.
Picornaviridae FMDV (type O1) VP0, VP1, and VP3 B/IC Mohana Subramanian et al. (2012)
FMDV (Asia1) VP0, VP1, and VP3 E. coli Guo et al. (2013)
Enterovirus 71 VP0, VP1, and VP3 B/IC Chung et al. (2006)
Enterovirus 71 VP0, VP1, and VP3 B/IC Chung et al. (2008)
Enterovirus 71 VP0, VP1, and VP3 Yeast Li et al. (2013a)
Circoviridae PCV2 Cap E. coli Wu et al. (2012)
PCV2 Cap Mammalian cells Chi et al. (2014)
PCV2 Cap E. coli Yin et al. (2010)
PCV2 Cap B/IC Bucarey et al. (2009)
PCV2 Cap B/IC Fort et al. (2008)
PCV2 Cap B/IC Martelli et al. (2011)
Papillomaviridae HPV16 L1 Mammalian cells Pastrana et al. (2001)
HPV L1 and L2 Yeast Bazan et al. (2009)
HPV L1 and L2 Plant Pineo et al. (2013)
HPV16 L1 B/IC Vidyasagar et al. (2014)
Filoviridae EBV/MBV VP40, GP, and NP B/IC Warfield et al. (2007b)
EBV VP40, NP, and GP B/IC Warfield et al. (2007a)
EBV GP and VP40 Mammalian cells Warfield et al. (2003)
MARV VP40 and GPs Mammalian cells Swenson et al. (2004)
MARV VP40 and GPs Mammalian cells Swenson et al. (2005)
ZEBV GP and Fc Mammalian cells Konduru et al. (2011)
PEMCV P1, 2A, 3C B/IC Jeoung et al. (2010)
PEMCV P1, 2A, 3C B/IC Jeoung et al. (2011)
Paramyxoviridae NDV NP, M, HN, F Avian cells McGinnes et al. (2010)
NDV F and M1 B/IC Park et al. (2014)
Bunyaviridae RVFV Nucleocapsids Mammalian cells Naslund et al. (2009)
RVFV N, Gn, and Gc B/IC Liu et al. (2008)
RVFV L, N, and M Mammalian cells Habjan et al. (2009)
UUK virus GN and GC Mammalian cells Overby et al. (2006)
Hantaviruses N, Gn, Gc glycoproteins Mammalian cells Acuna et al. (2013)
Orthomyxoviridae H5N1 HA, NA, M1, and M2 Mammalian cells Wu et al. (2010)
H5N1 HA, NA, and M1 B/IC Kang et al. (2009)
H5N1 HA and NA B/IC Bright et al. (2008)
H5N1 Gag, HA, and NA B/IC Haynes et al. (2009)
H1N1 NA and M1 Mammalian cells Easterbrook et al. (2012)
H1N1 HA and M1 B/IC Quan et al. (2010)
H1N1 HA, NA, and M1 B/IC Pyo et al. (2012)
H9N2 (AIV) HA and M1 B/IC Lee et al. (2011)
H3N2 HA and M1 B/IC Lee et al. (2013)
H7N9 HA, NA, and M1 B/IC Smith et al. (2013)
Reoviridae Rotavirus VP2, VP6, and VP7 B/IC Kim et al. (2002)
Rotavirus VP2, VP6, and VP7 B/IC Vieira et al. (2005)
Rotavirus VP2, VP6 B/IC Agnello et al. (2006)
Rotavirus VP2, VP6 B/IC Mena et al. (2005)
Rotavirus VP2, VP6, and VP7 B/IC Clark et al. (2009)
BTV VP2, VP3, VP5, VP7 B/IC Stewart et al. (2013)
Parvoviridae CPV VP2 E. coli Xu et al. (2014a)
GPV VP2 B/IC Chen et al. (2012)
GPV VP1, VP2, VP3 B/IC Ju et al. (2011)
PPV VP2 Mammalian cells Chen et al. (2011)
HPB19 virus VP2 E. coli Sanchez-Rodriguez et al. (2012)
HPB19 virus VP1, VP2 Yeast Chandramouli et al. (2013)
Polyomaviridae GHPV VP1 B/IC Zielonka et al. (2006)
GHPV VP1 and VP2 Yeast Zielonka et al. (2006)
JC polyomavirus VP1 B/IC Goldmann et al. (1999)
JC polyomavirus VP1 Yeast Sasnauskas et al. (2002)
Polyomavirus VP1 E. coli Shin and Folk (2003)
MCV VP1 B/IC Touze et al. (2010)
Flaviviridae JEV Envelope protein Mammalian cells Chiou et al. (2008)
JEV prM and envelope proteins B/IC Yamaji and Konishi (2013)
Dengue virus prM and envelope proteins Mammalian cells Zhang et al. (2011)
Dengue virus-2 prM and envelope proteins Yeast Liu et al. (2010)
West Nile virus Envelope glycoprotein E. coli Spohn et al. (2010)
West Nile virus prM and envelope proteins Mammalian cells Ohtaki et al. (2010)
HCV Core protein E. coli Lorenzo et al. (2001)
HCV Core protein B/IC Li et al. (2013b)
HCV E1 and E2 proteins Mammalian cells Garrone et al. (2011)
HCV E1, E2 protein B/IC Murata et al. (2003)
Caliciviridae RHDV VP60 B/IC Gromadzka et al. (2006)
RHDV VP60 B/IC Nagesha et al. (1995)
RHDV VP60 B/IC Young et al. (2006)
NV NV1 Mammalian cells Harrington et al. (2002)
NV Capsid protein Plant Lai and Chen (2012)
NV Capsid protein Plant Santi et al. (2008)
FCV VP1 B/IC Di Martino et al. (2007)

FMDV foot and mouth disease virus; B/IC baculovirus/insect cells, namely cloned genes into baculovirus constructs and infect insect cells to generate related protein for forming VLPs; PCV2 porcine circovirus type 2, CP capsid protein, ORF open reading frame, HPV 16 human papillomavirus 16, EBV Ebola virus, MBV Marburg virus, ZEBV Zaire Ebola virus, MARV Marburg virus, GPs glycoprotein, PEMCV porcine encephalomyocarditis virus, NDV Newcastle disease virus, RVFV Rift Valley fever virus, UUK virus uukuniemi virus (Bunyaviridae), HA hemagglutinin, NA neuraminidase, M1 matrix 1 protein, M2 matrix 2 protein, BTV Bluetongue virus, CPV canine parvovirus, GPV goose parvovirus, PPV porcine parvovirus, HPBV19 human parvovirus B19 virus, GHPV goose hemorrhagic polyomavirus; MCV a new human polyomavirus, known as Merkel cell polyomavirus; JEV Japanese encephalitis virus, HCV hepatitis C virus, RHDV rabbit hemorrhagic disease virus, NV Norwalk virus, FCV feline calicivirus

Prophylactic VLP vaccine

As a novel type of vaccine, VLPs offer a solution to the above problem, primarily because of its biological properties (Young et al. 2006). First, VLPs have a high level of safety without concerns of biosecurity since no viral genetic components are introduced during its production. Second, VLPs present conformational epitopes, which are arranged repeatedly on the surface. With such an arrangement, VLPs are more similar to the native virus; thus, VLPs can easily induce strong B cell responses in the absence of adjuvants (Ramqvist et al. 2007). Third, VLP vaccine can rapidly cope with epidemic viral diseases because of its short time required for proceeding from design to expression. For example, the development and preparation of VLP vaccines was only 8 weeks after the outbreak of influenza, but more than 5 months for attenuated vaccines (Cox 2005). Finally, VLPs, as pathogen-associated molecular patterns (PAMPs), can be recognized by pattern recognition receptors (PRRs) such as the Toll-like receptors (TLRs) of host cells and captured by antigen-presenting cells (e.g., dendritic cells) (Fakruddin et al. 2007), then can be taken up and processed via the MHC class I pathway (cross-presentation) for activation of CD8+ T cells, which are essential for the clearance of intracellular pathogens such as viruses. In addition, VLPs are inherent in a suitable size, and VLPs can also be taken up by dendritic cells (DCs) as exogenous antigens for processing and presentation by MHC class II and for directly promoting DC maturation and migration, essential for stimulation of the innate immune response, whose stimulate immunity patterns are similar as in the original virus (Grgacic and Anderson 2006; Keller et al. 2010; Ponterio et al. 2013; Raghunandan 2011) (Fig. 1). In this way, VLPs may have the advantages over the cognate live viruses for immune activation because some viruses that replicate in DCs are known to block activation and maturation of the cell through expression of particular viral proteins, while some VLPs which can resemble infectious viruses and retain their receptor binding regions are able to be taken up by antigen-presenting cells for class I presentation systems.

Fig. 1.

Fig. 1

Virus-like particles (VLPs) mimic the overall structure of virus particles, are recognized readily by the immune system, and present viral antigens in a similar pathway to authentic conformation inducing strong immune responses

The advance in molecular biology improved the expression system of the VLP-based vaccines. The number of reports on newly obtained VLPs has grown proportionally with the systems developed for the expression of these particles, and VLPs have been successfully used as a vaccine platform to which additional components of the virus or other virus or pathogen are attached or inserted and shown to stimulate both cellular and humoral immunity no matter how much the capsid protein the VLPs is composed of (Fig. 2). With the advantages in safe production process, short production time, and many available expression systems, there are numerous VLP-based vaccines under development and some are even currently in the market. Further examples of VLPs as vaccine candidates are shown in Table 1.

Fig. 2.

Fig. 2

A schematic diagram of the classification of different virus-like particles based on the number of viral surface proteins and the existence of lipid envelopes or not (adapted from Lua et al. 2014). For non-enveloped VLPs: (a) the single layered non-enveloped VLPs assembled by one protein (e.g., hepatitis B core antigen VLPs (Roose et al. 2013) and CPV VP2-VLPs (Xu et al. 2014)); (b) The single-layered non-enveloped VLPs assembled by two proteins (e.g., SARS coronavirus VLPs (Mortola and Roy 2004)); (c) Two-layered non-enveloped VLPs assembled by two proteins (e.g., papillomavirus L1 and L2 VLPs (McKee et al. 2015)); And (d) twolayered non-enveloped VLPs assembled by multiple proteins (e.g., FMDV-VLPs (Guo et al. 2013)); (e)The triple-layered VLPs assembled by multiple proteins (e.g., bluetongue virus (Stewart et al. 2013) and rotavirus VLPs (Parez et al. 2006)). For enveloped VLPs: (f) single-layered VLPs consisted of one protein (e.g., influenza virus ectodomain of matrix protein 2 (M2e) VLPs (Lee et al. 2014)); (g) Single-layered VLPs consisted of two protein (e.g., hantaviruses VLPs (Acuna et al. 2013)); (h) Two-layered VLPs consisted of two protein (e.g., hepatitis C VLPs (Bellier and Klatzmann 2013)), and (i) Two-layered VLPs consisted of multiple proteins (e.g., SARS coronavirus VLPs (Ho et al. 2004))

As shown in some studies, the VLPs can be used as an ideal carrier platform for foreign B cell and/or T cell epitopes to display practically any antigen in a highly immunogenic, multivalent format in vaccination experiments. In addition that some viral epitopes are relatively conserved, the epitope genes can be obtained through genetic engineering and subcloned into the plasmid vector. Thereafter, the genes are genetically incorporated into the gene sequence of coat/capsid protein of VLPs, and then VLPs can present the exogenous antigens by fusion expression (Arora et al. 2012; Kawano et al. 2013, 2014; Tyler et al. 2014; Ye et al. 2014). A simpler mean to produce such chimeric vaccines can be achieved by genetical insert rather than by chemical linking the peptide epitopes to the VLPs, and chimeric epitope peptides can be repeatedly arranged on the surface of VLPs, which have potential prospects (Peabody et al. 2008).

As proven by numerous experiments, the insertion of a foreign peptide sequence into the upstream or downstream of the viral structure proteins genes has little effect on the assembly of viruses such as HPV (Teunissen et al. 2013), hepatitis B virus (HBV) (Brandenburg et al. 2005; Mihailova et al. 2006), AMCV (Arcangeli et al. 2014), Rotavirus (Cortes-Perez et al. 2010), RNA bacteriophage Qβ (Tissot et al. 2010), and canine parvovirus (CPV) (Gilbert et al. 2004). Therefore, the insertion of foreign genes into VLPs through gene recombination technology renders VLPs suitable as an antigen presentation tool. HBc VLPs are the first reported VLPs that present exogenous antigens, and HBc protein dimers have a highly symmetrical and relatively stable icosahedral structure. Several sites to which exogenous antigen epitopes can be incorporated into the sequences of their proteins include N- and C-terminal and major immunodominant region (MIR) (Jegerlehner et al. 2002). Except viral epitopes, VLPs can also present immune-related factors such as CD40 ligand (Zhang et al. 2010) and cytotoxic T cell epitopes (Tartour et al. 2002), as well as immune-stimulating factors, such as antimicrobial peptides, interferons (IFN), proinflammatory cytokines, and chemokines when recognizing PAMPs in the early period of innate immunity (Liu et al. 2000; Vacher et al. 2013) (Table 2).

Table 2.

The examples of VLP-based delivery for exogenous antigen delivery or epitope

Virus Expression system Modification strategies Chimeric antigen(s) Ref.
Phage AP205 E. coli Chemical conjugation α-Helic regions of HIV gp41 Pastori et al. (2012)
HBV E. coli Fusion expression CFP-10 of tuberculosis Cortes-Perez et al. (2010)
HBV E. coli Fusion expression B and T cell epitopes of HCV Mihailova et al. (2006)
HBV HEK 293T Fusion expression VP4N20 of EV71 Cheong et al. (2009)
HBV E. coli Fusion expression CTL epitope Takeda et al. (2004)
HBV E. coli Fusion expression MAGE-3 Kazaks et al. (2008)
HBV E. coli Fusion expression CTL epitopes of HBV and HCV Sominskaya et al. (2010)
HBV E. coli Fusion expression Rubella virus E1 glycoprotein Skrastina et al. (2013)
HBV E. coli Fusion expression 4 HBx-derived epitopes* Ding et al. (2009)
HBV E. coli Fusion expression SP55 or SP70 epitope of EV71 Ye et al. (2014)
HBV E. coli Fusion expression (EDIII) of dengue viruses-2 Arora et al. (2012)
HEV B/IC Fusion expression B cell epitope Niikura et al. (2002)
Retrovirus 293T cells Fusion expression E1 and E2 envelope GP Huret et al. (2013)
Qβ bacteriophage E. coli Chemical conjugation CCR5 Hunter et al. (2009)
Qβ bacteriophage E. coli Chemical conjugation V3 and ECL2 of HIV Peabody et al. (2008)
Qβ bacteriophage E. coli Chemical conjugation Nicotine Cornuz et al. (2008)
Bacteriophage P22 E. coli Chemical conjugation Nucleoprotein of influenza Patterson et al. (2013)
Bacteriophage MS2 E. coli Fusion expression L2 peptide of HPV16 and HPV31 Tyler et al. (2014)
FHV B/IC Fusion expression ANTXR2 VWA Manayani et al. (2007)
BPV B/IC Fusion expression CCR5 Chackerian et al. (1999)
BPV B/IC Fusion expression CTL epitopes Liu et al. (2000)
Hepatitis E B/IC Fusion expression p18 peptide Jariyapong et al. (2013b)
HPV-16 Plant Fusion expression M2e2-24, M2e2-9 Matic et al. (2011)
HPV-16 Yeast Conjugation M2 from influenza A Ionescu et al. (2006)
RSV Insect Fusion expression N. cani surface protein Deo et al. (2013)
SV40 Insect Fusion expression CTL epitope from influenza A Kawano et al. (2014)
Rotavirus B/IC Fusion expression 14 amino acid epitope Peralta et al. (2009)
HaPyV Yeast Fusion expression GP33 CTL epitope of LCMV Mazeike et al. (2012)
Influenza virus A B/IC Fusion expression ESAT-6 Krammer et al. (2010)
IBDV Yeast Fusion expression HPV-16 E7 Martin Caballero et al. (2012)
PPV HEK-293 cells Fusion expression Residues165-200 Pan et al. (2008)

B/I baculovirus-insect cell system, CFP-10 Mycobacterium tuberculosis antigen culture filtrate protein 10, CCR5 the most important coreceptors that HIV used in the early stages of infection, BPV bovine papillomavirus virus, CP coat protein, HVJ hemagglutinating virus of Japan, SV simian virus 40, HBVc-VLP hepatitis B virus core protein virus-like particles, PA protective antigen of anthrax; ESAT-6 early secreted antigenic target-6, an important Mycobacterium tuberculosis T-cell antigen; p18 peptide V3 loop of HIV-1 gp120, VP4N20 the first 20 amino acids at the N-terminal of VP4 of EV71 genotype C4, MAGE-3 cancer-germline gene, FHV flock house virus; ANTXR2 VWA protective antigen-binding von Willebrand A domain of the ANTXR2 cellular receptor, CTL epitopes including human PV16 E7 protein, HIV IIIB gp120 P18, Nef, and reverse transcriptase (RT) proteins, and an HPV16 E7 linear B epitope; 4 HBx-derived epitopes* HBx(52–60), HBx(92–100), HBx(115–123) and a novel subdominant cytolytic T lymphocyte (CTL) epitope HBx(140–148); HaPyV hamster polyomavirus, LCMV lymphocytic choriomeningitis virus, M2e 2-24 ectodomain of the M2 protein (M2e), M2e 2-9 a shorter version of M2e containing the N-terminal highly conserved epitope, N. cani Neospora caninum, RSV rous sarcoma virus, residues 165-200 residues 165-200 from the Porcine circovirus 2 (PCV2) virus nucleoprotein, IBDV infectious bursal disease virus

*Four different dominant sequence derived from hepatitis B virus epitopes protein

VLP-based therapeutic vaccines

As an epitope vector, VLPs can present not only foreign antigens but also self-antigens. Several VLPs have been studied as vehicles for use in immune therapy especially as therapeutic vaccine to treat chronic diseases and cancer (Ramqvist et al. 2007). For immunotherapeutics, which represent a new promising class of vaccines developed to treat chronic diseases (Fulurija et al. 2008; Rohn et al. 2006; Sonderegger et al. 2006; Spohn et al. 2007), therapeutic vaccination has demonstrated that it involves active clearance of an infectious agent, infected cells, and especially tumor cells through breaking immune tolerance or bypassing the mechanisms by which the disease has evolved the immune system. It has been known that the CD8+ T lymphocytes (CTL) have played an important role in the control of tumor growth, so stimulation of specific CTL therefore represents one major goal in the design of cancer vaccines (Al-Barwani et al. 2014; Speiser et al. 2010; Zamarin and Postow 2015). VLPs can be good antigen delivery systems that efficiently introduce exogenous molecules into the MHC class I pathway, which showed to be important mediators of antitumor immunity in various animal models because of its ability to elicit tumor-specific CTL (Jegerlehner et al. 2002).

In addition, VLPs have the potential to become therapeutic vaccine to treat chronic diseases through molecular clone technology, thus overcoming the natural tolerance of the immune system and stimulating the immune system to create specific antibodies toward self-proteins (Spohn et al. 2005). With the identification of the pathophysiology of several chronic diseases, a few key pathogenic determinants have been deciphered. Such determinant-related proteins can be presented via VLPs by means of chemically cross-linking them to the surface of virus-like particles or genetic recombination to display. As a result, specific autoantibodies are produced, which are of considerable significance in ameliorating or even curing these chronic diseases (Tissot et al. 2010).

For example, to avoid a wide range of inflammatory reactions involving amyloid-β (Aβ)-specific B cells, Aβ1-6 peptide (DAEFRH) and RNA phage Qβ-VLPs (VLPs) are covalently linked when treating Alzheimer’s disease (AD) with VLP vaccines causing the production of antibodies specific to Aβ. The aforementioned strategies for the design of Aβ vaccine have been the subject of clinical trials (Chackerian et al. 2006; Wiessner et al. 2011). Other vaccines have been designed through similar methods, such as NGFQβ-VLPs, which are specific to nerve growth factors (NGF) for treating chronic pain (Rohn et al. 2011); bacteriophage Qβ-C-TNF4-23 VLPs, which are specific to TNF-α for rheumatoid arthritis (Spohn et al. 2007); and Qβ-EC1 VLPs specific to CCR5 for preventing HIV infection (Hunter et al. 2009). Another design strategy for the development of chimeric VLP vaccines is the production of corresponding antibodies by stimulation from immunity-related antigens present on the surface of VLPs. This is achieved by inserting pathogen-related gene expression proteins into the epitopes of particular viral structure proteins (Ogasawara et al. 2006; Pumpens and Grens 2001; Yao et al. 2004). For example, Cubas et al. insert surface glycoprotein-Trop2 expressing excessively on the surface of tumor cells into the SIV gag gene, which produced a chimeric vaccine to suppress the growth of tumor, because immunization with chimeric Trop2 VLPs can break tolerance to this self-protein and generate a specific cellular and humoral immune response significantly increasing the population of CD4+ and CD8+ T cells as well as natural killer (NK) and natural killer T cells (NKT) inside the tumor tissue, and these effects translated into a significant reduction in tumor growth (Cubas et al. 2011). Schiller et al. inserted an extracellular loop zone of chemokine receptor CCR5 of mice into bovine papillomavirus L1 capsid protein to produce autoantibodies to block CCR5. This step prevented HIV from entering cells and proliferation in cells (Chackerian et al. 1999). This approach is also employed to block B cells’ tolerance to autoantigens for the treatment of some chronic diseases (e.g., rheumatoid arthritis, osteoporosis, experimental autoimmune encephalitis, systemic lupus erythematosus, myocarditis, atherosclerosis, hypertension, Alzheimer’s disease, and obesity) by inducing therapeutically effective neutralizing autoreactive autoantibodies and is recognized as a potential treatment option for chronic diseases (Jennings and Bachmann 2008).

Chronic diseases are primarily caused by multiple pathogenic factors which make them difficult to cure; nevertheless, these therapeutic vaccines based on VLPs can produce antibodies aiming at autoantigens such as amyloid-β (Zamora et al. 2006), angiotensin II (Ang II) (Tissot et al. 2008), nerve growth factor (Rohn et al. 2011), allergens (Jegerlehner et al. 2002), and ghrelin (Andrade et al. 2013), which can help reduce the risk or cure certain diseases that had been at different stages of clinical trials (Jennings and Bachmann 2009; Roldao et al. 2010).

VLPs as versatile delivery vehicles

Ideal biological vectors should have the following biological characteristics: biocompatibility, solubility, and uptake efficiency, with targeted delivery and high drug loading. As a nanoscale material, VLPs have high potential in drug delivery (Boisgérault et al. 2002; Schott et al. 2011) (Table 3). VLPs fit the aforementioned demands in a certain degree among plenty of studies. First, VLPs are easy to be produced in large-scale quantities using the existing expression systems either as enveloped or nonenveloped VLPs (Fig. 3). Second, VLPs are capable of targeting the corresponding cell transport with its surface ligands by modification on the gene level (gene insertion) (Ungaro et al. 2013) or the protein level (chemical coupling) (Wei et al. 2009). Third, VLPs have good carrying capacity because of its large surface area and numerous amino acid residues on the surface (Patel and Swartz 2011). Fourth, VLPs are self-assembled by viral structure proteins under proper conditions which looks more like a protein cage with a large cavity space that can encapsulate numerous biological molecules, and as a result, expanded these molecules’ applications (Wang et al. 2011; Yang and Burkhard 2012). Finally, VLPs have thermodynamic stability because of its dodecahedral or icosahedral structure. To sum, VLPs have emerged as multifunctional platform systems for the development of bioderived nanomaterials and have good potential for application in drug delivery, genetic therapy, and other fields (Fig. 4).

Table 3.

VLPs derived from viral structural proteins as vehicle systems for biomedical applications

Virus Expression system VLP composition Structural information Cargo Ref.
Bacteriophage
 φCb5 Yeast Coat protein Icosahedral tRNA, nanoparticles, mRNA Freivalds et al. (2013)
 MS2 Yeast Capsid protein Icosahedral mRNA Li et al. (2014)
 MS2 Yeast Capsid protein Icosahedral Nonmethylated CG motifs Storni et al. (2004)
 MS2 E. coli Capsid protein Icosahedral HIV-1 gag mRNAs Sun et al. (2011)
 MS2 E. coli Coat protein Icosahedral IgG-binding Z domain Brown et al. (2009)
 MS2 E. coli Coat protein Icosahedral Antisense ODNs Wu et al. (2005)
 Qβ E. coli Coat protein Icosahedral CelB glycosidase Patterson et al. (2012)
Rotavirus E. coli VP6 Icosahedral DOX Zhao et al. (2011)
Rotavirus B/IC VP2, VP4, VP6 Icosahedral GFP Charpilienne et al. (2001)
Rotavirus B/IC VP2 Icosahedral GFP Cortes-Perez et al. (2010)
JC PyV E. coli VP1 Icosahedral GFP or tk gene Chen et al. (2010)
JC PyV E. coli VP1 Icosahedral Exogenous plasmid DNA Lin et al. (2014)
JCPyV Yeast VP1 Icosahedral IL-10 shRNA Chou et al. (2010)
HaPyV E. coli VP1 Icosahedral Plasmid DNA Voronkova et al. (2007)
CPV B/IC VP2 Icosahedral EGFP Gilbert et al. (2004)
HBV E. coli tHBcAg Icosahedral preS1 ligand Lee et al. (2012)
HBV B/IC HBcAg protein Icosahedral DNA fragment Brandenburg et al. (2005)
MrNv E.coli Capsid protein Icosahedral Plasmid DNA Jariyapong et al. (2013a)
HCV B/IC Core protein Icosahedral RGD peptide and IFN-α2a Li et al. (2013b)
CCMV Plant cell Capsid protein Spherical RNA derived from SINV Azizgolshani et al. (2013)
Simian virus 40 E. coli VP1 Spherical Quantum dots Li et al. (2009)
HIV-1 Mammals Nef7 Spherical HSV-1 TK gene Peretti et al. (2005)
HPV B/IC L1, L2 Icosahedral Plasmid DNA Malboeuf et al. (2007)

VP6 rotavirus capsid protein; DOX doxorubicin, an anticancer drug; ONDs antisense oligodeoxynucleotides, GFP green fluorescent protein, EGFP enhanced green fluorescent protein, RCNMV red clover necrotic mosaic virus, CPV canine parvovirus, liver-specific ligand a liver-specific ligand, tHBcAg truncated HBcAg, MrNv Macrobrachium rosenbergii nodavirus, DNA a small hepatitis B virus surface antigene (SHBs)-specific sequence, RGD Arg-Gly-Asp, PyV polyomavirus, HaPyV hamster polyomavirus, CCMV cowpea chlorotic mottle virus (a plant virus), RCNMV red clover necrotic mosaic virus, SINV a mammalian virus-Sindbis virus, HSV-1 TK gene herpes simplex virus-1 thymidine kinase gene

Fig. 3.

Fig. 3

Production process of VLPs derived from enveloped or nonenveloped through spontaneous self-assembling

Fig. 4.

Fig. 4

A schematic representation of assembly VLPs derived from enveloped or nonenveloped viruses which are efficient nanocarriers for cargo delivery

Drug carriers

The greatest adverse effect of chemotherapy in cancer treatment is toxicities to normal tissues, which severely limits the therapeutic effects of anticancer drugs. Anticancer drugs are tethered via the amino acid residues on the surface of VLPs, particularly the icosahedral VLPs, such as HBV, bacteriophage MS2, bacteriophage Qβ, and some dodecahedral VLPs, such as adenovirus (Ad) VLPs (Zochowska et al. 2009) with thermodynamic stability. Through mild chemical coupling reagents, anticancer drugs such as adriamycin and aleomycin can be loaded onto the surface of the aforementioned VLPs with hydrazone bonds created by amino acid residues. In addition, anticancer drug molecules can also be encapsulated into VLPs by the reversible process of self-assembly with changes of the external conditions, such as specific ionic concentration (Huhti et al. 2013). Moreover, VLPs have a proper particle size, good distribution, and biocompatibility as well as ligands on its surface for invasion into special cells. These ligands bind with receptors on the cell surface to help different VLPs to specifically deliver the drugs to various target cells mimicking the native virus. Common ligands are RGD motif (Marelli et al. 2013), transferrin (Singh et al. 2006), and so forth. At the same time, the anticancer drug bioavailability can be improved, that is, the improvement of the ability of the targeted transport and accumulation in target cells. Therefore, VLPs can be acceptable as effective biological vectors for carrying drugs.

VLPs as research surrogates

Studies on highly infective viruses and those without good cell culture systems in vitro or model animals, e.g., Ebola and Marburg viruses (Warfield et al. 2005) and human norovirus (Souza et al. 2013), must be performed under good experimental conditions. VLPs of these virus reserve the conformation of viral capsid protein, presenting the same ligands as those in the invading natural virus, which makes VLPs suitable as surrogates for basic research of these biosafety level 4 restricted viruses (Buonaguro et al. 2013). Therefore, VLPs are a good surrogate for simulating cell infection and to interact with host cells and viruses by labeling VLPs through the appropriate modification of the VLPs’ surface in chemical or genetic ways (Tscherne et al. 2010).

So far, significant progress has been made in the study of characterizing virus-specific surface receptors, pathways of virus entry, and the mechanisms of virus assembly by utilizing VLPs as a surrogate. The mechanism of the infectivity of HuNoV has demonstrated that amino acid residues in the P domain of the VP1 protein are responsible for the specificity of receptor binding, and this data comes from the study of surrogates—HuNoV virus-like particle (VLP) (Tan et al. 2003, 2008; Tan and Jiang 2005). The interaction of capsid ORF2 protein of HEV with heparan sulfate proteoglycans (HSPG) has been proven by Kalia et al., and the researchers observed these results through the expression of hepatitis E virus ORF2 capsid protein in the insect cell Tn5 and the self-assembly of HEV-LP in vitro (Kalia et al. 2009). To observe the process of absorption, entrance, and intracellular transport of the virus, HIV-VLPs were assembled in vitro by Jouvenet et al. using green fluorescent protein (GFP) and Gag, the major structural component of HIV-1, and HIV-VLPs have a very similar morphology compared with the VLPs assembled by the Gag protein (Jouvenet et al. 2008). The single VLPs are employed to simulate a single virus. This method avoids the interference caused by the intracellular replication of the natural virus to the infection of a single virus (Pokorski et al. 2011; Wei et al. 2011). Through the aid of an advanced confocal microscope, Goreliket et al. observed the cellular surface morphology and fluorescent signals during the period of viral entry by a single Cy3-labeled polyoma VLP. This technique can explain viral endocytosis and establish a model for the observation of endocytosis of other nanoparticles (Gorelik et al. 2002). Buonaguro et al. studied the interaction between virus and cellular receptors through HIV Pr55 gag-VLPs with the embedded HIV gp140 envelope proteins (i.e., embedded with extracellular functional regions gp120 and gp41) by double-transfected bacillus in vitro. Tscherne et al. created BlaM1 VLPs through the recombinant expression of beta-lactamase reporter protein (Bla) and influenza matrix protein-1. When the VLPs adsorbed on the target cells, Bla was dissociated from BlaM1 VLPs and entered the cells. The released Bla could then be detected by flow enzyme-linked immunosorbent assay (ELISA) or positive ELISA, but it could not be detected intracellularly in the presence of antibodies of influenza virus because Bla cannot enter the cell. Likewise, this method is employed to detect Ebola (EBOV) and Marburg (MARV) viruses. A VLP was created in vitro by inserting a GFP protein into the N-terminal of the VP2 protein of CPV. This VLP is considered as a probe that tracks the interaction between CPV and host cells. With morphological, biophysical, and antigenic properties similar to those of putative virions, VLPs represent a novel model for the study of virus-host interactions and virus assembly.

Conclusion and future perspectives

In the past 30 years, VLPs have been widely used in the biology field. The VLPs assembled by viral envelope glycoprotein or capsid proteins have been proven to have the ability to induce humoral and cellular immune responses in experimental mice, nonprimates, and even humans (Raghunandan 2011). VLPs, whatever the form of prophylactic vaccine (recombinant VLP vaccine or chimeric VLP vaccine) or the form of therapeutic vaccines is, have the potential to be used as a new generation of vaccine candidates against various viral infections (Guillen et al. 2013).

At present, virology, molecular biology, protein chemistry, inorganic chemistry, and materials science are being highly integrated in the pursuit of new materials with controlled physical properties. VLPs have shown to be amenable to both chemical and genetic modifications of their inner cavities as well as their outer surfaces. Owning to their versatile hierarchical assembly, VLPs have been providing a new approach for the targeted transportation of therapeutic drugs and other biological materials. Overall, in this article, we reviewed the development and current situation of VLPs as vaccines and carriers, thereby contributing to the understanding and discovery of new biological applications for VLPs.

Acknowledgments

This research was supported by grants from the National Science and Technology Support Program (2013BAD12B00), International Science &Technology Cooperation Program of China (2014DFA31890), and the Fundamental Research Funds for the Chinese Academy of Agricultural Sciences (2015ZL062).

Conflict of interest

The authors declare that they have no competing interests.

References

  1. Acuna R, Cifuentes-Munoz N, Marquez C, Bulling M, Klingstrom J, Mancini R, Lozach PY, Tischler ND. Hantavirus Gn and Gc glycoproteins self-assemble into virus-like particles. J Virol. 2013 doi: 10.1128/JVI.03118-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agnello D, Herve CA, Lavaux A, Darniot M, Guillon P, Charpilienne A, Pothier P. Intrarectal immunization with rotavirus 2/6 virus-like particles induces an antirotavirus immune response localized in the intestinal mucosa and protects against rotavirus infection in mice. J Virol. 2006;80(8):3823–3832. doi: 10.1128/JVI.80.8.3823-3832.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Al-Barwani F, Donaldson B, Pelham SJ, Young SL, Ward VK. Antigen delivery by virus-like particles for immunotherapeutic vaccination. Ther Deliv. 2014;5(11):1223–1240. doi: 10.4155/tde.14.74. [DOI] [PubMed] [Google Scholar]
  4. Amanna IJ, Slifka MK. Wanted, dead or alive: new viral vaccines. Antiviral Res. 2009;84(2):119–130. doi: 10.1016/j.antiviral.2009.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Andrade S, Pinho F, Ribeiro AM, Carreira M, Casanueva FF, Roy P, Monteiro MP. Immunization against active ghrelin using virus-like particles for obesity treatment. Curr Pharm Des. 2013;19(36):6551–6558. doi: 10.2174/13816128113199990506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Arcangeli C, Circelli P, Donini M, Aljabali AA, Benvenuto E, Lomonossoff GP, Marusic C. Structure-based design and experimental engineering of a plant virus nanoparticle for the presentation of immunogenic epitopes and as a drug carrier. J Biomol Struct Dyn. 2014;32(4):630–647. doi: 10.1080/07391102.2013.785920. [DOI] [PubMed] [Google Scholar]
  7. Arora U, Tyagi P, Swaminathan S, Khanna N. Chimeric hepatitis B core antigen virus-like particles displaying the envelope domain III of dengue virus type 2. J Nanobiotechnol. 2012;10:30. doi: 10.1186/1477-3155-10-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Azizgolshani O, Garmann RF, Cadena-Nava R, Knobler CM, Gelbart WM. Reconstituted plant viral capsids can release genes to mammalian cells. Virology. 2013;441(1):12–17. doi: 10.1016/j.virol.2013.03.001. [DOI] [PubMed] [Google Scholar]
  9. Bayer ME, Blumberg BS, Werner B. Particles associated with Australia antigen in the sera of patients with leukaemia, Down’s syndrome and hepatitis. Nature. 1968;218(5146):1057–1059. doi: 10.1038/2181057a0. [DOI] [PubMed] [Google Scholar]
  10. Bazan SB, de Alencar Muniz Chaves A, Aires KA, Cianciarullo AM, Garcea RL, Ho PL. Expression and characterization of HPV-16 L1 capsid protein in Pichia pastoris. Arch Virol. 2009;154(10):1609–1617. doi: 10.1007/s00705-009-0484-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bellier B, Klatzmann D. Virus-like particle-based vaccines against hepatitis C virus infection. Expert Rev Vaccines. 2013;12(2):143–154. doi: 10.1586/erv.13.10. [DOI] [PubMed] [Google Scholar]
  12. Blumberg BS, Alter HJ, Visnich S. A “new” antigen in leukemia sera. JAMA. 1965;191:541–546. doi: 10.1001/jama.1965.03080070025007. [DOI] [PubMed] [Google Scholar]
  13. Boisgérault F, Morón G, Leclerc C. Virus-like particles: a new family of delivery systems. Expert Rev Vaccines. 2002;1(1):101–109. doi: 10.1586/14760584.1.1.101. [DOI] [PubMed] [Google Scholar]
  14. Brady JN, Consigli RA. Chromatographic separation of the polyoma virus proteins and renaturation of the isolated VP1 major capsid protein. J Virol. 1978;27(2):436–442. doi: 10.1128/jvi.27.2.436-442.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brandenburg B, Stockl L, Gutzeit C, Roos M, Lupberger J, Schwartlander R, Gelderblom H, Sauer IM, Hofschneider PH, Hildt E. A novel system for efficient gene transfer into primary human hepatocytes via cell-permeable hepatitis B virus-like particle. Hepatology. 2005;42(6):1300–1309. doi: 10.1002/hep.20950. [DOI] [PubMed] [Google Scholar]
  16. Bright RA, Carter DM, Daniluk S, Toapanta FR, Ahmad A, Gavrilov V, Massare M, Pushko P, Mytle N, Rowe T, Smith G, Ross TM. Influenza virus-like particles elicit broader immune responses than whole virion inactivated influenza virus or recombinant hemagglutinin. Vaccine. 2007;25(19):3871–3878. doi: 10.1016/j.vaccine.2007.01.106. [DOI] [PubMed] [Google Scholar]
  17. Bright RA, Carter DM, Crevar CJ, Toapanta FR, Steckbeck JD, Cole KS, Kumar NM, Pushko P, Smith G, Tumpey TM, Ross TM. Cross-clade protective immune responses to influenza viruses with H5N1 HA and NA elicited by an influenza virus-like particle. PLoS One. 2008;3(1):e1501. doi: 10.1371/journal.pone.0001501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Brown SD, Fiedler JD, Finn MG. Assembly of hybrid bacteriophage Qbeta virus-like particles. Biochemistry. 2009;48(47):11155–11157. doi: 10.1021/bi901306p. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bucarey SA, Noriega J, Reyes P, Tapia C, Saenz L, Zuniga A, Tobar JA. 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(42):5781–5790. doi: 10.1016/j.vaccine.2009.07.061. [DOI] [PubMed] [Google Scholar]
  20. Buonaguro L, Tagliamonte M, Visciano ML. Chemokine receptor interactions with virus-like particles. Methods Mol Biol. 2013;1013:57–66. doi: 10.1007/978-1-62703-426-5_5. [DOI] [PubMed] [Google Scholar]
  21. Burns CC, Diop OM, Sutter RW, Kew OM. Vaccine-derived polioviruses. J Infect Dis. 2014;210(Suppl 1):S283–S293. doi: 10.1093/infdis/jiu295. [DOI] [PubMed] [Google Scholar]
  22. Chackerian B. Virus-like particles: flexible platforms for vaccine development. Expert Rev Vaccines. 2007;6(3):381–390. doi: 10.1586/14760584.6.3.381. [DOI] [PubMed] [Google Scholar]
  23. Chackerian B, Lowy DR, Schiller JT. Induction of autoantibodies to mouse CCR5 with recombinant papillomavirus particles. Proc Natl Acad Sci U S A. 1999;96(5):2373–2378. doi: 10.1073/pnas.96.5.2373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chackerian B, Rangel M, Hunter Z, Peabody DS. Virus and virus-like particle-based immunogens for Alzheimer’s disease induce antibody responses against amyloid-beta without concomitant T cell responses. Vaccine. 2006;24(37–39):6321–6331. doi: 10.1016/j.vaccine.2006.05.059. [DOI] [PubMed] [Google Scholar]
  25. Chandramouli S, Medina-Selby A, Coit D, Schaefer M, Spencer T, Brito LA, Zhang P, Otten G, Mandl CW, Mason PW, Dormitzer PR, Settembre EC. Generation of a parvovirus B19 vaccine candidate. Vaccine. 2013;31(37):3872–3878. doi: 10.1016/j.vaccine.2013.06.062. [DOI] [PubMed] [Google Scholar]
  26. Charpilienne A, Nejmeddine M, Berois M, Parez N, Neumann E, Hewat E, Trugnan G, Cohen J. Individual rotavirus-like particles containing 120 molecules of fluorescent protein are visible in living cells. J Biol Chem. 2001;276(31):29361–29367. doi: 10.1074/jbc.M101935200. [DOI] [PubMed] [Google Scholar]
  27. Chen LS, Wang M, Ou WC, Fung CY, Chen PL, Chang CF, Huang WS, Wang JY, Lin PY, Chang D. Efficient gene transfer using the human JC virus-like particle that inhibits human colon adenocarcinoma growth in a nude mouse model. Gene Ther. 2010;17(8):1033–1041. doi: 10.1038/gt.2010.50. [DOI] [PubMed] [Google Scholar]
  28. Chen Y, Guo W, Xu Z, Yan Q, Luo Y, Shi Q, Chen D, Zhu L, Wang X. A novel recombinant pseudorabies virus expressing parvovirus VP2 gene: immunogenicity and protective efficacy in swine. Virol J. 2011;8:307. doi: 10.1186/1743-422X-8-307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chen Z, Li C, Zhu Y, Wang B, Meng C, Liu G. Immunogenicity of virus-like particles containing modified goose parvovirus VP2 protein. Virus Res. 2012;169(1):306–309. doi: 10.1016/j.virusres.2012.08.009. [DOI] [PubMed] [Google Scholar]
  30. Cheong WS, Reiseger J, Turner SJ, Boyd R, Netter HJ. Chimeric virus-like particles for the delivery of an inserted conserved influenza A-specific CTL epitope. Antiviral Res. 2009;81(2):113–122. doi: 10.1016/j.antiviral.2008.10.003. [DOI] [PubMed] [Google Scholar]
  31. Chi JN, Wu CY, Chien MS, Wu PC, Wu CM, Huang C. The preparation of porcine circovirus type 2 (PCV2) virus-like particles using a recombinant pseudorabies virus and its application to vaccine development. J Biotechnol. 2014;181:12–19. doi: 10.1016/j.jbiotec.2014.04.006. [DOI] [PubMed] [Google Scholar]
  32. Chiou SS, Crill WD, Chen LK, Chang GJ. Enzyme-linked immunosorbent assays using novel Japanese encephalitis virus antigen improve the accuracy of clinical diagnosis of flavivirus infections. Clin Vaccine Immunol. 2008;15(5):825–835. doi: 10.1128/CVI.00004-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chou MI, Hsieh YF, Wang M, Chang JT, Chang D, Zouali M, Tsay GJ. In vitro and in vivo targeted delivery of IL-10 interfering RNA by JC virus-like particles. J Biomed Sci. 2010;17:51. doi: 10.1186/1423-0127-17-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Chroboczek J, Szurgot I, Szolajska E. Virus-like particles as vaccine. Acta Biochim Pol. 2014;61(3):531–539. [PubMed] [Google Scholar]
  35. Chung YC, Huang JH, Lai CW, Sheng HC, Shih SR, Ho MS, Hu YC. Expression, purification and characterization of enterovirus-71 virus-like particles. World J Gastroenterol. 2006;12(6):921–927. doi: 10.3748/wjg.v12.i6.921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Chung YC, Ho MS, Wu JC, Chen WJ, Huang JH, Chou ST, Hu YC. Immunization with virus-like particles of enterovirus 71 elicits potent immune responses and protects mice against lethal challenge. Vaccine. 2008;26(15):1855–1862. doi: 10.1016/j.vaccine.2008.01.058. [DOI] [PubMed] [Google Scholar]
  37. Clark KB, Lin SC, Humphrey C, Foytich K, Esona M, Wang Y, Liu M, Jiang B. Expression and characterization of human group C rotavirus virus-like particles in insect cells. Virology. 2009;387(2):267–272. doi: 10.1016/j.virol.2009.02.023. [DOI] [PubMed] [Google Scholar]
  38. Cornuz J, Zwahlen S, Jungi WF, Osterwalder J, Klingler K, van Melle G, Bangala Y, Guessous I, Muller P, Willers J, Maurer P, Bachmann MF, Cerny T. A vaccine against nicotine for smoking cessation: a randomized controlled trial. PLoS One. 2008;3(6):e2547. doi: 10.1371/journal.pone.0002547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Cortes-Perez NG, Sapin C, Jaffrelo L, Daou S, Grill JP, Langella P, Seksik P, Beaugerie L, Chwetzoff S, Trugnan G. Rotavirus-like particles: a novel nanocarrier for the gut. J Biomed Biotechnol. 2010;2010:317545. doi: 10.1155/2010/317545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Cox MM. Cell-based protein vaccines for influenza. Curr Opin Mol Ther. 2005;7(1):24–29. [PubMed] [Google Scholar]
  41. 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(3):251–263. doi: 10.1097/CJI.0b013e318209ee72. [DOI] [PubMed] [Google Scholar]
  42. Deo VK, Yoshimatsu K, Otsuki T, Dong J, Kato T, Park EY. Display of Neospora caninum surface protein related sequence 2 on Rous sarcoma virus-derived gag protein virus-like particles. J Biotechnol. 2013;165(1):69–75. doi: 10.1016/j.jbiotec.2013.02.013. [DOI] [PubMed] [Google Scholar]
  43. Di Martino B, Marsilio F, Roy P. Assembly of feline calicivirus-like particle and its immunogenicity. Vet Microbiol. 2007;120(1–2):173–178. doi: 10.1016/j.vetmic.2006.10.021. [DOI] [PubMed] [Google Scholar]
  44. Ding FX, Wang F, Lu YM, Li K, Wang KH, He XW, Sun SH. Multiepitope peptide-loaded virus-like particles as a vaccine against hepatitis B virus-related hepatocellular carcinoma. Hepatology. 2009;49(5):1492–1502. doi: 10.1002/hep.22816. [DOI] [PubMed] [Google Scholar]
  45. Easterbrook JD, Schwartzman LM, Gao J, Kash JC, Morens DM, Couzens L, Wan H, Eichelberger MC, Taubenberger JK. Protection against a lethal H5N1 influenza challenge by intranasal immunization with virus-like particles containing 2009 pandemic H1N1 neuraminidase in mice. Virology. 2012;432(1):39–44. doi: 10.1016/j.virol.2012.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Edman JC, Hallewell RA, Valenzuela P, Goodman HM, Rutter WJ. Synthesis of hepatitis B surface and core antigens in E. coli. Nature. 1981;291(5815):503–506. doi: 10.1038/291503a0. [DOI] [PubMed] [Google Scholar]
  47. Esteves K. Safety of oral poliomyelitis vaccine: results of a WHO enquiry. Bull World Health Organ. 1988;66(6):739–746. [PMC free article] [PubMed] [Google Scholar]
  48. Fachinger V, Bischoff R, Jedidia SB, Saalmuller A, Elbers K. The effect of vaccination against porcine circovirus type 2 in pigs suffering from porcine respiratory disease complex. Vaccine. 2008;26(11):1488–1499. doi: 10.1016/j.vaccine.2007.11.053. [DOI] [PubMed] [Google Scholar]
  49. Fakruddin JM, Lempicki RA, Gorelick RJ, Yang J, Adelsberger JW, Garcia-Pineres AJ, Pinto LA, Lane HC, Imamichi T. Noninfectious papilloma virus-like particles inhibit HIV-1 replication: implications for immune control of HIV-1 infection by IL-27. Blood. 2007;109(5):1841–1849. doi: 10.1182/blood-2006-02-001578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fort M, Sibila M, Allepuz A, Mateu E, Roerink F, Segales J. Porcine circovirus type 2 (PCV2) vaccination of conventional pigs prevents viremia against PCV2 isolates of different genotypes and geographic origins. Vaccine. 2008;26:1063–1071. doi: 10.1016/j.vaccine.2007.12.019. [DOI] [PubMed] [Google Scholar]
  51. Freivalds J, Kotelovica S, Voronkova T, Ose V, Tars K, Kazaks A. Yeast-expressed bacteriophage-like particles for the packaging of nanomaterials. Mol Biotechnol. 2013 doi: 10.1007/s12033-013-9686-0. [DOI] [PubMed] [Google Scholar]
  52. Fulurija A, Lutz TA, Sladko K, Osto M, Wielinga PY, Bachmann MF, Saudan P. Vaccination against GIP for the treatment of obesity. PLoS One. 2008;3:e3163. doi: 10.1371/journal.pone.0003163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Garrone P, Fluckiger AC, Mangeot PE, Gauthier E, Dupeyrot-Lacas P, Mancip J, Cangialosi A, Du Chene I, LeGrand R, Mangeot I, Lavillette D, Bellier B, Cosset FL, Tangy F, Klatzmann D, Dalba C. A prime-boost strategy using virus-like particles pseudotyped for HCV proteins triggers broadly neutralizing antibodies in macaques. Sci Transl Med. 2011;3(94):94ra71. doi: 10.1126/scitranslmed.3002330. [DOI] [PubMed] [Google Scholar]
  54. Gilbert L, Toivola J, Lehtomaki E, Donaldson L, Kapyla P, Vuento M, Oker-Blom C. Assembly of fluorescent chimeric virus-like particles of canine parvovirus in insect cells. Biochem Biophys Res Commun. 2004;313(4):878–887. doi: 10.1016/j.bbrc.2003.11.176. [DOI] [PubMed] [Google Scholar]
  55. Goldmann C, Petry H, Frye S, Ast O, Ebitsch S, Jentsch KD, Kaup FJ, Weber F, Trebst C, Nisslein T, Hunsmann G, Weber T, Luke W. Molecular cloning and expression of major structural protein VP1 of the human polyomavirus JC virus: formation of virus-like particles useful for immunological and therapeutic studies. J Virol. 1999;73(5):4465–4469. doi: 10.1128/jvi.73.5.4465-4469.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Gorelik J, Shevchuk A, Ramalho M, Elliott M, Lei C, Higgins CF, Lab MJ, Klenerman D, Krauzewicz N, Korchev Y. Scanning surface confocal microscopy for simultaneous topographical and fluorescence imaging: application to single virus-like particle entry into a cell. Proc Natl Acad Sci U S A. 2002;99(25):16018–16023. doi: 10.1073/pnas.252458399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Grgacic EV, Anderson DA. Virus-like particles: passport to immune recognition. Methods. 2006;40(1):60–65. doi: 10.1016/j.ymeth.2006.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Gromadzka B, Szewczyk B, Konopa G, Fitzner A, Kesy A. Recombinant VP60 in the form of virion-like particles as a potential vaccine against rabbit hemorrhagic disease virus. Acta Biochim Pol. 2006;53(2):371–376. [PubMed] [Google Scholar]
  59. Guillen G, Aguilar JC, Dueñas S, Hermida L, Iglesias E, Penton E, Lobaina Y, Lopez M, Mussachio A, Falcon V, Alvarez L, Martinez G, Gil L, Valdes I, Izquierdo A, Lazo L, Marcos E, Guzman G, Muzio V, Herrera L. Virus-like particles as nanovaccine candidates. Adv. Nat. Sci.: Nanosci. Nanotechnol. 2013;4(1):015005. [Google Scholar]
  60. Guo HC, Sun SQ, Jin Y, Yang SL, Wei YQ, Sun DH, Yin SH, Ma JW, Liu ZX, Guo JH, Luo JX, Yin H, Liu XT, Liu DX. Foot-and-mouth disease virus-like particles produced by a SUMO fusion protein system in Escherichia coli induce potent protective immune responses in guinea pigs, swine and cattle. Vet Res. 2013;44:48. doi: 10.1186/1297-9716-44-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Habjan M, Penski N, Wagner V, Spiegel M, Overby AK, Kochs G, Huiskonen JT, Weber F. Efficient production of Rift Valley fever virus-like particles: the antiviral protein MxA can inhibit primary transcription of bunyaviruses. Virology. 2009;385(2):400–408. doi: 10.1016/j.virol.2008.12.011. [DOI] [PubMed] [Google Scholar]
  62. Hagensee ME, Yaegashi N, Galloway DA. Self-assembly of human papillomavirus type 1 capsids by expression of the L1 protein alone or by coexpression of the L1 and L2 capsid proteins. J Virol. 1993;67(1):315–322. doi: 10.1128/jvi.67.1.315-322.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Harrington PR, Yount B, Johnston RE, Davis N, Moe C, Baric RS. Systemic, mucosal, and heterotypic immune induction in mice inoculated with Venezuelan equine encephalitis replicons expressing Norwalk virus-like particles. J Virol. 2002;76(2):730–742. doi: 10.1128/JVI.76.2.730-742.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Haynes JR, Dokken L, Wiley JA, Cawthon AG, Bigger J, Harmsen AG, Richardson C. Influenza-pseudotyped Gag virus-like particle vaccines provide broad protection against highly pathogenic avian influenza challenge. Vaccine. 2009;27(4):530–541. doi: 10.1016/j.vaccine.2008.11.011. [DOI] [PubMed] [Google Scholar]
  65. Ho Y, Lin PH, Liu CY, Lee SP, Chao YC. Assembly of human severe acute respiratory syndrome coronavirus-like particles. Biochem Biophys Res Commun. 2004;318(4):833–838. doi: 10.1016/j.bbrc.2004.04.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Horaud F. Ethical and safety considerations for the use in clinical trials of new attenuated poliovirus strains. Dev Biol Stand. 1993;78:149–154. [PubMed] [Google Scholar]
  67. Huhti L, Tamminen K, Vesikari T, Blazevic V. Characterization and immunogenicity of norovirus capsid-derived virus-like particles purified by anion exchange chromatography. Arch Virol. 2013;158(5):933–942. doi: 10.1007/s00705-012-1565-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hunter Z, Smyth HD, Durfee P, Chackerian B. Induction of mucosal and systemic antibody responses against the HIV coreceptor CCR5 upon intramuscular immunization and aerosol delivery of a virus-like particle based vaccine. Vaccine. 2009;28(2):403–414. doi: 10.1016/j.vaccine.2009.10.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Huret C, Desjardins D, Miyalou M, Levacher B, Amadoudji Zin M, Bonduelle O, Combadiere B, Dalba C, Klatzmann D, Bellier B. Recombinant retrovirus-derived virus-like particle-based vaccines induce hepatitis C virus-specific cellular and neutralizing immune responses in mice. Vaccine. 2013;31(11):1540–1547. doi: 10.1016/j.vaccine.2012.05.025. [DOI] [PubMed] [Google Scholar]
  70. Ionescu RM, Przysiecki CT, Liang X, Garsky VM, Fan J, Wang B, Troutman R, Rippeon Y, Flanagan E, Shiver J, Shi L. Pharmaceutical and immunological evaluation of human papillomavirus viruslike particle as an antigen carrier. J Pharm Sci. 2006;95(1):70–79. doi: 10.1002/jps.20493. [DOI] [PubMed] [Google Scholar]
  71. Jariyapong P, Chotwiwatthanakun C, Somrit M, Jitrapakdee S, Xing L, Cheng HR, Weerachatyanukul W. Encapsulation and delivery of plasmid DNA by virus-like nanoparticles engineered from Macrobrachium rosenbergii nodavirus. Virus Res. 2013 doi: 10.1016/j.virusres.2013.10.021. [DOI] [PubMed] [Google Scholar]
  72. Jariyapong P, Xing L, van Houten NE, Li TC, Weerachatyanukul W, Hsieh B, Moscoso CG, Chen CC, Niikura M, Cheng RH. Chimeric hepatitis E virus-like particle as a carrier for oral-delivery. Vaccine. 2013;31(2):417–424. doi: 10.1016/j.vaccine.2012.10.073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Jegerlehner A, Tissot A, Lechner F, Sebbel P, Erdmann I, Kundig T, Bachi T, Storni T, Jennings G, Pumpens P, Renner WA, Bachmann MF. A molecular assembly system that renders antigens of choice highly repetitive for induction of protective B cell responses. Vaccine. 2002;20(25–26):3104–3112. doi: 10.1016/S0264-410X(02)00266-9. [DOI] [PubMed] [Google Scholar]
  74. Jennings GT, Bachmann MF. The coming of age of virus-like particle vaccines. Biol Chem. 2008;389(5):521–536. doi: 10.1515/BC.2008.064. [DOI] [PubMed] [Google Scholar]
  75. Jennings GT, Bachmann MF. Immunodrugs: therapeutic VLP-based vaccines for chronic diseases. Annu Rev Pharmacol Toxicol. 2009;49:303–326. doi: 10.1146/annurev-pharmtox-061008-103129. [DOI] [PubMed] [Google Scholar]
  76. Jeoung HY, Lee WH, Jeong W, Ko YJ, Choi CU, An DJ. Immune responses and expression of the virus-like particle antigen of the porcine encephalomyocarditis virus. Res Vet Sci. 2010;89(2):295–300. doi: 10.1016/j.rvsc.2010.03.012. [DOI] [PubMed] [Google Scholar]
  77. Jeoung HY, Lee WH, Jeong W, Shin BH, Choi HW, Lee HS, An DJ. Immunogenicity and safety of virus-like particle of the porcine encephalomyocarditis virus in pig. Virol J. 2011;8:170. doi: 10.1186/1743-422X-8-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Jouvenet N, Bieniasz PD, Simon SM. Imaging the biogenesis of individual HIV-1 virions in live cells. Nature. 2008;454(7201):236–240. doi: 10.1038/nature06998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Ju H, Wei N, Wang Q, Wang C, Jing Z, Guo L, Liu D, Gao M, Ma B, Wang J. Goose parvovirus structural proteins expressed by recombinant baculoviruses self-assemble into virus-like particles with strong immunogenicity in goose. Biochem Biophys Res Commun. 2011;409(1):131–136. doi: 10.1016/j.bbrc.2011.04.129. [DOI] [PubMed] [Google Scholar]
  80. Kalia M, Chandra V, Rahman SA, Sehgal D, Jameel S. Heparan sulfate proteoglycans are required for cellular binding of the hepatitis E virus ORF2 capsid protein and for viral infection. J Virol. 2009;83(24):12714–12724. doi: 10.1128/JVI.00717-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Kang SM, Yoo DG, Lipatov AS, Song JM, Davis CT, Quan FS, Chen LM, Donis RO, Compans RW. Induction of long-term protective immune responses by influenza H5N1 virus-like particles. PLoS One. 2009;4(3):e4667. doi: 10.1371/journal.pone.0004667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Kawano M, Matsui M, Handa H. SV40 virus-like particles as an effective delivery system and its application to a vaccine carrier. Expert Rev Vaccines. 2013;12(2):199–210. doi: 10.1586/erv.12.149. [DOI] [PubMed] [Google Scholar]
  83. Kawano M, Morikawa K, Suda T, Ohno N, Matsushita S, Akatsuka T, Handa H, Matsui M. Chimeric SV40 virus-like particles induce specific cytotoxicity and protective immunity against influenza A virus without the need of adjuvants. Virology. 2014;448:159–167. doi: 10.1016/j.virol.2013.10.010. [DOI] [PubMed] [Google Scholar]
  84. Kazaks A, Balmaks R, Voronkova T, Ose V, Pumpens P. Melanoma vaccine candidates from chimeric hepatitis B core virus-like particles carrying a tumor-associated MAGE-3 epitope. Biotechnol J. 2008;3(11):1429–1436. doi: 10.1002/biot.200800160. [DOI] [PubMed] [Google Scholar]
  85. Keller SA, Bauer M, Manolova V, Muntwiler S, Saudan P, Bachmann MF. Cutting edge: limited specialization of dendritic cell subsets for MHC class II-associated presentation of viral particles. J Immunol. 2010;184:26–29. doi: 10.4049/jimmunol.0901540. [DOI] [PubMed] [Google Scholar]
  86. Kim Y, Chang KO, Kim WY, Saif LJ. Production of hybrid double- or triple-layered virus-like particles of group A and C rotaviruses using a baculovirus expression system. Virology. 2002;302(1):1–8. doi: 10.1006/viro.2002.1610. [DOI] [PubMed] [Google Scholar]
  87. Kirnbauer R, Booy F, Cheng N, Lowy DR, Schiller JT. Papillomavirus L1 major capsid protein self-assembles into virus-like particles that are highly immunogenic. Proc Natl Acad Sci U S A. 1992;89(24):12180–12184. doi: 10.1073/pnas.89.24.12180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Konduru K, Bradfute SB, Jacques J, Manangeeswaran M, Nakamura S, Morshed S, Wood SC, Bavari S, Kaplan GG. Ebola virus glycoprotein Fc fusion protein confers protection against lethal challenge in vaccinated mice. Vaccine. 2011;29(16):2968–2977. doi: 10.1016/j.vaccine.2011.01.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Krammer F, Schinko T, Messner P, Palmberger D, Ferko B, Grabherr R. Influenza virus-like particles as an antigen-carrier platform for the ESAT-6 epitope of Mycobacterium tuberculosis. J Virol Methods. 2010;167(1):17–22. doi: 10.1016/j.jviromet.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine. 2012;31(1):58–83. doi: 10.1016/j.vaccine.2012.10.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Lai H, Chen Q. Bioprocessing of plant-derived virus-like particles of Norwalk virus capsid protein under current Good Manufacture Practice regulations. Plant Cell Rep. 2012;31(3):573–584. doi: 10.1007/s00299-011-1196-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lee DH, Park JK, Lee YN, Song JM, Kang SM, Lee JB, Park SY, Choi IS, Song CS. H9N2 avian influenza virus-like particle vaccine provides protective immunity and a strategy for the differentiation of infected from vaccinated animals. Vaccine. 2011;29(23):4003–4007. doi: 10.1016/j.vaccine.2011.03.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lee KW, Tey BT, Ho KL, Tan WS. Delivery of chimeric hepatitis B core particles into liver cells. J Appl Microbiol. 2012;112(1):119–131. doi: 10.1111/j.1365-2672.2011.05176.x. [DOI] [PubMed] [Google Scholar]
  94. Lee DH, Bae SW, Park JK, Kwon JH, Yuk SS, Song JM, Kang SM, Kwon YK, Kim HY, Song CS. Virus-like particle vaccine protects against H3N2 canine influenza virus in dog. Vaccine. 2013;31(32):3268–3273. doi: 10.1016/j.vaccine.2013.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Lee YN, Kim MC, Lee YT, Hwang HS, Cho MK, Lee JS, Ko EJ, Kwon YM, Kang SM. AS04-adjuvanted virus-like particles containing multiple M2 extracellular domains of influenza virus confer improved protection. Vaccine. 2014;32(35):4578–4585. doi: 10.1016/j.vaccine.2014.06.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Li ML, Cripe TP, Estes PA, Lyon MK, Rose RC, Garcea RL. Expression of the human papillomavirus type 11 L1 capsid protein in Escherichia coli: characterization of protein domains involved in DNA binding and capsid assembly. J Virol. 1997;71(4):2988–2995. doi: 10.1128/jvi.71.4.2988-2995.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Li F, Zhang ZP, Peng JC, Cui ZQ, Pang DW, Li K, Wei HP, Zhou YF, Wen JK, Zhang XE. Imaging viral behavior in mammalian cells with self-assembled capsid-quantum-dot hybrid particles. Small. 2009;5(6):718–726. doi: 10.1002/smll.200801303. [DOI] [PubMed] [Google Scholar]
  98. Li HY, Han JF, Qin CF, Chen R. Virus-like particles for enterovirus 71 produced from Saccharomyces cerevisiae potently elicits protective immune responses in mice. Vaccine. 2013;31(32):3281–3287. doi: 10.1016/j.vaccine.2013.05.019. [DOI] [PubMed] [Google Scholar]
  99. Li X, Xu X, Jin A, Jia Q, Zhou H, Kang S, Lou Y, Gao J, Lu J. Self-assembled HCV core virus-like particles targeted and inhibited tumor cell migration and invasion. Nanoscale Res Lett. 2013;8(1):401. doi: 10.1186/1556-276X-8-401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. 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. 2014;134(7):1683–1694. doi: 10.1002/ijc.28482. [DOI] [PubMed] [Google Scholar]
  101. Lin MC, Wang M, Fang CY, Chen PL, Shen CH, Chang D. Inhibition of BK virus replication in human kidney cells by BK virus large tumor antigen-specific shRNA delivered by JC virus-like particles. Antiviral Res. 2014;103:25–31. doi: 10.1016/j.antiviral.2013.12.013. [DOI] [PubMed] [Google Scholar]
  102. Liu WJ, Liu XS, Zhao KN, Leggatt GR, Frazer IH. Papillomavirus virus-like particles for the delivery of multiple cytotoxic T cell epitopes. Virology. 2000;273(2):374–382. doi: 10.1006/viro.2000.0435. [DOI] [PubMed] [Google Scholar]
  103. Liu L, Celma CC, Roy P. Rift Valley fever virus structural proteins: expression, characterization and assembly of recombinant proteins. Virol J. 2008;5:82. doi: 10.1186/1743-422X-5-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. 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(1):53–59. doi: 10.1007/s11262-009-0418-2. [DOI] [PubMed] [Google Scholar]
  105. Lorenzo LJ, Duenas-Carrera S, Falcon V, Acosta-Rivero N, Gonzalez E, de la Rosa MC, Menendez I, Morales J. Assembly of truncated HCV core antigen into virus-like particles in Escherichia coli. Biochem Biophys Res Commun. 2001;281(4):962–965. doi: 10.1006/bbrc.2001.4449. [DOI] [PubMed] [Google Scholar]
  106. Lua LH, Connors NK, Sainsbury F, Chuan YP, Wibowo N, Middelberg AP. Bioengineering virus-like particles as vaccines. Biotechnol Bioeng. 2014;111(3):425–440. doi: 10.1002/bit.25159. [DOI] [PubMed] [Google Scholar]
  107. Malboeuf CM, Simon DA, Lee YE, Lankes HA, Dewhurst S, Frelinger JG, Rose RC. Human papillomavirus-like particles mediate functional delivery of plasmid DNA to antigen presenting cells in vivo. Vaccine. 2007;25(17):3270–3276. doi: 10.1016/j.vaccine.2007.01.067. [DOI] [PubMed] [Google Scholar]
  108. Manayani DJ, Thomas D, Dryden KA, Reddy V, Siladi ME, Marlett JM, Rainey GJ, Pique ME, Scobie HM, Yeager M, Young JA, Manchester M, Schneemann A. A viral nanoparticle with dual function as an anthrax antitoxin and vaccine. PLoS Pathog. 2007;3(10):1422–1431. doi: 10.1371/journal.ppat.0030142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Marelli UK, Rechenmacher F, Sobahi TR, Mas-Moruno C, Kessler H. Tumor targeting via integrin ligands. Front Oncol. 2013;3:222. doi: 10.3389/fonc.2013.00222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Martelli P, Ferrari L, Morganti M, De Angelis E, Bonilauri P, Guazzetti S, Caleffi A, Borghetti P. One dose of a porcine circovirus 2 subunit vaccine induces humoral and cell-mediated immunity and protects against porcine circovirus-associated disease under field conditions. Vet Microbiol. 2011;149(3–4):339–351. doi: 10.1016/j.vetmic.2010.12.008. [DOI] [PubMed] [Google Scholar]
  111. Martin Caballero J, Garzon A, Gonzalez-Cintado L, Kowalczyk W, Jimenez Torres I, Calderita G, Rodriguez M, Gondar V, Bernal JJ, Ardavin C, Andreu D, Zurcher T, von Kobbe C. Chimeric infectious bursal disease virus-like particles as potent vaccines for eradication of established HPV-16 E7-dependent tumors. PLoS One. 2012;7(12):e52976. doi: 10.1371/journal.pone.0052976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Matic S, Rinaldi R, Masenga V, Noris E. Efficient production of chimeric human papillomavirus 16 L1 protein bearing the M2e influenza epitope in Nicotiana benthamiana plants. BMC Biotechnol. 2011;11:106. doi: 10.1186/1472-6750-11-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Mazeike E, Gedvilaite A, Blohm U. Induction of insert-specific immune response in mice by hamster polyomavirus VP1 derived virus-like particles carrying LCMV GP33 CTL epitope. Virus Res. 2012;163(1):2–10. doi: 10.1016/j.virusres.2011.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. McAleer WJ, Buynak EB, Maigetter RZ, Wampler DE, Miller WJ, Hilleman MR. Human hepatitis B vaccine from recombinant yeast. Nature. 1984;307(5947):178–180. doi: 10.1038/307178a0. [DOI] [PubMed] [Google Scholar]
  115. McGinnes LW, Pantua H, Laliberte JP, Gravel KA, Jain S, Morrison TG. Assembly and biological and immunological properties of Newcastle disease virus-like particles. J Virol. 2010;84(9):4513–4523. doi: 10.1128/JVI.01931-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. McKee SJ, Bergot AS, Leggatt GR. Recent progress in vaccination against human papillomavirus-mediated cervical cancer. Rev Med Virol. 2015;25(Suppl 1):54–71. doi: 10.1002/rmv.1824. [DOI] [PubMed] [Google Scholar]
  117. Mena JA, Ramirez OT, Palomares LA. Quantification of rotavirus-like particles by gel permeation chromatography. J Chromatogr B Analyt Technol Biomed Life Sci. 2005;824(1–2):267–276. doi: 10.1016/j.jchromb.2005.07.034. [DOI] [PubMed] [Google Scholar]
  118. Mihailova M, Boos M, Petrovskis I, Ose V, Skrastina D, Fiedler M, Sominskaya I, Ross S, Pumpens P, Roggendorf M, Viazov S. Recombinant virus-like particles as a carrier of B- and T-cell epitopes of hepatitis C virus (HCV) Vaccine. 2006;24(20):4369–4377. doi: 10.1016/j.vaccine.2006.02.051. [DOI] [PubMed] [Google Scholar]
  119. Mohana Subramanian B, Madhanmohan M, Sriraman R, Chandrasekhar Reddy RV, Yuvaraj S, Manikumar K, Rajalakshmi S, Nagendrakumar SB, Rana SK, Srinivasan VA. Development of foot-and-mouth disease virus (FMDV) serotype O virus-like-particles (VLPs) vaccine and evaluation of its potency. Antiviral Res. 2012;96(3):288–295. doi: 10.1016/j.antiviral.2012.09.019. [DOI] [PubMed] [Google Scholar]
  120. Moon KB, Lee J, Kang S, Kim M, Mason HS, Jeon JH, Kim HS. Overexpression and self-assembly of virus-like particles in Nicotiana benthamiana by a single-vector DNA replicon system. Appl Microbiol Biotechnol. 2014;98(19):8281–8290. doi: 10.1007/s00253-014-5901-6. [DOI] [PubMed] [Google Scholar]
  121. Mortola E, Roy P. Efficient assembly and release of SARS coronavirus-like particles by a heterologous expression system. FEBS Lett. 2004;576(1–2):174–178. doi: 10.1016/j.febslet.2004.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Murata K, Lechmann M, Qiao M, Gunji T, Alter HJ, Liang TJ. Immunization with hepatitis C virus-like particles protects mice from recombinant hepatitis C virus-vaccinia infection. Proc Natl Acad Sci U S A. 2003;100:6753–6758. doi: 10.1073/pnas.1131929100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Nagesha HS, Wang LF, Hyatt AD, Morrissy CJ, Lenghaus C, Westbury HA. Self-assembly, antigenicity, and immunogenicity of the rabbit haemorrhagic disease virus (Czechoslovakian strain V-351) capsid protein expressed in baculovirus. Arch Virol. 1995;140(6):1095–1108. doi: 10.1007/BF01315418. [DOI] [PubMed] [Google Scholar]
  124. Naslund J, Lagerqvist N, Habjan M, Lundkvist A, Evander M, Ahlm C, Weber F, Bucht G. Vaccination with virus-like particles protects mice from lethal infection of Rift Valley fever virus. Virology. 2009;385(2):409–415. doi: 10.1016/j.virol.2008.12.012. [DOI] [PubMed] [Google Scholar]
  125. Niikura M, Takamura S, Kim G, Kawai S, Saijo M, Morikawa S, Kurane I, Li TC, Takeda N, Yasutomi Y. Chimeric recombinant hepatitis E virus-like particles as an oral vaccine vehicle presenting foreign epitopes. Virology. 2002;293(2):273–280. doi: 10.1006/viro.2001.1240. [DOI] [PubMed] [Google Scholar]
  126. Ogasawara Y, Amexis G, Yamaguchi H, Kajigaya S, Leppla SH, Young NS. Recombinant viral-like particles of parvovirus B19 as antigen carriers of anthrax protective antigen. In Vivo. 2006;20(3):319–324. [PubMed] [Google Scholar]
  127. Ohtaki N, Takahashi H, Kaneko K, Gomi Y, Ishikawa T, Higashi Y, Kurata T, Sata T, Kojima A. Immunogenicity and efficacy of two types of West Nile virus-like particles different in size and maturation as a second-generation vaccine candidate. Vaccine. 2010;28(40):6588–6596. doi: 10.1016/j.vaccine.2010.07.055. [DOI] [PubMed] [Google Scholar]
  128. Overby AK, Popov V, Neve EP, Pettersson RF. Generation and analysis of infectious virus-like particles of uukuniemi virus (bunyaviridae): a useful system for studying bunyaviral packaging and budding. J Virol. 2006;80(21):10428–10435. doi: 10.1128/JVI.01362-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Pan Q, He K, Huang K. Development of recombinant porcine parvovirus-like particles as an antigen carrier formed by the hybrid VP2 protein carrying immunoreactive epitope of porcine circovirus type 2. Vaccine. 2008;26(17):2119–2126. doi: 10.1016/j.vaccine.2008.02.037. [DOI] [PubMed] [Google Scholar]
  130. Parez N, Fourgeux C, Mohamed A, Dubuquoy C, Pillot M, Dehee A, Charpilienne A, Poncet D, Schwartz-Cornil I, Garbarg-Chenon A. Rectal immunization with rotavirus virus-like particles induces systemic and mucosal humoral immune responses and protects mice against rotavirus infection. J Virol. 2006;80(4):1752–1761. doi: 10.1128/JVI.80.4.1752-1761.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Park JK, Lee DH, Yuk SS, To EO, Kwon JH, Noh JY, Kim BY, Choi SW, Kang SM, Lee JB, Park SY, Choi IS, Song CS. Virus-like particle vaccine confers protection against a lethal NDV challenge in chickens and allows DIVA strategy. Clin Vaccine Immunol. 2014 doi: 10.1128/CVI.00636-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Pastori C, Tudor D, Diomede L, Drillet AS, Jegerlehner A, Rohn TA, Bomsel M, Lopalco L. Virus like particle based strategy to elicit HIV-protective antibodies to the alpha-helic regions of gp41. Virology. 2012;431(1–2):1–11. doi: 10.1016/j.virol.2012.05.005. [DOI] [PubMed] [Google Scholar]
  133. Pastrana DV, Vass WC, Lowy DR, Schiller JT. NHPV16 VLP vaccine induces human antibodies that neutralize divergent variants of HPV16. Virology. 2001;279(1):361–369. doi: 10.1006/viro.2000.0702. [DOI] [PubMed] [Google Scholar]
  134. Patel KG, Swartz JR. Surface functionalization of virus-like particles by direct conjugation using azide-alkyne click chemistry. Bioconjug Chem. 2011;22(3):376–387. doi: 10.1021/bc100367u. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Patterson DP, Schwarz B, El-Boubbou K, van der Oost J, Prevelige PE, Douglas T. Virus-like particle nanoreactors: programmed encapsulation of the thermostable CelB glycosidase inside the P22 capsid. Soft Matter. 2012;8(39):10158. doi: 10.1039/c2sm26485d. [DOI] [Google Scholar]
  136. Patterson DP, Rynda-Apple A, Harmsen AL, Harmsen AG, Douglas T. Biomimetic antigenic nanoparticles elicit controlled protective immune response to influenza. ACS Nano. 2013;7(4):3036–3044. doi: 10.1021/nn4006544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. 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. 2008;380(1):252–263. doi: 10.1016/j.jmb.2008.04.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Peralta A, Molinari P, Taboga O. Chimeric recombinant rotavirus-like particles as a vehicle for the display of heterologous epitopes. Virol J. 2009;6:192. doi: 10.1186/1743-422X-6-192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Peretti S, Schiavoni I, Pugliese K, Federico M. Cell death induced by the herpes simplex virus-1 thymidine kinase delivered by human immunodeficiency virus-1-based virus-like particles. Mol Ther. 2005;12(6):1185–1196. doi: 10.1016/j.ymthe.2005.06.474. [DOI] [PubMed] [Google Scholar]
  140. Pineo CB, Hitzeroth II, Rybicki EP. Immunogenic assessment of plant-produced human papillomavirus type 16 L1/L2 chimaeras. Plant Biotechnol J. 2013;11(8):964–975. doi: 10.1111/pbi.12089. [DOI] [PubMed] [Google Scholar]
  141. Plotkin SA. Vaccines: past, present and future. Nat Med. 2005;11(4 Suppl):S5–S11. doi: 10.1038/nm1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Pokorski JK, Hovlid ML, Finn MG. Cell targeting with hybrid Qβ virus-like particles displaying epidermal growth factor. Chembiochem: Eur J Chem Biol. 2011;12(16):2441–2447. doi: 10.1002/cbic.201100469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Ponterio E, Petrizzo A, Di Bartolo I, Buonaguro FM, Buonaguro L, Ruggeri FM. Pattern of activation of human antigen presenting cells by genotype GII.4 norovirus virus-like particles. J Transl Med. 2013;11:127. doi: 10.1186/1479-5876-11-127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Pumpens P, Grens E. HBV core particles as a carrier for B cell/T cell epitopes. Intervirology. 2001;44(2–3):98–114. doi: 10.1159/000050037. [DOI] [PubMed] [Google Scholar]
  145. Pyo HM, Masic A, Woldeab N, Embury-Hyatt C, Lin L, Shin YK, Song JY, Babiuk S, Zhou Y. Pandemic H1N1 influenza virus-like particles are immunogenic and provide protective immunity to pigs. Vaccine. 2012;30(7):1297–1304. doi: 10.1016/j.vaccine.2011.12.083. [DOI] [PubMed] [Google Scholar]
  146. Quan FS, Vunnava A, Compans RW, Kang SM. Virus-like particle vaccine protects against 2009 H1N1 pandemic influenza virus in mice. PLoS One. 2010;5(2):e9161. doi: 10.1371/journal.pone.0009161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Raghunandan R. Virus-like particles: innate immune stimulators. Expert Rev Vaccines. 2011;10(4):409–411. doi: 10.1586/erv.11.37. [DOI] [PubMed] [Google Scholar]
  148. Ramqvist T, Andreasson K, Dalianis T. Vaccination, immune and gene therapy based on virus-like particles against viral infections and cancer. Expert Opin Biol Ther. 2007;7(7):997–1007. doi: 10.1517/14712598.7.7.997. [DOI] [PubMed] [Google Scholar]
  149. Rohn TA, Jennings GT, Hernandez M, Grest P, Beck M, Zou Y, Kopf M, Bachmann MF. Vaccination against IL-17 suppresses autoimmune arthritis and encephalomyelitis. Eur J Immunol. 2006;36:2857–2867. doi: 10.1002/eji.200636658. [DOI] [PubMed] [Google Scholar]
  150. Rohn TA, Ralvenius WT, Paul J, Borter P, Hernandez M, Witschi R, Grest P, Zeilhofer HU, Bachmann MF, Jennings GT. A virus-like particle-based anti-nerve growth factor vaccine reduces inflammatory hyperalgesia: potential long-term therapy for chronic pain. J Immunol. 2011;186(3):1769–1780. doi: 10.4049/jimmunol.1000030. [DOI] [PubMed] [Google Scholar]
  151. Roldao A, Mellado MC, Castilho LR, Carrondo MJ, Alves PM. Virus-like particles in vaccine development. Expert Rev Vaccines. 2010;9(10):1149–1176. doi: 10.1586/erv.10.115. [DOI] [PubMed] [Google Scholar]
  152. Roose K, Baets SD, Schepens B, Saelens X. Hepatitis B core-based virus-like particles to present heterologous epitopes. Expert Rev Vaccines. 2013;12(2):183–198. doi: 10.1586/erv.12.150. [DOI] [PubMed] [Google Scholar]
  153. Sanchez-Rodriguez SP, Munch-Anguiano L, Echeverria O, Vazquez-Nin G, Mora-Pale M, Dordick JS, Bustos-Jaimes I. Human parvovirus B19 virus-like particles: in vitro assembly and stability. Biochimie. 2012;94(3):870–878. doi: 10.1016/j.biochi.2011.12.006. [DOI] [PubMed] [Google Scholar]
  154. Sanders BP, Liu Y, Brandjes A, van Hoek V, de Los Rios Oakes I, Lewis J, Wimmer E, Custers JH, Schuitemaker H, Cello J, Edo-Matas D. Brunenders: a partially attenuated historic poliovirus type I vaccine strain. J Gen Virol. 2015 doi: 10.1099/vir.0.000197. [DOI] [PubMed] [Google Scholar]
  155. Santi L, Batchelor L, Huang Z, Hjelm B, Kilbourne J, Arntzen CJ, Chen Q, Mason HS. An efficient plant viral expression system generating orally immunogenic Norwalk virus-like particles. Vaccine. 2008;26(15):1846–1854. doi: 10.1016/j.vaccine.2008.01.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Sasnauskas K, Bulavaite A, Hale A, Jin L, Knowles WA, Gedvilaite A, Dargeviciute A, Bartkeviciute D, Zvirbliene A, Staniulis J, Brown DW, Ulrich R. Generation of recombinant virus-like particles of human and non-human polyomaviruses in yeast Saccharomyces cerevisiae. Intervirology. 2002;45(4–6):308–317. doi: 10.1159/000067922. [DOI] [PubMed] [Google Scholar]
  157. Schott JW, Galla M, Godinho T, Baum C, Schambach A. Viral and non-viral approaches for transient delivery of mRNA and proteins. Curr Gene Ther. 2011;11(5):382–398. doi: 10.2174/156652311797415872. [DOI] [PubMed] [Google Scholar]
  158. Shin YC, Folk WR. Formation of polyomavirus-like particles with different VP1 molecules that bind the urokinase plasminogen activator receptor. J Virol. 2003;77(21):11491–11498. doi: 10.1128/JVI.77.21.11491-11498.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Singh P, Destito G, Schneemann A, Manchester M. Canine parvovirus-like particles, a novel nanomaterial for tumor targeting. J Nanobiotechnol. 2006;4:2. doi: 10.1186/1477-3155-4-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Skrastina D, Petrovskis I, Petraityte R, Sominskaya I, Ose V, Lieknina I, Bogans J, Sasnauskas K, Pumpens P. Chimeric derivatives of hepatitis B virus core particles carrying major epitopes of the rubella virus E1 glycoprotein. Clin Vaccine Immunol. 2013;20(11):1719–1728. doi: 10.1128/CVI.00533-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Smith GE, Flyer DC, Raghunandan R, Liu Y, Wei Z, Wu Y, Kpamegan E, Courbron D, Fries LF, 3rd, Glenn GM. Development of influenza H7N9 virus like particle (VLP) vaccine: homologous A/Anhui/1/2013 (H7N9) protection and heterologous A/chicken/Jalisco/CPA1/2012 (H7N3) cross-protection in vaccinated mice challenged with H7N9 virus. Vaccine. 2013;31(40):4305–4313. doi: 10.1016/j.vaccine.2013.07.043. [DOI] [PubMed] [Google Scholar]
  162. Sominskaya I, Skrastina D, Dislers A, Vasiljev D, Mihailova M, Ose V, Dreilina D, Pumpens P. Construction and immunological evaluation of multivalent hepatitis B virus (HBV) core virus-like particles carrying HBV and HCV epitopes. Clin Vaccine Immunol. 2010;17(6):1027–1033. doi: 10.1128/CVI.00468-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Sonderegger I, Rohn TA, Kurrer MO, Iezzi G, Zou Y, Kastelein RA, Bachmann MF, Kopf M. Neutralization of IL-17 by active vaccination inhibits IL-23-dependent autoimmune myocarditis. Eur J Immunol. 2006;36:2849–2856. doi: 10.1002/eji.200636484. [DOI] [PubMed] [Google Scholar]
  164. Souza AC, Vasques RM, Inoue-Nagata AK, Lacorte C, Maldaner FR, Noronha EF, Nagata T. Expression and assembly of Norwalk virus-like particles in plants using a viral RNA silencing suppressor gene. Appl. Microbiol. Biotechnol. 2013;97(20):9021–9027. doi: 10.1007/s00253-013-5077-5. [DOI] [PubMed] [Google Scholar]
  165. Speiser DE, Schwarz K, Baumgaertner P, Manolova V, Devevre E, Sterry W, Walden P, Zippelius A, Conzett KB, Senti G, Voelter V, Cerottini JP, Guggisberg D, Willers J, Geldhof C, Romero P, Kundig T, Knuth A, Dummer R, Trefzer U, Bachmann MF. Memory and effector CD8 T-cell responses after nanoparticle vaccination of melanoma patients. J Immunother. 2010;33(8):848–858. doi: 10.1097/CJI.0b013e3181f1d614. [DOI] [PubMed] [Google Scholar]
  166. Spohn G, Schwarz K, Maurer P, Illges H, Rajasekaran N, Choi Y, Jennings GT, Bachmann MF. Protection against osteoporosis by active immunization with TRANCE/RANKL displayed on virus-like particles. J Immunol. 2005;175(9):6211–6218. doi: 10.4049/jimmunol.175.9.6211. [DOI] [PubMed] [Google Scholar]
  167. Spohn G, Guler R, Johansen P, Keller I, Jacobs M, Beck M, Rohner F, Bauer M, Dietmeier K, Kundig TM, Jennings GT, Brombacher F, Bachmann MF. A virus-like particle-based vaccine selectively targeting soluble TNF-alpha protects from arthritis without inducing reactivation of latent tuberculosis. J Immunol. 2007;178(11):7450–7457. doi: 10.4049/jimmunol.178.11.7450. [DOI] [PubMed] [Google Scholar]
  168. Spohn G, Keller I, Beck M, Grest P, Jennings GT, Bachmann MF. Active immunization with IL-1 displayed on virus-like particles protects from autoimmune arthritis. Eur J Immunol. 2008;38(3):877–887. doi: 10.1002/eji.200737989. [DOI] [PubMed] [Google Scholar]
  169. Spohn G, Jennings GT, Martina BE, Keller I, Beck M, Pumpens P, Osterhaus AD, Bachmann MF. A VLP-based vaccine targeting domain III of the West Nile virus E protein protects from lethal infection in mice. Virol J. 2010;7:146. doi: 10.1186/1743-422X-7-146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Stewart M, Dubois E, Sailleau C, Breard E, Viarouge C, Desprat A, Thiery R, Zientara S, Roy P. Bluetongue virus serotype 8 virus-like particles protect sheep against virulent virus infection as a single or multi-serotype cocktail immunogen. Vaccine. 2013;31(3):553–558. doi: 10.1016/j.vaccine.2012.11.016. [DOI] [PubMed] [Google Scholar]
  171. Storni T, Ruedl C, Schwarz K, Schwendener RA, Renner WA, Bachmann MF. Nonmethylated CG motifs packaged into virus-like particles induce protective cytotoxic T cell responses in the absence of systemic side effects. J Immunol. 2004;172(3):1777–1785. doi: 10.4049/jimmunol.172.3.1777. [DOI] [PubMed] [Google Scholar]
  172. 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(1):124–128. doi: 10.1016/j.bbrc.2011.02.122. [DOI] [PubMed] [Google Scholar]
  173. Swenson DL, Warfield KL, Kuehl K, Larsen T, Hevey MC, Schmaljohn A, Bavari S, Aman MJ. Generation of Marburg virus-like particles by co-expression of glycoprotein and matrix protein. FEMS Immunol Med Microbiol. 2004;40(1):27–31. doi: 10.1016/S0928-8244(03)00273-6. [DOI] [PubMed] [Google Scholar]
  174. Swenson DL, Warfield KL, Negley DL, Schmaljohn A, Aman MJ, Bavari S. Virus-like particles exhibit potential as a pan-filovirus vaccine for both Ebola and Marburg viral infections. Vaccine. 2005;23(23):3033–3042. doi: 10.1016/j.vaccine.2004.11.070. [DOI] [PubMed] [Google Scholar]
  175. Takeda S, Shiosaki K, Kaneda Y, Nakasatomi T, Yoshizaki H, Someya K, Konno Y, Eda Y, Kino Y, Yamamoto N, Honda M. Hemagglutinating virus of Japan protein is efficient for induction of CD4+ T-cell response by a hepatitis B core particle-based HIV vaccine. Clin Immunol. 2004;112(1):92–105. doi: 10.1016/j.clim.2004.04.001. [DOI] [PubMed] [Google Scholar]
  176. Tan M, Jiang X. Norovirus and its histo-blood group antigen receptors: an answer to a historical puzzle. Trends Microbiol. 2005;13(6):285–293. doi: 10.1016/j.tim.2005.04.004. [DOI] [PubMed] [Google Scholar]
  177. Tan M, Huang P, Meller J, Zhong W, Farkas T, Jiang X. Mutations within the P2 domain of norovirus capsid affect binding to human histo-blood group antigens: evidence for a binding pocket. J Virol. 2003;77(23):12562–12571. doi: 10.1128/JVI.77.23.12562-12571.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Tan M, Fang P, Chachiyo T, Xia M, Huang P, Fang Z, Jiang W, Jiang X. Noroviral P particle: structure, function and applications in virus-host interaction. Virology. 2008;382(1):115–123. doi: 10.1016/j.virol.2008.08.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Tartour E, Benchetrit F, Haicheur N, Adotevi O, Fridman WH. Synthetic and natural non-live vectors: rationale for their clinical development in cancer vaccine protocols. Vaccine. 2002;20(Suppl 4):A32–A39. doi: 10.1016/S0264-410X(02)00385-7. [DOI] [PubMed] [Google Scholar]
  180. Teunissen EA, de Raad M, Mastrobattista E. Production and biomedical applications of virus-like particles derived from polyomaviruses. J Control Release. 2013;172(1):305–321. doi: 10.1016/j.jconrel.2013.08.026. [DOI] [PubMed] [Google Scholar]
  181. Tissot AC, Maurer P, Nussberger J, Sabat R, Pfister T, Ignatenko S, Volk HD, Stocker H, Muller P, Jennings GT, Wagner F, Bachmann MF. Effect of immunisation against angiotensin II with CYT006-AngQb on ambulatory blood pressure: a double-blind, randomised, placebo-controlled phase IIa study. Lancet. 2008;371(9615):821–827. doi: 10.1016/S0140-6736(08)60381-5. [DOI] [PubMed] [Google Scholar]
  182. Tissot AC, Renhofa R, Schmitz N, Cielens I, Meijerink E, Ose V, Jennings GT, Saudan P, Pumpens P, Bachmann MF. Versatile virus-like particle carrier for epitope based vaccines. PLoS One. 2010;5(3):e9809. doi: 10.1371/journal.pone.0009809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Touze A, Gaitan J, Arnold F, Cazal R, Fleury MJ, Combelas N, Sizaret PY, Guyetant S, Maruani A, Baay M, Tognon M, Coursaget P. Generation of Merkel cell polyomavirus (MCV)-like particles and their application to detection of MCV antibodies. J Clin Microbiol. 2010;48(5):1767–1770. doi: 10.1128/JCM.01691-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Tscherne DM, Manicassamy B, Garcia-Sastre A. An enzymatic virus-like particle assay for sensitive detection of virus entry. J Virol Methods. 2010;163(2):336–343. doi: 10.1016/j.jviromet.2009.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Tyler M, Tumban E, Peabody DS, Chackerian B. The use of hybrid virus-like particles to enhance the immunogenicity of a broadly protective HPV vaccine. Biotechnol Bioeng. 2014;111(12):2398–2406. doi: 10.1002/bit.25311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Ungaro F, Conte C, Quaglia F, Tornesello ML, Buonaguro FM, Buonaguro L. VLPs and particle strategies for cancer vaccines. Expert Rev Vaccines. 2013;12(10):1173–1193. doi: 10.1586/14760584.2013.836909. [DOI] [PubMed] [Google Scholar]
  187. Vacher G, Kaeser MD, Moser C, Gurny R, Borchard G. Recent advances in mucosal immunization using virus-like particles. Mol Pharm. 2013;10(5):1596–1609. doi: 10.1021/mp300597g. [DOI] [PubMed] [Google Scholar]
  188. Valenzuela P, Medina A, Rutter WJ, Ammerer G, Hall BD. Synthesis and assembly of hepatitis B virus surface antigen particles in yeast. Nature. 1982;298(5872):347–350. doi: 10.1038/298347a0. [DOI] [PubMed] [Google Scholar]
  189. Vidyasagar P, Sridevi VN, Rajan S, Praveen A, Srikanth A, Abhinay G, Siva Kumar V, Verma RR, Rajendra L. Generation and characterization of neutralizing monoclonal antibodies against baculo-expressed HPV 16 VLPs. Eur J Microbiol Immunol (Bp) 2014;4(1):56–64. doi: 10.1556/EuJMI.4.2014.1.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Vieira HL, Estevao C, Roldao A, Peixoto CC, Sousa MF, Cruz PE, Carrondo MJ, Alves PM. Triple layered rotavirus VLP production: kinetics of vector replication, mRNA stability and recombinant protein production. J Biotechnol. 2005;120(1):72–82. doi: 10.1016/j.jbiotec.2005.03.026. [DOI] [PubMed] [Google Scholar]
  191. Voronkova T, Kazaks A, Ose V, Ozel M, Scherneck S, Pumpens P, Ulrich R. Hamster polyomavirus-derived virus-like particles are able to transfer in vitro encapsidated plasmid DNA to mammalian cells. Virus Genes. 2007;34(3):303–314. doi: 10.1007/s11262-006-0028-1. [DOI] [PubMed] [Google Scholar]
  192. Wang T, Zhang Z, Gao D, Li F, Wei H, Liang X, Cui Z, Zhang XE. Encapsulation of gold nanoparticles by simian virus 40 capsids. Nanoscale. 2011;3(10):4275–4282. doi: 10.1039/c1nr10568j. [DOI] [PubMed] [Google Scholar]
  193. Wang HB, Fang G, Yu WZ, Du F, Fan CX, Liu QL, Hao LX, Liu Y, Zheng JS, Qin ZY, Xia W, Zhang SY, Yin ZD, Jing Q, Zhang YX, Huang RN, Yang RP, Tong WB, Qi Q, Guan XJ, Jing YL, Ma QL, Wang J, Ma XZ, Chen N, Zheng HR, Li YQ, Ma C, Su QR, Reilly KH, Luo HM, Wu XP, Wen N, Yang WZ. An outbreak of type Pi vaccine-derived poliovirus in Sichuan Province, China: emergence and circulation in an under-immunized population. PLoS One. 2014;9(12):e113880. doi: 10.1371/journal.pone.0113880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Warfield KL, Bosio CM, Welcher BC, Deal EM, Mohamadzadeh M, Schmaljohn A, Aman MJ, Bavari S. Ebola virus-like particles protect from lethal Ebola virus infection. Proc Natl Acad Sci U S A. 2003;100(26):15889–15894. doi: 10.1073/pnas.2237038100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  195. Warfield KL, Swenson DL, Demmin G, Bavari S. Filovirus-like particles as vaccines and discovery tools. Expert Rev Vaccines. 2005;4(3):429–440. doi: 10.1586/14760584.4.3.429. [DOI] [PubMed] [Google Scholar]
  196. Warfield KL, Posten NA, Swenson DL, Olinger GG, Esposito D, Gillette WK, Hopkins RF, Costantino J, Panchal RG, Hartley JL, Aman MJ, Bavari S. Filovirus-like particles produced in insect cells: immunogenicity and protection in rodents. J Infect Dis. 2007;196(Suppl 2):S421–S429. doi: 10.1086/520612. [DOI] [PubMed] [Google Scholar]
  197. Warfield KL, Swenson DL, Olinger GG, Kalina WV, Aman MJ, Bavari S. Ebola virus-like particle-based vaccine protects nonhuman primates against lethal Ebola virus challenge. J Infect Dis. 2007;196(Suppl 2):S430–S437. doi: 10.1086/520583. [DOI] [PubMed] [Google Scholar]
  198. Wei B, Wei Y, Zhang K, Wang J, Xu R, Zhan S, Lin G, Wang W, Liu M, Wang L, Zhang R, Li J. Development of an antisense RNA delivery system using conjugates of the MS2 bacteriophage capsids and HIV-1 TAT cell-penetrating peptide. Biomed Pharmacother. 2009;63(4):313–318. doi: 10.1016/j.biopha.2008.07.086. [DOI] [PubMed] [Google Scholar]
  199. Wei HJ, Chang W, Lin SC, Liu WC, Chang DK, Chong P, Wu SC. Fabrication of influenza virus-like particles using M2 fusion proteins for imaging single viruses and designing vaccines. Vaccine. 2011;29(41):7163–7172. doi: 10.1016/j.vaccine.2011.05.077. [DOI] [PubMed] [Google Scholar]
  200. Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, Jennings GT, Luond R, Ortmann R, Reichwald J, Zurini M, Mir A, Bachmann MF, Staufenbiel M. The second-generation active Abeta immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci. 2011;31(25):9323–9331. doi: 10.1523/JNEUROSCI.0293-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Wu M, Sherwin T, Brown WL, Stockley PG. Delivery of antisense oligonucleotides to leukemia cells by RNA bacteriophage capsids. Nanomedicine. 2005;1(1):67–76. doi: 10.1016/j.nano.2004.11.011. [DOI] [PubMed] [Google Scholar]
  202. Wu CY, Yeh YC, Yang YC, Chou C, Liu MT, Wu HS, Chan JT, Hsiao PW. Mammalian expression of virus-like particles for advanced mimicry of authentic influenza virus. PLoS One. 2010;5(3):e9784. doi: 10.1371/journal.pone.0009784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  203. Wu PC, Lin WL, Wu CM, Chi JN, Chien MS, Huang C. Characterization of porcine circovirus type 2 capsid particle assembly and its application to virus-like particle vaccine development. Appl Microbiol Biotechnol. 2012;95(6):1501–1507. doi: 10.1007/s00253-012-4015-2. [DOI] [PubMed] [Google Scholar]
  204. Xu J, Guo HC, Wei YQ, Dong H, Han SC, Ao D, Sun DH, Wang HM, Cao SZ, Sun SQ. Self-assembly of virus-like particles of canine parvovirus capsid protein expressed from Escherichia coli and application as virus-like particle vaccine. Appl Microbiol Biotechnol. 2014 doi: 10.1007/s00253-013-5485-6. [DOI] [PubMed] [Google Scholar]
  205. Yamaji H, Konishi E. Production of Japanese encephalitis virus-like particles in insect cells. Bioengineered. 2013;4(6):438–442. doi: 10.4161/bioe.24514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Yang YK, Burkhard P. Encapsulation of gold nanoparticles into self-assembling protein nanoparticles. J. Nanobiotechnol. 2012;10:42. doi: 10.1186/1477-3155-10-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Yao Q, Zhang R, Guo L, Li M, Chen C. Th cell-independent immune responses to chimeric hemagglutinin/simian human immunodeficiency virus-like particles vaccine. J Immunol. 2004;173(3):1951–1958. doi: 10.4049/jimmunol.173.3.1951. [DOI] [PubMed] [Google Scholar]
  208. Ye X, Ku Z, Liu Q, Wang X, Shi J, Zhang Y, Kong L, Cong Y, Huang Z. Chimeric virus-like particle vaccines displaying conserved enterovirus 71 epitopes elicit protective neutralizing antibodies in mice through divergent mechanisms. J Virol. 2014;88(1):72–81. doi: 10.1128/JVI.01848-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Yin S, Sun S, Yang S, Shang Y, Cai X, Liu X. Self-assembly of virus-like particles of porcine circovirus type 2 capsid protein expressed from Escherichia coli. Virol J. 2010;7:166. doi: 10.1186/1743-422X-7-166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Young SL, Wilson M, Wilson S, Beagley KW, Ward V, Baird MA. Transcutaneous vaccination with virus-like particles. Vaccine. 2006;24(26):5406–5412. doi: 10.1016/j.vaccine.2006.03.052. [DOI] [PubMed] [Google Scholar]
  211. Zamarin D, Postow MA. Immune checkpoint modulation: rational design of combination strategies. Pharmacol Ther. 2015;150:23–32. doi: 10.1016/j.pharmthera.2015.01.003. [DOI] [PubMed] [Google Scholar]
  212. Zamora E, Handisurya A, Shafti-Keramat S, Borchelt D, Rudow G, Conant K, Cox C, Troncoso JC, Kirnbauer R. Papillomavirus-like particles are an effective platform for amyloid-beta immunization in rabbits and transgenic mice. J Immunol. 2006;177(4):2662–2670. doi: 10.4049/jimmunol.177.4.2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Zhang R, Zhang S, Li M, Chen C, Yao Q. Incorporation of CD40 ligand into SHIV virus-like particles (VLP) enhances SHIV-VLP-induced dendritic cell activation and boosts immune responses against HIV. Vaccine. 2010;28(31):5114–5127. doi: 10.1016/j.vaccine.2010.03.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Zhang S, Liang M, Gu W, Li C, Miao F, Wang X, Jin C, Zhang L, Zhang F, Zhang Q, Jiang L, Li M, Li D. Vaccination with dengue virus-like particles induces humoral and cellular immune responses in mice. Virol J. 2011;8:333. doi: 10.1186/1743-422X-8-333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Zhao Q, Chen W, Chen Y, Zhang L, Zhang J, Zhang Z. Self-assembled virus-like particles from rotavirus structural protein VP6 for targeted drug delivery. Bioconjug Chem. 2011;22(3):346–352. doi: 10.1021/bc1002532. [DOI] [PubMed] [Google Scholar]
  216. Zielonka A, Gedvilaite A, Ulrich R, Luschow D, Sasnauskas K, Muller 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(1–2):128–137. doi: 10.1016/j.virusres.2006.02.010. [DOI] [PubMed] [Google Scholar]
  217. Zochowska M, Paca A, Schoehn G, Andrieu JP, Chroboczek J, Dublet B, Szolajska E. Adenovirus dodecahedron, as a drug delivery vector. PLoS One. 2009;4(5):e5569. doi: 10.1371/journal.pone.0005569. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Applied Microbiology and Biotechnology are provided here courtesy of Nature Publishing Group

RESOURCES