ArticlePDF AvailableLiterature Review

Gold nanoparticles for preparation of antibodies and vaccines against infectious diseases

Taylor & Francis
Expert Review of Vaccines
Authors:

Abstract and Figures

Introduction: Vaccination remains very effective in stimulating protective immune responses against infections. An important task in antibody and vaccine preparation is to choose an optimal carrier that will ensure a high immune response. Particularly promising in this regard are nanoscale particle carriers. An antigen that is adsorbed or encapsulated by nanoparticles can be used as an adjuvant to optimize the immune response during vaccination. A very popular antigen carrier used for immunization and vaccination is gold nanoparticles, with are being used to make new vaccines against viral, bacterial, and parasitic infections. Areas covered: This review summarizes what is currently known about the use of gold nanoparticles as an antigen carrier and adjuvant to prepare antibodies in vivo and design vaccines against viral, bacterial, and parasitic infections. The basic principles, recent advances, and current problems in the use of gold nanoparticles are discussed. Expert opinion: Gold nanoparticles can be used as adjuvants to increase the effectiveness of vaccines by stimulating antigen-presenting cells and ensuring controlled antigen release. Studying the characteristics of the immune response obtained from the use of gold nanoparticles as a carrier and an adjuvant will permit the particles’ potential for vaccine design to be increased.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ierv20
Expert Review of Vaccines
ISSN: 1476-0584 (Print) 1744-8395 (Online) Journal homepage: https://www.tandfonline.com/loi/ierv20
Gold nanoparticles for preparation of antibodies
and vaccines against infectious diseases
Lev A. Dykman
To cite this article: Lev A. Dykman (2020): Gold nanoparticles for preparation of
antibodies and vaccines against infectious diseases, Expert Review of Vaccines, DOI:
10.1080/14760584.2020.1758070
To link to this article: https://doi.org/10.1080/14760584.2020.1758070
Accepted author version posted online: 19
Apr 2020.
Published online: 28 Apr 2020.
Submit your article to this journal
Article views: 160
View related articles
View Crossmark data
REVIEW
Gold nanoparticles for preparation of antibodies and vaccines against infectious
diseases
Lev A. Dykman
Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, Russia
ABSTRACT
Introduction: Vaccination remains very effective in stimulating protective immune responses against
infections. An important task in antibody and vaccine preparation is to choose an optimal carrier that
will ensure a high immune response. Particularly promising in this regard are nanoscale particle carriers.
An antigen that is adsorbed or encapsulated by nanoparticles can be used as an adjuvant to optimize
the immune response during vaccination. a very popular antigen carrier used for immunization and
vaccination is gold nanoparticles, with are being used to make new vaccines against viral, bacterial, and
parasitic infections.
Areas covered: This review summarizes what is currently known about the use of gold nanoparticles as
an antigen carrier and adjuvant to prepare antibodies in vivo and design vaccines against viral, bacterial,
and parasitic infections. The basic principles, recent advances, and current problems in the use of gold
nanoparticles are discussed.
Expert opinion: Gold nanoparticles can be used as adjuvants to increase the effectiveness of vaccines
by stimulating antigen-presenting cells and ensuring controlled antigen release. Studying the charac-
teristics of the immune response obtained from the use of gold nanoparticles as a carrier and an
adjuvant will permit the particlespotential for vaccine design to be increased.
ARTICLE HISTORY
Received 22 January 2020
Accepted 16 April 2020
KEYWORDS
Vaccine; gold nanoparticles;
nanocarriers; adjuvant;
immunization; antibodies;
infections
1. Introduction
Vaccination remains very effective in stimulating protective
immune responses against infections. Among the most
advanced diagnostic tools is the immunochemical method,
based on the use of highly pathogen-specific antibodies. An
important task in antibody and vaccine preparation is to
choose an optimal carrier (delivery system) that would ensure
a high immune response. Such adjuvant carriers can deposit
antigens at the injection sites, enhance their presentation to
immunocompetent cells, and induce the production of neces-
sary cytokines. Particularly promising in this regard are various
nanoscale particle carriers [116]. Such nanocarriers can be
used as adjuvants in the preparation of antibodies and next-
generation vaccines. The term nanovaccinology[17] is com-
ing into common scientific use.
Of note, the stimulation of antibody production in response
to antigens adsorbed on colloidal particles was discovered as
early as the beginning of the twentieth century [1820]. In the
same period, studies began on the effect of colloidal metal
particles on the cells of the immune system [21,22]. In recent
decades, significant progress has been made in understanding
the immunogenicity of nanoparticles, the response of the
immune cells, the nature of immunomodulation and immuno-
suppression, and the cyto- and immunotoxicity of nanoparti-
cles, with account taken of their physicalchemical properties
[2338].
Cells of the immune system constitute the first barrier to
the entry of nanoparticles into animal tissues and cells. In this
context, of undoubted interest is the study of the interaction
of nanoparticles with phagocytic cells, the mechanisms of
intracellular penetration, and the response of immune cells
to nanoparticles [39]. Depending on their size, nanoparticles
penetrate macrophages through receptor-mediated endocy-
tosis (phagocytosis) or pinocytosis and are localized mainly in
lysosomes and in perinuclear space. Often, they are wrapped
inside fringed vesicles. Some researchers believe that a key
part in nanoparticle uptake by macrophages is played by
scavenger receptors [40,41]. Nanoparticle penetration into
macrophages may enhance the respiratory activity of the
cells and, in some cases, the release of inflammatory mediators
(cytokines, prostaglandins, etc.) and the activation of the
immune response genes [42,43].
Of great interest are studies on the penetration of nano-
particles not only into macrophages but also into other
immune system cells, in particular dendritic cells. Dendritic
cells treated with gold nanoparticles can affect the activation
of CD8
+
T cells, which leads to epitope-specific cytotoxic
T-lymphocyte responses in vitro [44]. Compared with the use
of the native antigen, the internalization of antigen-
conjugated nanoparticles into dendritic cells increases the
immune response, enhancing lymphocyte proliferation [45].
In addition, nanoparticles can activate other immune system
CONTACT Lev A. Dykman dykman_l@ibppm.ru Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences,
Saratov, Russia
EXPERT REVIEW OF VACCINES
https://doi.org/10.1080/14760584.2020.1758070
© 2020 Informa UK Limited, trading as Taylor & Francis Group
cells neutrophils [46,47], lymphocytes [48,49], and mono-
cytes [50,51].
Thus, nanoparticle penetration into the immune cells,
which causes cytokine induction, stimulation of T cells, activa-
tion of the immune response genes, enhanced antigen pro-
cessing, and antibody secretion by B cells, offers a good
possibility of using nanoparticles as carriers and adjuvants in
the preparation of antibodies and vaccines against infections
[5255]. In particular, various nanoparticles are being used to
make new vaccines against viral [5660], bacterial [61], and
parasitic [62] infections. On the basis of nanocarriers, subunit
[63] and peptide [64] vaccines are being developed for the
oral [65], intranasal [66], and percutaneous [67] routes of
administration.
A very popular antigen carrier used for immunization and
vaccination is gold nanoparticles (GNPs) [6871]. Owing to
their unique physicochemical properties, ease of preparation,
and low toxicity, GNPs are widely used in various fields of
biomedical research [72]. For the first time, GNPs were used
as carriers of haptens for antibody preparation in 1986 [73].
Since then, a large number of reports have been published in
which this method was used and improved to develop anti-
bodies to a number of haptens and complete antigens [70]. It
was found that adjuvant properties are inherent in GNPs
themselves [7476].
This review summarizes what is now known about the use
of GNPs to prepare antibodies and vaccines against viral,
bacterial, and parasitic infections.
2. Gold nanoparticles in the design of antiviral
vaccines
GNPs were first used to develop vaccines by Demenev et al. in
1996 [77], who examined the protective properties of an
experimental tick-borne encephalitis vaccine. For tick-borne
encephalitis virus and other flaviviruses, a slowly sedimenting
virus-specific antigen was described that had complement-
binding activity, was not associated with the virion fraction,
and did not have infectivity or hemagglutinating activity. This
antigen was called a soluble antigen [78]. The vaccine was
prepared by conjugating GNPs (average diameter, 15 nm) to
the soluble antigen. White laboratory mice were vaccinated
intraperitoneally three times, each at 32 μg of antigen per
injection. The protective properties of the experimental and
commercial vaccines were compared on the basis of the aver-
age survival time and the protection coefficients after vaccina-
tion of mice challenged with a virus-containing suspension
(100,000 and 10,000 LD
50
). The survival time and the thera-
peutic and protective efficacy obtained when the experimen-
tal vaccine was used were about 1.5 times greater than when
the commercial vaccine was used.
Many studies on GNP immunization and vaccination have
used such important objects as influenza virus and foot and
mouth disease (FMD) virus. Highly specific antibodies were
obtained from the immunization of animals with 17-nm
GNPs coupled to the pFMDV and pH5N1 antigens of these
viruses [79]. BALB/C white mice were vaccinated intraperito-
neally. Both antisera obtained had a high titer. The same
research team evaluated the effect of GNP size on the immune
response to the synthetic FMD peptide pFMDV [80]. GNPs with
diameters of 2, 5, 8, 12, 17, 37, and 50 nm were used. BALB/C
mice were immunized intraperitoneally six times, each at 1 μg
of antigen per injection. The antigens were mixed with com-
plete Freunds adjuvant (CFA). The antibody titer was maximal
for 8-, 12- and 17-nm GNPs. The titer of the antisera obtained
by using GNPs was threefold higher than when keyhole limpet
hemocyanin (KLH) was used as an adjuvant and did not
require further purification from contaminant antibodies
(Figure 1). The amount of GNPs that had accumulated in the
spleen correlated with the magnitude of the immune
response elicited by the GNP/pFMDV conjugate.
Double intraperitoneal immunization of rats with 15-nm
GNPs complexed with the M1 antigen of the influenza virus
of the attenuated strain PR8 at a dose of 20 mg/kg produced
high-titer antibodies [81]. The production of IFN-γand inter-
leukins (ILs) 1βand 6 was stimulated, and the respiration of
peritoneal macrophages and spleen lymphocytes, was acti-
vated. Similar results were produced by double subcutaneous
immunization of guinea pigs with 12 μg of 15-nm GNPs con-
jugated to a synthetic VP
1
peptide of the FMD virus capsid
[82]. The titer of the resultant antibodies was not lower than
that obtained with CFA.
When guinea pigs were injected with FMD viruslike parti-
cles complexed with gold nanostars (50 μg, single injection),
the titer of specific and neutralizing antibodies and the stimu-
lation of T lymphocyte proliferation were much greater than
Article Highlights
Gold nanoparticles have been used to prepare antibodies and vac-
cines against more than 45 pathogens of viral, bacterial, and parasitic
infections.
The antibody elaboration during immunization with antigens conju-
gated to gold nanoparticles enhances the secretion of cytokines.
Vaccination of animals with antigens conjugated to gold nanoparti-
cles decreases the symptoms of infections and protects the animals
challenged with virulent pathogens (up to lethal doses).
By programmingthe activation state of dendritic or other antigen-
presenting cells, nanoparticles directly affect the induction of cellular
and humoral immunity.
Studying the characteristics of the immune response obtained from
the use of gold nanoparticles as a carrier and an adjuvant will permit
the particlespotential for vaccine design to be increased.
Figure 1. Efficacy of using keyhole limpet hemocyanin (KLH) and GNPs as
nanocarriers. (a) Antibodies obtained by using KLH recognize the epitopes of
both the antigen and the carrier protein. (b) Antibodies obtained by using GNPs
recognize only the antigens epitopes [80].
2L. A. DYKMAN
those in guinea pigs immunized with the antigen in the pre-
sence of mineral oil ISA206 as an adjuvant. The proposed
nanovaccine more effectively protected virus-infected animals
[83].
A prototype of the intranasal influenza a vaccine was first
proposed by Gills group [84]. The synthetic peptide M2e,
a conserved extracellular domain of influenza virus matrix
protein 2 conjugated to 12-nm GNPs, was used as an immu-
nogen (Figure 2). As an adjuvant, soluble CpG (cytosinephos-
phateguanosine) oligodeoxynucleotide (CpG ODN) was used.
BALB/C mice were immunized twice at an interval of 21 days,
each time with 8.2 μg of M2e per animal. The conjugate
induced the production of specific IgG (with stimulation of
subtypes of both IgG1 and IgG2a), which could recognize M2e
and native M2 on influenza a viruses and had a protective
effect against a lethal dose of PR8-H1N1 virus. Subsequently,
Gills group published more detailed results from the use of
the proposed prototype vaccine to induce protective immu-
nity against the influenza a virus serotypes H1N1, H3N2, and
H5N1 [85,86]. Bimler et al. [87] showed that even at 15 months
after being vaccinated with the GNP/M2e+CpG conjugate, the
mice retained high titers of M2e-specific antibodies. These
mice were protected against lethal H1N1 influenza. In addi-
tion, the antibody titers increased after a challenge with influ-
enza a and remained elevated for 3 months. This suggests that
old mice retain effective M2e-specific memory B cells.
When mice were immunized with the M2e antigen of influenza
a virus, the most effective complex immunogens were M2e +
GNPs and CpG + GNPs [88]. When they were used, the titer of
the obtained antibodies was the highest. In addition, the immu-
nogens activated the respiratory activity of lymphoid cells and the
production of proinflammatory cytokines to a greater extent than
did other immunogens, including a commercial vaccine. The adju-
vant effect of GNPs is probably associated with the more efficient
penetration of the conjugate into phagocytic cells, which leads to
improved presentation of the antigen to antibody-forming cells.
In general, GNPs as an antigen carrier and adjuvant are
often combined with other immune response stimulants,
most commonly with CFA and CpG ODNs. It is known that
when CpG motifs are unmethylated, they act as immunosti-
mulants [89]. The immunostimulatory activity of synthetic
ODNs containing CpG sequences may be similar to the activity
of bacterial DNA [90]. According to Wei et al. [91], GNPs
coupled to CpG ODNs effectively enhance the intracellular
penetration of nanoparticles into macrophages and signifi-
cantly increase the secretion of proinflammatory cytokines
tumor necrosis factor α(TNF-α) and IL-6. Moreover, the
immunostimulatory effect of GNPs conjugated to CpG ODNs
is often significantly higher than that of the same concentra-
tions of soluble CpG ODNs [92,93]. Of note, the optimal immu-
nization method is the simultaneous use of two conjugates:
GNPsantigen and CpG ODNsGNPs [94](Figure 3). The
immune response to an intranasal influenza vaccination can
be enhanced with gold nanorods coupled to polyinosinic-
polycytidylic acid as an adjuvant, as well as with CpG [95].
Wang et al. [96,97] proposed the use of two H3N2 influenza
aantigens for vaccination hemagglutinin and flagellin in
complex with 18-nm GNPs. In a single intranasal immuniza-
tion, both GNP/hemagglutinin/flagellin and GNP/hemaggluti-
nin complexes induced higher levels of antibody production
in mice than did a mixture of soluble hemagglutinin and
flagellin. Immunization elicited a strong mucosal and systemic
immune response, which protected the hosts from lethal
influenza. In addition, it was shown in vitro that the conjugates
promote the maturation of dendritic cells for the processing
and presentation of antigens, stimulate the production of
cytokines, and promote antigen-specific T-cell immunity.
GNPs were used to prepare antibodies to the surface anti-
gens of porcine transmissible gastroenteritis virus [98,99]. Mice
were immunized once intraperitoneally with 70 μg of antigen
15-nm GNPs; guinea pigs, twice subcutaneously with 125 μg;
and rabbits, three times subcutaneously with 220 μg. The
resulting virus-neutralizing antibodies from all animal species
Figure 2. Scheme of vaccine design. M2e is conjugated to GNPs. By keeping M2e in excess in the solution, complete surface-coverage of GNPs with M2e is ensured
at all times. Soluble CpG is added to the conjugate [86].
Figure 3. Average antibody titers obtained with various immunostimulant
combinations [94].
EXPERT REVIEW OF VACCINES 3
had a much higher titer than did antibodies developed against
the native virus. Immunization with the GNP/antigen complex
significantly enhanced the respiratory activity of peritoneal
macrophages and spleen lymphocytes and increased the con-
tent of IFN-γ, IL-1β, and IL-6 in the blood plasma of the
immunized animals. In addition, there was an increase in the
number of follicles in the spleen, indicating the activation of
humoral immunity.
A conjugate of 15-nm GNPs with a glycoprotein isolated
from fixed rabies virus, strain Moscow 3253, was used to
develop highly specific antibodies to the virus [100]. In the
first immunization, 25 μg of the antigen was intraperitoneally
injected into mice; in the subsequent four immunizations,
50 μg was used. The immunogenicity of the purified glyco-
protein was higher than that of whole rabies virus, and the
glycoprotein conjugated to GNPs was best able to induce
virus-neutralizing antibodies. The resulting antibodies were
used to make a diagnostic agent based on solid-phase immu-
noassay. Antirabies virus antibodies were also prepared by
using the GNP-conjugated ribonucleoprotein of an attenuated
rabies virus [101].
To prepare antibodies against West Nile fever virus, Niikura
et al. [102] used antigen conjugates with GNPs of various sizes
and shapes: 20- and 40-nm nanospheres, 40 × 20-nm nanorods,
and 40 × 40 × 40-nm nanocubes. Mice were immunized twice
intraperitoneally with 100 ng of the conjugate per animal.
Forty-nm nanospheres induced the highest concentration of
specific antibodies, while the titer of the antibodies obtained
with the other GNPs was nearly 2 times lower. The macro-
phages and dendritic cells absorbed larger numbers of nanor-
ods, which suggests that the production of antibodies does not
depend on the efficiency of absorption of various GNPs.
Nanorods enhanced the production of IL-1βand IL-18, while
nanospheres and nanocubes enhanced that of TNF-α,IL-6,IL-
12, and granulocytemacrophage colony-stimulating factor.
Stone et al. [103] examined the immune response to nanor-
ods coupled to the glycoprotein antigen of respiratory syncy-
tial virus. The conjugate-treated human dendritic cells induced
an immune response (proliferation) of primary T cells.
The influence of the particle shape and size on the effec-
tiveness of the immune response was also investigated in [94].
Mice were injected with an antigen coupled to gold nano-
spheres (diameters, 15 and 50 nm), nanorods (70 × 15 nm),
nanoshells (SiO
2
core, 140 nm; gold shell, 27 nm), and nanos-
tars (core, 12 nm; rays, 1525 nm). The antibody titers
increased from 1:160 to 1:10240 in the sequence native anti-
gen < nanorods < nanoshells < GNPs-15 nm < GNPs-50 nm. It
was concluded that spherical GNPs with diameters of 50 and
15 nm are the optimal antigen carrier and adjuvant for
immunization.
The V3βpeptide of the HIV-1 gp120 protein in complex with
2-nm gold glyconanoparticles was used as an immunogen to
prepareaprototypeHIVvaccine[104]. Rabbits were immunized
three times intramuscularly with 50 μgofthecomplex.The
resulting antibodies had a high titer and neutralizing activity
against HIV-1. The HIV-1 Gag p17 peptide conjugated to 2-nm
gold glyconanoparticles increased the proliferation of HIV-
specific CD4
+
and CD8
+
T cells and induced the secretion of
the highly functional TNF-αand IL-1βcytokines, as compared
to the unconjugated peptide [105]. Also, GNPs conjugated with
the gp120 and gp41 viral proteins were used as a platform for
thedeliveryofimmunogensinthepreparationofanHIV-1
vaccine [106].
Ten-nm GNPs were complexed simultaneously with CpG
ODN and with the recombinant hepatitis B virus core antigen
in the form of viruslike particles [107]. Mice were immunized
four times intraperitoneally with 50 μg of the conjugate. The
titer of the resultant antibodies was twofold higher than that
of the antibodies prepared without GNPs. The proliferation of
the CD4
+
and CD8
+
T cells, the secretion of IL-4 and IFN-γ, and
the stimulation of both Th1 and Th2 immune responses were
increased. That is, the conjugates showed a strong cellular and
humoral immunostimulatory ability.
The surface antigen of hepatitis B virus (HBsAg) was con-
jugated to gold nanocages [108]. The immunogenic properties
of the conjugate were examined in vitro in RAW 264.7 macro-
phages. The conjugate was intensely absorbed by the macro-
phages and contributed to the processing of the antigens and
to the secretion of IL-4.
For a prototype hepatitis E vaccine, vaccine monomers
were conjugated to gold fluorescent nanoclusters [109]. The
intraperitoneally injected conjugate not only enhanced the
Th1/Th2 immune response in mice but also reduced the toxi-
city of the vaccine. In addition, the own fluorescence of the
gold clusters made it possible to track the dynamic behavior
of the vaccine in vivo. GNPs were also used to prepare anti-
peptide antibodies to the binding sites of the E2 protein of
hepatitis C virus [110].
A prototype dengue virus vaccine was developed by using
gold nanorods functionalized with protein E virus (DENVE)
[111]. Immunization of mice with GNP/DENVE significant
increased IgG synthesis and splenocyte proliferation.
Quach et al. [112] conjugated differently sized GNPs to
domain III of envelope glycoprotein derived from serotype 2
of dengue virus (EDIII) as a dengue subunit vaccine. They used
GNPs with average particle diameters of 20, 40, and 80 nm.
The GNP/EDIII conjugate was used to immunize BALB/C mice
three times subcutaneously. The conjugate induced high con-
centrations of antibodies that provide serotype-specific neu-
tralization of dengue virus. The concentration of the
antibodies depended on both size and concentration of the
GNPs, and this made it possible to modulate the immune
response by adjusting these parameters. In addition, the con-
jugate activated the complement system, induced proliferative
responses in splenocytes and CD4
+
and CD8
+
T cells, and
induced the secretion of IL-4 and IFN-γ. Eighty-nm GNPs per-
formed the best.
Using the surface spike glycoprotein of avian coronavirus,
which causes infectious bronchitis in birds, Chen et al. [113]
obtained viruslike particles by incubating 100-nm GNPs in
a solution containing an optimized concentration of the viral
proteins. After free proteins were removed, the particles with
the antigen were shown by TEM to be morphologically similar
to the natural viral particles. BALB/C mice and specific-
pathogen-free chickens were vaccinated intramuscularly with
a single dose of 10 μg. Compared to free protein
4L. A. DYKMAN
immunization, conjugate vaccination showed improved anti-
gen delivery to lymphoid organs, higher antibody titers, an
increased response of spleen T cells, and a reduction in symp-
toms of coronavirus infection. a comparison with
a commercial whole inactivated viral vaccine also showed
that the GNP conjugate provided better antiviral protection.
GNPs conjugated to the S protein of severe acute respiratory
syndrome-related coronavirus induced a strong IgG response
upon immunization of mice [114].
GNPs were also used to make a conjugate with the antigen
of the Newcastle disease virus of birds [115]. Ross chickens
were vaccinated once subcutaneously with a mixture of GNPs
and the vaccine antigen. High-titer specific antibodies were
obtained, and increased secretion of IFN-γwas noted.
The capsid (Cap) protein of a pathogenic porcine circovirus
was conjugated to 23-nm GNPs [116]. In vitro studies showed
that GNPs contributed to Cap protein phagocytosis. Mice
immunized twice subcutaneously with GNP/Cap showed
high production of virus-neutralizing antibodies. Similar
results were obtained with classical swine fever virus anti-
gen [117].
Soliman et al. [118] investigated the use of gold nanospheres
and nanorods as an adjuvant to develop antibodies against the
Rift Valley fever virus. They noted that the immune response
obtained by a single subcutaneous immunization of rats
depended on the shape of the GNPs. Peak antibody concentra-
tions were detected on day 30 after immunization with nanorods
and on day 45 with nanospheres. However, the content of IFN-γ
and IL-10 was higher with nanospheres than it was with nanorods.
Research is also underway to design GNP-based vaccines
against other dangerous pathogens, such as Ebola, Zika,
Marburg, Lassa, and Nipah.
Table 1 summarizes the literature data on the viral antigens
that have been conjugated with GNP carriers and then used
for immunization of animals.
3. Gold nanoparticles in the preparation of
antibacterial vaccines
GNPs have also been intensely used to develop antibodies and
vaccines against bacterial infections. For the first time, 15-nm
GNPs were used as an adjuvant to prepare antibodies to the
surface antigens of Yersinia pseudotuberculosis [119]. Rabbits
received two subcutaneous injections, each with 1 μg of the
antigen. The serum titer was as high as when CFA was used,
but the amount of antigen injected when GNPs were used as
an adjuvant was smaller by two orders of magnitude. GNPs as
an antigen carrier activated the phagocytic activity of the
lymphoid cells. Similar results were obtained by the same
research team in the preparation of antibodies against
Y. enterocolitica and Brucella abortus.
The efficacy of 15-nm GNPs coated with Y. pestis F1 antigen
in animal immunization was evaluated by Gregory et al. [120].
The conjugate (0.93 μg) was administered to mice once
Table 1. Conjugates of GNPs with viral antigens used for immunization and vaccination of animals.
Viral antigen GNP size and shape
Functionalization approach
(ligand)
Chemical nature of
antigen Refs.
Tick-borne encephalitis virus Nanospheres, 15 nm Adsorption Isolated protein [77]
Foot and mouth disease virus Nanospheres, 17 nm Chemosorption (cysteine) Synthetic peptide [79]
-
- Nanospheres, 2, 5, 8, 12, 17, 37, and 50 nm Chemosorption (cysteine) Synthetic peptide [80]
-
- Nanospheres, 15 nm Adsorption Synthetic peptide [82]
-
- Nanostars Adsorption Virus-like particles [83]
Influenza virus Nanospheres, 17 nm Chemosorption (cysteine) Synthetic peptide [79]
-
- Nanospheres, 15 nm Adsorption Isolated peptide [81]
-
- Nanospheres, 12 nm Adsorption Synthetic peptide [84]
-
- Nanospheres, 12 nm Chemosorption (cysteine) Synthetic peptide [87]
-
- Nanospheres, 15 nm Adsorption Synthetic peptide [88]
-
- Nanospheres, 18 nm Chemosorption (N
3
-PEG-SH,
SH-NTA)
Recombinant
proteins
[96]
Porcine transmissible gastroenteritis virus Nanospheres, 15 nm Adsorption Isolated protein [99]
Rabies virus Nanospheres, 15 nm Adsorption Isolated glycoprotein [100]
-
- Nanospheres, 15 nm Adsorption Isolated
ribonucleoprotein
[101]
West Nile fever virus Nanospheres, 20 and 40 nm; Nanorods, 40 × 20 nm;
Nanocubes, 40 nm
Chemosorption (PSS-MA) Isolated protein [102]
Respiratory syncytial virus Nanorods, 21 × 57 nm Chemosorption (EDC) Recombinant
glycoprotein
[103]
HIV Glyconanoparticles, 2 nm Adsorption Synthetic peptide [104]
Hepatitis B virus Nanospheres, 10 nm Adsorption Recombinant protein [107]
-
- Nanocages, 50 nm Adsorption Recombinant protein [108]
Hepatitis E virus Nanoclusters Direct synthesis Hepatitis E vaccine [109]
Hepatitis C virus Nanospheres, 15 nm Adsorption Synthetic peptide [110]
Dengue virus Nanospheres, 20, 40, and 80 nm Adsorption Recombinant
glycoprotein
[112]
Avian coronavirus Nanospheres, 100 nm Adsorption Recombinant protein [113]
Severe acute respiratory syndrome-related
coronavirus
Nanospheres, 40 nm Chemosorption (BSPP) Recombinant protein [114]
Newcastle disease virus Nanospheres Mixing Newcastle disease
vaccine
[115]
Porcine circovirus Nanospheres, 23 nm Adsorption Recombinant protein [116]
Classical swine fever virus Nanospheres, 24 nm Adsorption Recombinant protein [117]
Rift Valley fever virus Nanospheres, 20 nm; Nanorods, 40 × 20 nm Adsorption Isolated protein [118]
EXPERT REVIEW OF VACCINES 5
intramuscularly. The animals injected with GNP/F1 generated
the greatest IgG antibody response to the F1 antigen, as
compared to those injected with unconjugated F1 antigen in
phosphate-buffered saline or in Alhydrogel®. Sera obtained
against the F1 antigen coupled to GNPs were able to bind to
the F1 antigen in an enzyme-linked immunoassay. Similar
results were produced by Kireev et al. [121].
GNPs decorated with thiolated glycosides (synthetic analogs of
the capsular polysaccharides of Neisseria meningitidis)werepro-
posed as a synthetic antigen for use in immunization [122].
Conjugates of 15-nm GNPs with Salmonella typhimurium surface
antigens stimulated macrophage respiratory activity and the activ-
ity of the mitochondrial enzymes succinate dehydrogenase and
glycerophosphate dehydrogenase [42]. This stimulation may be
a significant factor determining the adjuvant properties of GNPs.
GNPs were included in the transmucosal delivery of tetanus
toxoid, used for vaccination against tetanus, a deadly disease
caused by Clostridium tetani [123]. Subcutaneous injection of
tetanus toxoid conjugated to 25-nm GNPs into mice gener-
ated a systemic response but did not cause any mucosal
response. However, the conjugate generated a significantly
higher mucosal response after oral administration. For enhan-
cing the immune response to tetanus toxoid, plant adjuvants
(saponins) from Quillaja saponaria [124] and Asparagus race-
mosus [125] were used together with GNPs. GNPs were also
used in the preparation of a vaccine against botulism,
a particularly dangerous disease caused by C. botulinum [126].
GNPs (average diameter, ~ 2 nm) were conjugated to a syn-
thetic tetrasaccharide epitope, an analog of the capsular polysac-
charide of Streptococcus pneumoniae type 14 [127,128]. The
obtained glyconanoparticles conjugated with T-helper peptide
generated specific high-titer IgG after a single intradermal immu-
nization of mice with 3 μg of the conjugate. An increase in the
content of the cytokines IL-2, IL-4, IL-5, IL-17, and IFN-γconfirmed
that the nanoparticles activate T helpers. The antisaccharide anti-
bodies stimulated the phagocytosis of type 14 bacteria by human
leukocytes, which indicates the functionality of the antibodies.
A new strategy for preparing antibodies to tuberculin was
described [129] that used the adjuvant properties of GNPs.
Tuberculin is a mixture of the surface antigens of various types
of mycobacteria (Mycobacterium tuberculosis, M. bovis,and
M. avium). When injected intradermally, it causes a specific
delayed-type hypersensitivity response in infected or vaccinated
patients. Polyclonal antituberculin antibodies were raised by
injecting 7.5 μg of tuberculin conjugated to 15-nm GNPs into
rabbits four times intramuscularly. The obtained antibodies had
a high titer, whereas in the control animals, which were given
nonconjugated tuberculin, no antibodies were found in the
blood serum. The antituberculin antibodies were used to detect
mycobacteria by ELISA and by light and electron microscopy.
GNPs were found to partially remove the toxic effect of
tuberculin on rat peritoneal macrophages owing to their
penetration into the intracellular space. This decrease in toxi-
city contributed to the better development of the humoral
response and to the elaboration of antituberculin antibodies.
In addition, GNPs decorated with tuberculin enhanced the
adhesion of phagocytic cells to microbial cells. Preliminary
vaccination results for GNPs conjugated to guinea pig tuber-
culin showed a protective effect [129].
For immunoassay of mycobacteria, GNPs were conjugated
to the penta- and hexasaccharide fragments of mycobacterial
lipoarabinomannan by using aglycon spacers of various
lengths [130]. Rabbit polyclonal antibodies to these conju-
gates were developed. The antisera obtained by hyperimmu-
nization of rabbits detected with a high titer both
lipoarabinomannan oligosaccharides and mycobacterial cells.
Gao et al. [131] coated 30-nm GNPs with bacterial vesicles of
the outer membrane of Escherichia coli (Figure 4). When injected
three times subcutaneously, the conjugate induced rapid activa-
tion and maturation of dendritic cells in the lymph nodes of the
vaccinated mice. In addition, vaccination with the conjugate
gave rise to antibodies that had a higher avidity than those
obtained by vaccination with outer membrane vesicles only.
The production of IFN-γand IL-17 increased too, which indicates
strong Th1 and Th17 cellular responses to the bacteria.
Another prototype vaccine against E. coli was demon-
strated by Sanchez-Villamil et al. [132]. GNPs were coated
with the specific immunogenic LomW and EscC antigens of
the enterohemorrhagic strain E. coli O157: H7. Mice were
injected three times subcutaneously with GNPs/LomW, GNPs/
EscC, or a combination thereof, containing equivalent
amounts of both immunogens. As a result, higher-titer IgG
were found in sera and higher-titer IgA were found in feces.
Serum IgG titers correlated with a significant decrease in the
intestinal colonization of E. coli at 3 days after inoculation. The
sera from the mice immunized with antigen-coated GNPs
reduced the adhesion of E. coli O157: H7 and two other
E. coli pathotypes to human intestinal epithelial cells. In addi-
tion, the sera had antigen-specific bactericidal properties.
An effective vaccine against the human pathogen Listeria
monocytogenes was made by conjugating the listeriolysin
O peptide LLO91-99 to gold glyconanoparticles [133].
a single intraperitoneal or intravenous immunization of mice
with the conjugate induced a specific T-cell response and
protected the animals against Listeria infection. The protection
correlated with an increase in the number of splenic CD4
+
and
CD8
+
T cells, NK cells, and CD8α
+
dendritic cells, and it also
correlated with an increase in the production of the cytokines
IL-12, IFN-γ, TNF-α, and MCP-1 after infection. Subsequently, it
was shown that vaccination of pregnant mice with gold gly-
conanoparticles carrying L. monocytogenes peptides protects
Figure 4. Schematic illustration of modulation of antibacterial immunity with
bacterial-membrane-coated nanoparticles [130].
6L. A. DYKMAN
newborns from listeriosis [134]. Newborn mice born to vacci-
nated females were free of bacteria and healthy, whereas
nonvaccinated mice had obvious lesions of the brain and skin.
An interesting vaccination schedule against glanders,
a disease caused by Burkholderia mallei, was proposed by
Gregory et al. [135]. GNPs (average diameter, 15 nm) were
first covalently bound to recombinant protein carriers the
Hc fragment of tetanus toxin, the hemolysin coregulated pro-
tein produced by both B. mallei and B. pseudomallei, and the
flagellin produced by B. pseudomallei.Theconjugatessopre-
pared were functionalized with purified LPS from a nonvirulent
B. thailandensis strain. Mice were immunized three times intra-
nasally, and the dose used was 0.93 μg. The GNP/protein/LPS
conjugates generated significantly higher antibody titers than
did native LPS. In addition, they improved protection against
a lethal inhalation challenge of B. mallei in the murine model of
infection. The proposed scheme in the form of aerosol immu-
nization was successfully tested on rhesus monkeys [136].
a similar approach was developed for vaccination against
B. pseudomallei myeloidosis [137]. Mice immunized three
times subcutaneously with GNPs/protein/LPS generated high-
titer antibodies. Importantly, the immunized animals survived
nearly 100% and their lungs were less contaminated with bac-
teria after a lethal infection with B. pseudomallei.
Dakterzada et al. [138] evaluated the efficacy of GNPs con-
jugated with the N-terminal domains of Pseudomonas aeruginosa
flagellin as an immunogen in the vaccination of mice.
Nanoparticles were complexed with a recombinant flagellin (1-
161). Flagellin, GNP/flagellin, and flagellin emulsified with CFA
were administered twice subcutaneously to BALB/C mice at
adoseof10μg. The mice given GNPs/flagellin elaborated
more antiflagellin antibodies than did those that either were
not immunized at all or were given adjuvant-free flagellin. In
a whole-cell ELISA, these antibodies efficiently recognized native
flagellin on the bacterial cells. An assay of opsonophagocytosis
demonstrated the functional activity and specificity of the anti-
flagellin antibodies prepared by using the GNP/flagellin conju-
gate against the homologous strain.
The acidic serine protease AprV2 secreted by virulent
Dichelobacter nodosus, the causative agent of infectious podo-
dermatitis, was conjugated to 20-nm GNPs [139]. The nano-
vaccine induced higher IgG responses than did native AprV2
or the vaccine adjuvanted with monophosphoryl lipid A.
Conjugates of 15-nm GNPs with two isolated Francisella
tularensis antigens, a protective antigenic complex and
a glycosylated protein complex, were used to obtain antitular-
emia sera and vaccinate animals [140]. Subcutaneous immu-
nization of mice with GNPs decorated with a glycosylated
protein complex was more effective than was immunization
with an unconjugated antigen. This greater effectiveness was
manifested as an increase in the protection ability and titers of
antibodies. The conjugation of GNPs to both antigens in the
immunization of rabbits afforded sera with a high titer of
specific antibodies in a relatively short period and with mini-
mal consumption of the antigen. The use in ELISA of the
immunoglobulins isolated from the sera allowed detection of
F. tularensis cells of different subspecies, enabling their further
use in the manufacture of diagnostic agents for tularemia.
Recombinant immunodominant antigens of Coxiella burne-
tii, the causative agent of Q fever, were conjugated to GNPs
[141]. Immunization of mice and guinea pigs gave rise to
specific antibodies and activated the response of the CD4
+
T cells. The protective effect of the GNP-associated antigens
was comparable to that of the commercial vaccine used.
Conjugates of 15-nm GNPs with seven Vibrio cholerae anti-
gens were synthesized in [142]. Rabbits were immunized five
times subcutaneously, and the resultant polyclonal antibodies
to the test antigens showed a high specific activity. By using
white laboratory mice, the protective activity of the conju-
gates was evaluated during infection of vaccinated animals,
with a commercial vaccine as a control. The cholera vaccine
prototypes made with GNPs as a carrier and an adjuvant
corresponded to the commercial chemical vaccine in terms
of protection efficiency.
Table 2 summarizes the literature data on the bacterial
antigens that have been conjugated with GNP carriers and
then used for immunization of animals.
4. Gold nanoparticles in the preparation of
antiparasitic vaccines
Besides being used as an adjuvant in immunization against
viral and bacterial diseases, GNPs are also used to develop
antibodies and vaccines against parasitic infections. Parween
et al. [143] reported that 17-nm GNPs in complex with recom-
binant peptides of the C-terminal 19kDa fragment of mero-
zoite surface protein 1 from the malaria pathogen Plasmodium
falciparum had a pronounced immunogenic activity when
combined with the adjuvant Alhydrogel®. Mice were immu-
nized three times subcutaneously at a dose of 25 μg.
Antibodies against the weakly immunogenic peptides conju-
gated to GNPs recognized the native protein on the parasites
surface and inhibited the invasion of P. falciparum in an in vitro
assay.
Another immunogen was proposed to make an antimalarial
vaccine the P. falciparum surface protein Pfs25, expressed in
zygotes and ookinetes [144]. The recombinant protein Pfs25
was attached to various GNPs, including nanospheres, nanos-
tars, nanocages, and nanoprisms. Mice were immunized with
the resulting conjugates three times intramuscularly at a dose
of 10 μg. This yielded high-titer antibodies, the highest titers
being obtained with gold nanospheres and nanostars. In
membrane feeding assays, the antibodies blocked the trans-
mission of parasites to mosquitoes.
GNPs were also used to prepare anti-idiotypic antibodies [145].
These antibodies were used for an indirect and a competitive
ELISA of the opisthorchiasis pathogen Opisthorchis felineus.Work
is underway to develop a vaccine against Babesia canis,thecau-
sative agent of canine piroplasmosis, by using the adjuvant prop-
erties of GNPs.
For vaccination against schistosomiasis, a parasitic disease
caused by Schistosoma mansoni trematodes, gold nanorods
were complexed with the recombinant protein rSm2, located
on the surface of larvae and adult worms [146]. Mice were
immunized three times intraperitoneally with 2 μg of the
conjugate. After immunization, a Th1 immunological response
EXPERT REVIEW OF VACCINES 7
was observed with higher production of IFN-γ, mainly by CD4
+
and CD8
+
T cells. In addition, the conjugate activated dendritic
cells in vitro, enhancing the expression of MHCII and MHCI and
the production of IL-1β. After vaccination with the conjugate,
followed by a challenge with S. mansoni cercariae, the number
of worms in the livers portal vein and the number of eggs in
the liver tissues decreased, as compared with the control
group.
Table 3 summarizes the literature data on the parasite
antigens that have been conjugated with GNP carriers and
then used for immunization of animals.
5. Expert opinion
GNPs have been used to prepare antibodies and vaccines
against more than 45 pathogens of viral, bacterial, and para-
sitic infections. The sizes of the GNPs used for this purpose
have ranged from 2 to 100 nm. Moreover, in most studies,
particle diameters ranging from 15 to 50 were recognized as
optimal. Variously shaped GNPs have been used, including
nanospheres, nanorods, nanocages, nanostars, nanocubes,
nanoshells, nanoprisms, and nanoclusters. Most researchers
have indicated that the best effect is achieved with gold
nanospheres as antigen carriers.
Various methods have been used to functionalize GNPs with
target antigens. Conjugation has been carried out both by
physical adsorption and by chemisorption. Passive adsorption
of an antigen on the particle surface occurs because of the
electrostatic and hydrophobic interactions. Coulomb
interactions between the H
2
N groups of lysine and citrate
ions on the surface of gold nanoparticles have also been
reported. Recently, information has appeared about an impor-
tant role of the SH groups of cysteine molecules in protein
binding to the surface of gold particles. It is known that
between sulfur and gold atoms, there arises a donoracceptor
(semipolar) bond (dative bound). This fact has led to the use of
alkanethiol linkers (chemisorption method) for more stable
attachment of biomolecules to GNPs. In addition, GNPs have
often been conjugated to protein or polysaccharide linkers
before being functionalized with an antigen. Most often, gold
glyconanoparticles have been used as immunomodulators.
What other adjuvants can be used with GNPs remains an
open question. Specific antibodies against target antigens
have been obtained by using GNPs as the sole adjuvant in
combination with immune response enhancers such as CFA,
CpG-ODNs, MontanideTM, AdvaxTM, Alhydrogel®, Imiquimod,
monophosphoryl lipid A, ISA206, and saponins. Moreover,
both natural (native) antigens and genetically engineered
(recombinant) ones have been used as immunogens.
Various animals have been used for immunization and vac-
cination, including mice, rats, guinea pigs, rabbits, chickens, and
monkeys. The methods used to administer immunogens have
included intraperitoneal, subcutaneous, intradermal, intramus-
cular, intravenous, intranasal, and oral. Moreover, the immuni-
zation doses of the antigens varied within a very wide range
for example, from 100 ng to 50 μg for mice or from 1 μgto
220 μg for rabbits. The frequency of administration also varied
from single to sixfold.
Table 2. Conjugates of GNPs with bacterial antigens used for immunization and vaccination of animals.
Bacterial antigen GNP size and shape Functionalization approach (ligand) Chemical nature of antigen Refs.
Yersinia pseudotuberculosis Nanospheres, 15 nm Adsorption Isolated protein [119]
Yersinia pestis Nanospheres, 15 nm Chemosorption (NHS, EDC) Recombinant protein [120]
-
- Nanospheres, 15 nm Adsorption Isolated protein [121]
Neisseria meningitides*Nanospheres, 5 nm Chemosorption (thiols) Synthetic carbohydrate [122]
Salmonella typhimurium Nanospheres, 15 nm Adsorption Isolated protein [42]
Clostridium tetani*Glyconanoparticles, 25 nm Adsorption Isolated protein [123]
Streptococcus pneumoniae*Glyconanoparticles, 2 nm Direct synthesis Synthetic carbohydrate [127]
Mycobacterium tuberculosis Nanospheres, 15 nm Adsorption Isolated peptide [129]
-
-* Nanospheres, 15 nm Adsorption Synthetic carbohydrate [130]
Escherichia coli Nanospheres, 30 nm Adsorption Isolated bacterial membrane [131]
-
- Nanospheres, 15 nm Chemosorption (MHDA) Recombinant proteins [132]
Listeria monocytogenes*Glyconanoparticles, 1.5 nm Direct synthesis Synthetic peptides [133]
Burkholderia mallei*Nanospheres, 15 nm Chemosorption (NHS, EDC) Isolated LPS [135]
Burkholderia pseudomallei Nanospheres, 15 nm Chemosorption (NHS, EDC) Isolated LPS [137]
Pseudomonas aeruginosa Nanospheres, 15 nm Adsorption Recombinant protein [138]
Dichelobacter nodosus Nanospheres, 20 nm Adsorption Recombinant protein [139]
Francisella tularensis Nanospheres, 15 nm Adsorption Isolated proteins [140]
Coxiella burnetii Nanospheres, 15 nm Adsorption Recombinant proteins [141]
Vibrio cholerae Nanospheres, 15 nm Adsorption Isolated proteins and LPS [142]
*
Glyconanovaccine
Table 3. Conjugates of GNPs with parasite antigens used for immunization and vaccination of animals.
Parasite antigen GNP size and shape
Functionalization approach
(ligand)
Chemical nature of
antigen Refs.
Plasmodium
falciparum
Nanospheres, 17 nm Adsorption Recombinant peptides [143]
-
- Nanospheres, 30 nm; nanostars, 50 nm; nanocages, 60 nm;
nanoprisms, 40 nm
Chemosorption (4-ATP) Recombinant protein [144]
Babesia canis Nanospheres, 15 nm Adsorption Isolated proteins [unpublished
data]
Schistosoma
mansoni
Nanorods Adsorption Recombinant protein [146]
8L. A. DYKMAN
The antibody elaboration during immunization with GNP
antigen conjugates enhanced the secretion of cytokines
most often, IFN-γ, TNF-α, IL-1β, IL-4, IL-6, IL-10, and IL-12,
and less often, IL-2, IL-5, IL-17, IL-18, MCP-1, and GM-CSF.
Several researchers have noted the stimulation of the respira-
tory activity of peritoneal macrophages and spleen lympho-
cytes and the activation of the macrophage mitochondrial
enzymes succinate dehydrogenase and glycerophosphate
dehydrogenase. Immunization with GNPantigen conjugates
enhanced the proliferation of the CD4
+
and CD8
+
T cells and
NK cells and stimulated both Th1 and Th2 immune responses.
Other effects noted were the activation of dendritic cell
maturation for antigen processing and presentation, increased
expression of MHCII and MHCI, and activation of NLRP3
inflammasomes. The active uptake of GNPs by macrophages
contributed to the processing of antigens. Splenocytes of the
animals immunized with GNPs had higher proliferation rates
than the cells immunized with the native antigen alone, which
shows the ability of GNP-based carriers to induce enhanced
cellular responses to the associated antigens. Often, GNPs
decorated with bacterial antigens enhanced the phagocytic
adhesion of microbial cells.
Antibodies obtained by using GNPs have been successfully
applied to the immunochemical identification of pathogens
with various solid-phase and microscopic methods and with
biosensor devices. Vaccination of animals with GNPantigen
conjugates decreased the symptoms of infections and pro-
tected the animals challenged with virulent pathogens (up
to lethal doses). In some cases, the vaccinated animals sur-
vived 100%. In most studies, the low toxicity of the gold carrier
itself was noted [147].
Currently, the immunostimulatory activity of nanomaterials,
including GNPs, as adjuvants is explained by the following
mechanisms (Figure 5). Antigennanocarrier conjugates are
more effectively delivered to the lymph nodes and induce
a depot effect, which contributes to the steady and gradual
release of the antigens. The use of nanoparticles as adjuvants
helps antigens to concentrate on the membrane of dendritic
cells and activates their maturation and the expression of
MHCI and MHCII. The spatial organization (multivalence) of
antigens on the surface of the particles enhances the prolif-
eration of T cells and the activation of B cells. Both events are
accompanied by the release of soluble mediators such as
cytokines, chemokines, and immunomodulating molecules,
which regulate the immune response.
Antigen-presenting cells are crucial for the response to
vaccines. Among the antigen-presenting cells, dendritic cells
are especially important for the primary immune response,
because they control the activation of the CD4
+
and CD8
+
T cells, which assist in the induction of antibodies and exhibit
direct cytotoxic activity. Therefore, the key application of
nanoparticles in immunology is the modulation of the func-
tions of antigen-presenting cells. By programmingthe activa-
tion state of dendritic or other antigen-presenting cells,
nanoparticles directly affect the induction of cellular and
humoral immunity [148].
It should be emphasized that GNPs are not biodegradable.
Therefore, the biodistribution and excretion kinetics have to
be studied comprehensively for different animal models. As
the excretion of accumulated particles from the liver and
spleen can take up to 34 months, the question as to the
injected doses and possible inflammation processes is still of
great importance. Bioaccumulated GNPs can interfere with
different diagnostic techniques, or accumulated GNPs can
exhibit catalytic properties. All these concerns, together with
potential toxicity, are big limitations of GNPs on a successfully
clinical translation. Nowadays, despite the huge numbers of
studies regarding the synthesis and functionalization of GNPs
(different shapes, coatings, sizes, charges, etc.), there are very
few nanomaterial-based pharmaceuticals on the market.
Figure 5. Mechanisms of immune system activation by engineered nanomaterials [11].
EXPERT REVIEW OF VACCINES 9
The synthetic and natural polymeric biodegradable nanoma-
terials also can serve as antigen carriers. The advantages of
nanoparticles of this type are that they are utilized well in the
organism, the target substance is highly efficiently involved, there
is a higher capability to overcome different physiological barriers,
and there are less systematic side effects. The action mechanisms
of biodegradable nanoparticles and GNPs as carriers of antigens
intheimmunesystemareprobablysimilar.Thesetwotypesof
nanoobjects can compete in the development of a new genera-
tion of vaccines, taking into account data on bioinertness, low
toxicity, and the good excretion of gold nanoparticles from the
organism with the involvement of a hepatobiliary system [147].
Thus, GNPs, which have adjuvant properties, can be an
excellent tool in the design of effective vaccines against infec-
tious diseases.
Acknowledgments
I thank Mr DN Tychinin for his help in preparation of the manuscript.
Funding
This work was supported by the Russian Science Foundation under grant
no. 19-14-00077.
Declaration of interest
The author has no relevant affiliations or financial involvement with any
organization or entity with a financial interest in or financial conflict with
the subject matter or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties.
Reviewer Disclosures
Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
ORCID
Lev A. Dykman http://orcid.org/0000-0003-2440-6761
References
Papers of special note have been highlighted as either of interest ()orof
considerable interest (••) to readers.
1. Gregory AE, Titball R, Williamson D. Vaccine delivery using
nanoparticles. Front Cell Infect Microbiol. 2013;3:13.
2. Zaman M, Good MF, Toth I. Nanovaccines and their mode of action.
Methods. 2013;60(3):226231.
3. Prashant CK, Kumar M, Dinda AK. Nanoparticle based tailoring of
adjuvant function: the role in vaccine development. J Biomed
Nanotechnol. 2014;10(9):23172331.
4. Zhao L, Seth a, Wibowo N, et al. Nanoparticle vaccines. Vaccine.
2014;32(3):327337.
5. Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine
delivery systems. Pharm Res. 2014;31(10):25632582.
6. Zhu M, Wang R, Nie G. Applications of nanomaterials as vaccine
adjuvants. Hum Vaccin Immunother. 2014;10(9):27612774.
7. Liu Y, Xu Y, Tian Y, et al. Functional nanomaterials can optimize the
efficacy of vaccines. Small. 2014;10(22):45054520. .
8. Irvine DJ, Hanson MC, Rakhra K, et al. Synthetic nanoparticles for
vaccines and immunotherapy. Chem Rev. 2015;115
(19):1110911146. .
9. Maughan CN, Preston SG, Williams GR. Particulate inorganic adju-
vants: recent developments and future outlook. J Pharm
Pharmacol. 2015;67(3):426449.
10. Gupta a, Das S, Schanen B, et al. Adjuvants in micro- to nanoscale:
current state and future direction. Wiley Interdiscip Rev Nanomed
Nanobiotechnol. 2016;8(1):6184.
11. Sun B, Xia T. Nanomaterial-based vaccine adjuvants. J Mater Chem
B. 2016;4(33):54965509.
12. Yang L, Li W, Kirberger M, et al. Design of nanomaterial based
systems for novel vaccine development. Biomater Sci. 2016;4
(5):785802. .
13. Andrade LM, Cox L, Versiani AF, et al. a growing world of small
things: a brief review on the nanostructured vaccines. Future Virol.
2017;12(12):767779. .
14. Shen Y, Hao T, Ou S, et al. Applications and perspectives of nano-
materials in novel vaccine development. MedChemComm. 2017;9
(2):226238. .
15. Li X, Wang X, Ito a. Tailoring inorganic nanoadjuvants towards
next-generation vaccines. Chem Soc Rev. 2018;47(13):49544980.
16. Hess KL, Medintz IL, Jewell CM. Designing inorganic nanomaterials
for vaccines and immunotherapies. Nano Today. 2019;27:7398.
17. Facciolà a, Visalli G, Laganà P, et al. The new era of vaccines: the
nanovaccinology. Eur Rev Med Pharmacol Sci. 2019;23
(16):71637182. .
18. Steabben DB. Studies on the physiological action of colloids. The
action of colloidal substances on blob-elements and antibody
content. Br J Exp Path. 1925;6:113.
19. Pacheco G. Studies on the action of metallic colloids on
immunization. Mem Inst Oswaldo Cruz. 1925;18:119149.
20. Zozaya J. Carbohydrates adsorbed on colloids as antigens. J Exp
Med. 1932;55(3):325351.
21. Gros O, OConnor JM. Einige Beobachtungen bei colloidalen
Metallen mit Rucksicht auf ihre physikalisch-chemischen
Eigenschaften und deren pharmakologische Wirkungen. Archiv
F Exp Path U Pharmakol. 1911;64(56):456467.
22. Zilber LA, Friese WW. Über die antigene Eigenschaften kolloidaler
Metalle. Zh Eksp Biol. 1929;11:128135.
23. Syed S, Zubair a, Frieri M. Immune response to nanomaterials:
implications for medicine and literature review. Curr Allergy
Asthma Rep. 2013;13(1):5057.
24. Farrera C, Fadeel B. It takes two to tango: understanding the
interactions between engineered nanomaterials and the immune
system. Eur J Pharm Biopharm. 2015;95:312.
25. Petrarca C, Clemente E, Amato V, et al. Engineered metal based
nanoparticles and innate immunity. Clin Mol Allergy. 2015;13(1):13.
26. Kononenko V, Narat M, Drobne D. Nanoparticle interaction with the
immune system. Arh Hig Rada Toksikol. 2015;66(2):97108.
27. Luo Y-H, Chang LW, Lin P. Metal-based nanoparticles and the
immune system: activation, inflammation, and potential
applications. Biomed Res Int. 2015;2015:143720.
28. Seth a, Oh D-B, Lim YT. Nanomaterials for enhanced immunity as
an innovative paradigm in nanomedicine. Nanomedicine (Lond).
2015;10(6):959975.
29. Bracho-Sanchez E, Xia CQ, Clare-Salzler MJ, et al. Micro and nano
material carriers for immunomodulation. Am J Transplant. 2016;16
(12):33623370. .
30. Ilinskaya AN, Dobrovolskaia MA. Understanding the immunogeni-
city and antigenicity of nanomaterials: past, present and future.
Toxicol Appl Pharmacol. 2016;299:7077.
This review extensively explores the current understanding of
the immunogenicity and antigenicity of nanomaterials.
31. Fang RH, Zhang L. Nanoparticle-based modulation of the immune
system. Annu Rev Chem Biomol Eng. 2016;7(1):13.113.22.
32. David CAW, Owen a, Liptrott NJ. Determining the relationship
between nanoparticle characteristics and immunotoxicity: key chal-
lenges and approaches. Nanomedicine (Lond). 2016;11
(11):14471464.
10 L. A. DYKMAN
33. Moyano DF, Liu Y, Peer D, et al. Modulation of immune response
using engineered nanoparticle surfaces. Small. 2016;12(1):7682. .
34. Najafi-Hajivar S, Zakeri-Milani P, Mohammadi H, et al. Overview on
experimental models of interactions between nanoparticles and
the immune system. Biomed Pharmacother. 2016;83:13651378.
35. Chattopadhyay S, Chen J-Y, Chen H-W, et al. Nanoparticle vaccines
adopting virus-like features for enhanced immune potentiation.
Nanotheranostics. 2017;1(3):244260. .
36. Frey M, Bobbala S, Karabin N, et al. Influences of nanocarrier
morphology on therapeutic immunomodulation. Nanomedicine
(Lond). 2018;13(14):17951811. .
37. Kelly HG, Kent SJ, Wheatley AK. Immunological basis for enhanced
immunity of nanoparticle vaccines. Expert Rev Vaccines. 2019;18
(3):269280.
38. Fadeel B. Hide and seek: nanomaterial interactions with the
immune system. Front Immunol. 2019;10:133.
39. Dykman LA, Khlebtsov NG. Uptake of engineered gold nanoparti-
cles into mammalian cells. Chem Rev. 2014;114(2):12581288.
40. Patel PC, Giljohann DA, Daniel WL, et al. Scavenger receptors med-
iate cellular uptake of polyvalent oligonucleotide-functionalized
gold nanoparticles. Bioconjug Chem. 2010;21(12):22502256. .
41. Franca a, Aggarwal P, Barsov EV, et al. Macrophage scavenger
receptor a mediates the uptake of gold colloids by macrophages
in vitro. Nanomedicine (Lond). 2011;6(7):11751188.
42. Staroverov SA, Aksinenko NM, Gabalov KP, et al. Effect of gold
nanoparticles on the respiratory activity of peritoneal
macrophages. Gold Bull. 2009;42(2):153156.
43. Yen H-J, Hsu S-H, Tsai C-L. Cytotoxicity and immunological
response of gold and silver nanoparticles of different sizes. Small.
2009;5(13):15531561.
44. Cheung W-H, Chan VS-F, Pang H-W, et al. Conjugation of latent
membrane protein (LMP)-2 epitope to gold nanoparticles as highly
immunogenic multiple antigenic peptides for induction of Epstein
Barr virus-specific cytotoxic T-Lymphocyte responses in vitro.
Bioconjug Chem. 2009;20(1):2431. .
45. Cruz LJ, Rueda F, Cordobilla B, et al. Targeting nanosystems to
human DCs via Fc receptor as an effective strategy to deliver
antigen for immunotherapy. Mol Pharm. 2011;8(1):104116. .
46. Bartneck M, Keul HA, Singh S, et al. Rapid uptake of gold nanorods
by primary human blood phagocytes and immunomodulatory
effects of surface chemistry. ACS Nano. 2010;4(6):30733086. .
47. Chekanov AV, Baranova OA, Levin AD, et al. Influence of gold
nanoparticles on activation of human blood neutrophils.
Biophysics. 2013;58:385388.
48. Le Guevel X, Palomares F, Torres MJ, et al. Nanoparticle size influ-
ences the proliferative responses of lymphocyte subpopulations.
RSC Adv. 2015;5(104):8530585309. .
49. Liptrott NJ, Kendall E, Nieves DJ, et al. Partial mitigation of gold
nanoparticle interactions with human lymphocytes by surface func-
tionalization with a mixed matrix. Nanomedicine (Lond). 2014;9
(16):24672479. .
50. Li Y, Italiani P, Casals E, et al. Assessing the immunosafety of
engineered nanoparticles with a novel in vitro model based on
human primary monocytes. ACS Appl Mater Interfaces. 2016;8
(42):2843728447. .
51. Srijampa S, Buddhisa S, Ngernpimai S, et al. Effects of gold nano-
particles with different surface charges on cellular internalization
and cytokine responses in monocytes. BioNanoSci. 2019;9
(3):580586. .
52. Blecher K, Nasir a, Friedman a. The growing role of nanotechnol-
ogy in combating infectious disease. Virulence. 2011;2
(5):395401.
53. Fan Y, Moon JJ. Particulate delivery systems for vaccination against
bioterrorism agents and emerging infectious pathogens. Wiley
Interdiscip Rev Nanomed Nanobiotechnol. 2017;9(1):e1403.
54. Neto LMM, Kipnis a, Junqueira-Kipnis AP. Role of metallic nanopar-
ticles in vaccinology: implications for infectious disease vaccine
development. Front Immunol. 2017;8:239.
55. Pati R, Shevtsov M, Sonawane a. Nanoparticle vaccines against
infectious diseases. Front Immunol. 2018;9:2224.
56. Sokolova V, Westendorf AM, Buer J, et al. The potential of nano-
particles for the immunization against viral infections. J Mater
Chem B. 2015;3(24):47674779. .
57. Glass JJ, Kent SJ, De Rose R. Enhancing dendritic cell activation and
HIV vaccine effectiveness through nanoparticle vaccination. Expert
Rev Vaccines. 2016;15(6):719729.
58. Liu Y, Chen C. Role of nanotechnology in HIV/AIDS vaccine
development. Adv Drug Deliv Rev. 2016;103:7689.
59. Deng L, Wang B-Z. a perspective on nanoparticle universal influ-
enza vaccines. ACS Infect Dis. 2018;4(12):16561665.
60. Al-Halifa S, Gauthier L, Arpin D, et al. Nanoparticle-based vaccines
against respiratory viruses. Front Immunol. 2019;10:22.
61. Lin LC-W, Chattopadhyay S, Lin J-C, et al. Advances and opportu-
nities in nanoparticle- and nanomaterial-based vaccines against
bacterial infections. Adv Healthc Mater. 2018;7(13):e1701395.
62. Huang W-C, Sia ZR, Lovell JF. Adjuvant and antigen systems for
malaria transmission-blocking vaccines. Adv Biosys. 2018;2(10):
e1800011.
63. Vartak a, Sucheck SJ. Recent advances in subunit vaccine carriers.
Vaccines (Basel). 2016;4(2):12.
64. Fujita Y, Taguchi H. Current status of multiple antigen-presenting
peptide vaccine systems: application of organic and inorganic
nanoparticles. Chem Cent J. 2011;5(1):48.
65. Marasini N, Skwarczynski M, Toth I. Oral delivery of nanoparticle-based
vaccines. Expert Rev Vaccines. 2014;13(11):13611376.
66. Wang S, Liu H, Zhang X, et al. Intranasal and oral vaccination with
protein-based antigens: advantages, challenges and formulation
strategies. Protein Cell. 2015;6(7):480503. .
67. Kaurav M, Minz S, Sahu K, et al. Nanoparticulate mediated transcu-
taneous immunization: myth or reality. Nanomedicine. 2016;12
(4):10631081. .
68. Comber JD, Bamezai a. Gold nanoparticles (AuNPs): a new frontier
in vaccine delivery. J Nanomedine Biotherapeutic Discov. 2015;5
(4):4.
69. Salazar-González JA, González-Ortega O, Rosales-Mendoza S. Gold
nanoparticles and vaccine development. Expert Rev Vaccines.
2015;14(9):11971211.
70. Dykman LA, Khlebtsov NG. Immunological properties of gold
nanoparticles. Chem Sci. 2017;8(3):17191735.
71. Carabineiro SAC. Applications of gold nanoparticles in nanomedi-
cine: recent advances in vaccines. Molecules. 2017;22:857.
•• This review explores the important role of gold nanoparicles in
vaccine design and their use as an adjuvant and a carrier.
72. Dykman LA, Khlebtsov NG. Gold Nanoparticles in Biomedical
Applications. Boca Raton: CRC Press; 2017.
73. Shiosaka S, Kiyama H, Wanaka a, et al. a new method for producing
a specific and high titre antibody against glutamate using colloidal
gold as a carrier. Brain Res. 1986;382(2):399403. .
74. Dykman LA, Sumaroka MV, Staroverov SA, et al. Immunogenic
properties of colloidal gold. Biol Bull Russ Acad Sci. 2004;31
(1):7579. .
75. Dykman LA, Staroverov SA, Bogatyrev VA, et al. Adjuvant proper-
ties of gold nanoparticles. Nanotechnol Russ. 2010;5(1112):748-
761. .
76. Dykman LA, Staroverov SA, Bogatyrev VA, et al. Gold nanoparticles
as an antigen carrier and an adjuvant. Hauppauge: Nova Science
Publishers; 2010.
77. Demenev VA, Shchinova MA, Ivanov LI, et al. Perfection of metho-
dical approaches to designing vaccines against tick-borne
encephalitis. Vopr Virusol. 1996;41(3):107110.
78. Gaidamovich SY, Demenev VA, Obukhova VR. Differentiation in the
complement fixation test of the viruses of tick-borne encephalitis
complex by means of a type-specific soluble antigen. Acta Virol.
1985;29(2):143149.
79. Chen Y-S, Hung Y-C, Liau I, et al. Assessment of the in vivo toxicity
of gold nanoparticles. Nanoscale Res Lett. 2009;4(8):858864. .
80. Chen Y-S, Hung Y-C, Liau I, et al., Assessment of gold nanoparticles
as a size-dependent vaccine carrier for enhancing the antibody
response against synthetic foot-and-mouth disease virus peptide.
Nanotechnology. 2010;21(19): 195101..
EXPERT REVIEW OF VACCINES 11
This research article extensively explores the use of gold nano-
particles as a vaccine carrier for enhancing the antibody
response against synthetic foot-and-mouth disease virus
peptide.
81. Mezhenny PV, Staroverov SA, Volkov AA, et al. Construction of
conjugates of colloidal selenium and colloidal gold with the pro-
tein of influenza virus and the study of their immunogenic
properties. Bull Saratov State Agrarian Univ. 2013;2:2932.
82. Dykman LA, Staroverov SA, Mezhenny PV, et al. Use of a synthetic
foot-and-mouth disease virus peptide conjugated to gold nano-
particles for enhancing immunological response. Gold Bull. 2015;48
(12):93101. .
83. Teng Z, Sun S, Chen H, et al. Golden-star nanoparticles as adjuvant
effectively promotes immune response to foot-and-mouth disease
virus-like particles vaccine. Vaccine. 2018;36(45):67526760. .
84. Tao W, Ziemer KS, Gill HS. Gold nanoparticleM2e conjugate cofor-
mulated with CpG induces protective immunity against influenza
a virus. Nanomedicine (Lond). 2014;9(2):237251. .
•• This research article extensively explores the mechanisms of
protective immunity against influenza a virus, with gold nano-
particle conjugates used as an adjuvant.
85. Tao W, Gill HS. M2e-immobilized gold nanoparticles as influenza
a vaccine: role of soluble M2e and longevity of protection. Vaccine.
2015;33(20):23072315.
86. Tao W, Hurst B, Shakya AK, et al. Consensus M2e peptide conju-
gated to gold nanoparticles confers protection against H1N1, H3N2
and H5N1 influenza a viruses. Antiviral Res. 2017;141:6272.
87. Bimler L, Song AY, Le DT, et al. AuNP-M2e + sCpG vaccination of
juvenile mice generates lifelong protective immunity to influenza
a virus infection. Immun Ageing. 2019;16(1):23. .
88. Dykman LA, Staroverov SA, Fomin AS. Effect of M2e peptidegold
nanoparticle conjugates on development of anti-influenza antibo-
dies. Gold Bull. 2018;51(4):197203.
89. Weiner GJ, Liu HM, Wooldridge JE, et al. Immunostimulatory oligo-
deoxynucleotides containing the CpG motif are effective as
immune adjuvants in tumor antigen immunization. Proc Natl
Acad Sci USA. 1997;94(20):1083310837. .
90. Rothenfusser S, Tuma E, Wagner M, et al. Recent advances in
immunostimulatory CpG oligonucleotides. Curr Opin Mol Ther.
2003;5(2):98106.
91. Wei M, Chen N, Li J, et al. Polyvalent immunostimulatory nanoa-
gents with self-assembled CpG oligonucleotide-conjugated gold
nanoparticles. Angew Chem Int Ed Engl. 2012;51(5):12021206. .
92. Chen N, Wei M, Sun Y, et al. Self-assembly of poly-adenine-tailed
CpG oligonucleotide-gold nanoparticle nanoconjugates with
immunostimulatory activity. Small. 2014;10(2):368375. .
93. Zhang H. Gao X-D Nanodelivery systems for enhancing the immu-
nostimulatory effect of CpG oligodeoxynucleotides. Mater Sci Eng
C. 2017;70:935946.
94. Dykman LA, Staroverov SA, Fomin AS, et al. Gold nanoparticles as
an adjuvant: influence of size, shape, and technique of combina-
tion with CpG on antibody production. Int Immunopharmacol.
2018;54:163168.
95. Tazaki T, Tabata K, Ainai a, et al. Shape-dependent adjuvanticity of
nanoparticle-conjugated RNA adjuvants for intranasal inactivated
influenza vaccines. RSC Adv. 2018;8(30):1652716536. .
96. Wang C, Zhu W, Wang B-Z. Dual-linker gold nanoparticles as adju-
vanting carriers for multivalent display of recombinant influenza
hemagglutinin trimers and flagellin improve the immunological
responses in vivo and in vitro. Int J Nanomedicine.
2017;12:47474762.
97. Wang C, Zhu W, Luo Y, et al. Gold nanoparticles conjugating recombi-
nant influenza hemagglutinin trimers and flagellin enhanced mucosal
cellular immunity. Nanomedicine. 2018;14(4):13491360. .
98. Staroverov SA, Vidyasheva IV, Gabalov KP, et al. Immunostimulatory
effect of gold nanoparticles conjugated with transmissible gastro-
enteritis virus. Bull Exp Biol Med. 2011;151(4):436439. .
99. Staroverov SA, Volkov AA, Mezhennyy PV, et al. Prospects for the
use of spherical gold nanoparticles in immunization. Appl
Microbiol Biotechnol. 2019;103(1):437447. .
100. Sharapova NA, Kireev MN, Abramova EG, et al. Isolation of glico-
proteid from the fixed rabies virus, strain Moscow 3253ʹ, and
constructing of dot-immunoassay diagnosticum on its basis. Acta
Biomedica Scientifica. 2012;5:347350.
101. Gavrilova YK, Generalov SV, Kireev MN, et al. Development of the
scheme of obtaining antibodies to the ribonucleoprotein of atte-
nuated rabies virus. Zh Microbiol Epidemiol Immunobiol.
2019;5:38.
102. Niikura K, Matsunaga T, Suzuki T, et al. Gold nanoparticles as
a vaccine platform: influence of size and shape on immunological
responses in vitro and in vivo. ACS Nano. 2013;7(5):39263938. .
103. Stone JW, Thornburg NJ, Blum DL, et al. Gold nanorod vaccine for
respiratory syncytial virus. Nanotechnology. 2013;24(29):295102. .
104. Di Gianvincenzo P, Calvo J, Perez S, et al. Negatively charged
glyconanoparticles modulate and stabilize the secondary structures
of a gp120 V3 loop peptide: toward fully synthetic HIV vaccine
candidates. Bioconjug Chem. 2015;26(4):755765. .
105. Climent N, García I, Marradi M, et al. Loading dendritic cells with
gold nanoparticles (GNPs) bearing HIV-peptides and mannosides
enhance HIV-specific T cell responses. Nanomedicine. 2018;14
(2):339351. .
106. Lin F Development of gold nanoparticle-based antigen delivery
platform for vaccines against HIV-1 [Graduate Theses and
Dissertations]. Iowa State University. 2015.
107. Wang Y, Wang Y, Kang N, et al. Construction and immunological
evaluation of CpG-Au@HBc virus-like nanoparticles as a potential
vaccine. Nanoscale Res Lett. 2016;11(1):338. .
108. Yavuz E, Bagriacik EU Gold-based nano-adjuvants. Proc. IEEE 7-th
International Conference on Nanomaterials: Applications and
Properties, Odesa, Ukraine. 2017;04NB06-104NB06-4.
109. Wang H, Ding Y, Su S, et al. Assembly of hepatitis E vaccine by
in situgrowth of gold clusters as nano-adjuvants: an efficient way
to enhance the immune responses of vaccination. Nanoscale Horiz.
2016;1(5):394398. .
110. Olenina LV, Kolesanova EF, Gervaziev YV, et al. Preparation of
anti-peptide antibodies to the protein binding sites E2 of hepatitis
C virus with CD81. Med Immunol. 2001;3:231.
111. Versiani AF, Andrade LM, Martins EMN, et al. Gold nanoparticles
and their applications in biomedicine. Future Virol. 2016;11
(4):293309. .
112. Quach QH, Ang SK, Chu J-HJ, et al. Size-dependent neutralizing
activity of gold nanoparticle-based subunit vaccine against dengue
virus. Acta Biomater. 2018;78:224235.
113. Chen H-W, Huang C-Y, Lin S-Y, et al. Synthetic virus-like particles pre-
pared via protein corona formation enable effective vaccination in an
avian model of coronavirus infection. Biomaterials. 2016;106:111118.
114. Sekimukai H, Iwata-Yoshikawa N, Fukushi S, et al. Gold
nanoparticle-adjuvanted S protein induces a strong
antigen-specific IgG response against severe acute respiratory
syndrome-related coronavirus infection, but fails to induce protec-
tive antibodies and limit eosinophilic infiltration in lungs. Microbiol
Immunol. 2020;64(1):3351. .
115. Kareem S, Altimimi MB, Jarullah BA. Improvement of Newcastle
disease virus vaccine by using gold nanoparticles and some natural
food additives. J Thi-Qar Sci. 2017;6:5964.
116. Ding P, Zhang T, Li Y, et al. Nanoparticle orientationally dis-
played antigen epitopes improve neutralizing antibody level in
a model of porcine circovirus type 2. Int J Nanomedicine.
2017;12:52395254.
117. Li Y, Jin Q, Ding P, et al. Gold nanoparticles enhance immune
responses in mice against recombinant classical swine fever virus
E2 protein. Biotechnol Lett. 2020. doi:10.1007/s10529-020-02853-w.
118. Soliman MG, Mohamed AF, El Sayed RA, et al. Immune enhancing
potential of sphere and rod gold nanoparticles to Rift Valley fever
vaccine relative to time: in vitro study. Eur J Biomed Pharm Sci.
2017;4:529536.
119. Staroverov SA, Ermilov DN, Shcherbakov AA, et al. Generation of
antibodies to Yersinia pseudotuberculosis antigens using the colloid
gold particles as an adjuvant. Zh Mikrobiol Epidemiol Immunobiol.
2003;3:5457.
12 L. A. DYKMAN
120. Gregory AE, Williamson ED, Prior JL, et al. Conjugation of Y. pestis
F1-antigen to gold nanoparticles improves immunogenicity.
Vaccine. 2012;30(48):67776782. .
121. Kireev MN, Polunina TA, Guseva NP, et al. Studying the immuno-
genic properties of plague microbe capsule antigen F1 conjugated
with nanoparticles of colloid gold and silver. Prob Particularly
Dangerous Infect. 2008;2(2(96)):4345. .
122. Manea F, Bindoli C, Fallarini S, et al. Multivalent,
saccharide-functionalized gold nanoparticles as fully synthetic ana-
logs of type a Neisseria meningitidis Antigens. Adv Mater. 2008;20
(22):43484352. .
123. Pokharkar V, Bhumkar D, Suresh K, et al. Gold nanoparticles as
a potential carrier for transmucosal vaccine delivery. J Biomed
Nanotechnol. 2011;7(1):5759. .
124. Barhate G, Gautam M, Gairola S, et al. Quillaja saponaria extract as
mucosal adjuvant with chitosan functionalized gold nanoparticles
for mucosal vaccine delivery: stability and immunoefficiency
studies. Int J Pharm. 2013;441(12):636642. .
125. Barhate G, Gautam M, Gairola S, et al. Enhanced mucosal immune
responses against tetanus toxoid using novel delivery system com-
prised of chitosan-functionalized gold nanoparticles and botanical
adjuvant: characterization, immunogenicity, and stability
assessment. J Pharm Sci. 2014;103(11):34483456.
126. Rabiee Rudsari a, Ebrahimi F, Arefpour Torabi MA. Study of adju-
vant capability of the gold nanoparticles on the immunity of
botulinum neurotoxin serotype E in mouse. Sci J Adv Defence Sci
Technol. 2013;4:8792.
127. Safari D, Marradi M, Chiodo F, et al. Gold nanoparticles as carriers
for a synthetic Streptococcus pneumoniae type 14 conjugate
vaccine. Nanomedicine (Lond). 2012;5(5):651662. .
128. Vetro M, Safari D, Fallarini S, et al. Preparation and immunogenicity
of gold glyco-nanoparticles as antipneumococcal vaccine model.
Nanomedicine (Lond). 2017;12(1):1323. .
129. Staroverov SA, Dykman LA. Use of gold nanoparticles for the pre-
paration of antibodies to tuberculin, the immunoassay of myco-
bacteria, and animal vaccination. Nanotechnol Russia. 2013;8
(1112):816822.
130. Burygin GL, Abronina PI, Podvalnyy NM, et al. Preparation and
in vivo evaluation of glyco-gold nanoparticles carrying synthetic
mycobacterial hexaarabinofuranoside. Beilstein J Nanotechnol.
2020;11:480493.
131. Gao W, Fang RH, Thamphiwatana S, et al. Modulating antibacterial
immunity via bacterial membrane-coated nanoparticles. Nano Lett.
2015;15(2):14031409. .
132. Sanchez-Villamil JI, Tapia D, Torres AG. Development of a gold
nanoparticle vaccine against enterohemorrhagic Escherichia coli
O157:H7. mBio. 2019;10(4):e0186919.
133. Rodriguez-Del Rio E, Marradi M, Calderon-Gonzalez R, et al.
a gold glyco-nanoparticle carrying a listeriolysin O peptide and
formulated with Advax
TM
delta inulin adjuvant induces robust
T-cell protection against listeria infection. Vaccine. 2015;33
(12):14651473. .
134. Calderón-Gonzalez R, Terán-Navarro H, Frande-Cabanes E, et al.
Pregnancy vaccination with gold glyco-nanoparticles carrying Listeria
monocytogenes peptides protects against listeriosis and brain- and
cutaneous-associated morbidities. Nanomaterials. 2016;6(8):151. .
135. Gregory AE, Judy BM, Qazi O, et al. a gold nanoparticle-linked
glycoconjugate vaccine against Burkholderia mallei.
Nanomedicine. 2015;11(2):447456. .
136. Torres AG, Gregory AE, Hatcher CL, et al. Protection of non-human
primates against glanders with a gold nanoparticle glycoconjugate
vaccine. Vaccine. 2015;33(5):686692. .
137. Muruato LA, Tapia D, Hatcher CL, et al. Use of reverse vaccinology
in the design and construction of nanoglycoconjugate vaccines
against Burkholderia pseudomallei. Clin Vaccine Immunol. 2017;24
(11):e0020617. .
138. Dakterzada F, Mohabati Mobarez a, Habibi Roudkenar M, et al.
Induction of humoral immune response against Pseudomonas aer-
uginosa flagellin
(1-161)
using gold nanoparticles as an adjuvant.
Vaccine. 2016;34(12):14721479. .
139. Srivastva P Evaluation of a candidate GNP-APRV2 footrot nanovac-
cine in mice. CSK-Himachal Pradesh Agricultural University
[Graduate Theses and Dissertations] 2017.
140. Dykman LA, Volokh OA, Kuznetsova EM, et al. Immunogenicity of
conjugates of protective antigen complexes of tularemia microbe
with gold nanoparticles. Nanotechnol Russ. 2018;13(78):384392. .
141. Van Schaik EJ, Gregory AE, Audette GF, et al. Nanovaccines against
intracellular pathogens using Coxiella burnetii as a model organism.
Immune aspects of biopharmaceuticals and nanomedicines.
Singapore: Pan Stanford Publishing; 2018.
142. Dykman LA, Volokh OA, Gromova OV, et al. Obtaining and char-
acteristic of antibodies to Vibrio cholerae protective antigens con-
jugated with gold nanoparticles. Dokl Biochem Biophys.
2020;490:19-12.
143. Parween S, Gupta PK, Chauhan VS. Induction of humoral immune
response against PfMSP-119 and PvMSP-119 using gold nanopar-
ticles along with alum. Vaccine. 2011;29(13):24512460.
144. Kumar R, Ray PC, Datta D, et al. Nanovaccines for malaria using
Plasmodium falciparum antigen Pfs25 attached gold nanoparticles.
Vaccine. 2015;33(39):50645071. .
145. Bulashev AK, Borovikov SN, Serikova SS, et al. Development of an
ELISA using anti-idiotypic antibody for diagnosis of opisthorchiasis.
Folia Parasitol. 2016;63:025.
146. Assis NRG, Caires AJ, Figueiredo BC, et al. The use of gold nanorods
as a new vaccine platform against schistosomiasis. J Control
Release. 2018;275:4052.
147. Khlebtsov NG, Dykman LA. Biodistribution and toxicity of engi-
neered gold nanoparticles: a review of in vitro and in vivo studies.
Chem Soc Rev. 2011;40(3):16471671.
148. Vijayan V, Mohapatra a, Uthaman S, et al. Recent advances in
nanovaccines using biomimetic immunomodulatory materials.
Pharmaceutics. 2019;11:534.
This review summarizes the recent advances in the design of
nanovaccines using biomimetic immunomodulatory materials.
EXPERT REVIEW OF VACCINES 13
... 16 PepGNP-Dengue is a candidate vaccine that has been designed to elicit a specific cytotoxic CD8+ T cell response, without inducing a humoral one. It is composed of synthetic T cell-selective multi-valent DENV peptides carried on ultrasmall gold nanoparticles (GNPs), chosen for their immunogenic properties, 17 layered with carbohydrates mimicking a bacterial pattern ('self-adjuvanting' vaccine) and designed to mimic the process of antigen presentation during natural infection. 18 Their intradermal administration aims to partially mimic a natural infection from a mosquito bite, target skin antigen-presenting cells (APCs), and induce a protective local CD8+ response. ...
Article
Full-text available
Background Vaccines that minimize the risk of vaccine-induced antibody-dependent enhancement and severe dengue are needed to address the global health threat posed by dengue. This study assessed the safety and immunogenicity of a gold nanoparticle (GNP)-based, multi-valent, synthetic peptide dengue vaccine candidate (PepGNP-Dengue), designed to provide protective CD8+ T cell immunity, without inducing antibodies. Methods In this randomized, double-blind, vehicle-controlled, phase 1 trial (NCT04935801), healthy naïve individuals aged 18–45 years recruited at the Centre for primary care and public health, Lausanne, Switzerland, were randomly assigned to receive PepGNP-Dengue or comparator (GNP without peptides [vehicle-GNP]). Randomization was stratified into four groups (low dose [LD] and high dose [HD]), allocation was double-blind from participants and investigators. Two doses were administered by intradermal microneedle injection 21 days apart. Primary outcome was safety, secondary outcome immunogenicity. Analysis was by intention-to-treat for safety, intention-to-treat and per protocol for immunogenicity. Findings 26 participants were enrolled (August–September 2021) to receive PepGNP-Dengue (LD or HD, n = 10 each) or vehicle-GNP (LD or HD, n = 3 each). No vaccine-related serious adverse events occurred. Most (90%) related adverse events were mild; injection site pain and transient discoloration were most frequently reported. Injection site erythema occurred in 58% of participants. As expected, PepGNP-Dengue did not elicit anti-DENV antibodies of significance. Significant increases were observed in specific CD8+ T cells and dengue dextramer+ memory cell subsets in the LD PepGNP-Dengue but not in the HD PepGNP-Dengue or vehicle-GNP groups, specifically PepGNP-activated CD137+CD69+CD8+ T cells (day 90, +0.0318%, 95% CI: 0.0088–0.1723, p = 0.046), differentiated effector memory (TemRA) and central memory (Tcm) CD8+ T cells (day 35, +0.8/10⁵ CD8+, 95% CI: 0.19–5.13, p = 0.014 and +1.34/10⁵ CD8+, 95% CI: 0.1–7.34, p = 0.024, respectively). Interpretation Results provide proof of concept that a synthetic nanoparticle-based peptide vaccine can successfully induce virus-specific CD8+ T cells. The favourable safety profile and cellular responses observed support further development of PepGNP-Dengue. Funding Emergex Vaccines Holding Limited.
... The AuNPs lead to paralysis and then the death of worms. The authors attributed this to changes in the enzymatic activity of the worms [26] in addition to its effect on a group of protozoa [27][28][29] and the morphological changes in the protoscolices. The morphological distortions of the protoscolices exposed to gold nanoparticles were attributed to the loss of the plasma membrane function or defects in the equilibrium of ions on both sides of the membrane plasma. ...
Article
Many scolicidal agents have been used to destroy fertile protoscolices, but these scolicidal agents have side effects, highlighting the need for research on effective and non-toxic replacement scolicidal agents. Gold nanoparticles are biocompatible and non-toxic. The current study examined the effects of AuNPs in killing the protoscolices of Echinococcus granulosus in vitro using eosin staining. The protoscolices were treated with 0.2, 0.4, 0.8, or 1.0 mg/mL of gold nanoparticles (AuNPs) for 15, 30, 45, or 60 minutes. A concentration of 1.0 mg/mL was the most efficient in killing the protoscolices after 60 minutes exposure, reaching 96%, followed by 0.8 mg/mL (84.5%), whereas 0.4 and 0.2 mg/mL of AuNPs achieved a death rate of 76.8% and 68.5%, respectively. The loss of the protoscolices was lower at shorter exposure times with the same concentration of AuNPs and increased as the AuNP concentration was increased at the same exposure time. Significant differences were found between the different groups compared to the control group.
Article
The knowledge base in the field of vaccination research and development has greatly improved. In the present era, utilizing a novel vaccine design and optimization strategy improves the vaccination efficiency and activity. In this regard, a novel drug delivery system produces nanosized, vaccine-loaded carrier molecules, which is called Nanovaccine. The advancement in nanovaccine and improved technology comes under preclinical and clinical trials, whereas different routes of administration strategy are applied against infectious diseases. The nanovaccine has high efficiency and immunogenicity compared to the traditional vaccine. Its long-lasting effect reduces the booster dose as well. One of the factors like routes of administration affects the efficiency of nanovaccine. The parenteral and mucosal vaccination is a traditional administration approach. In order to increase the safety and effectiveness of vaccines, innovative administration methods such as needless injection and polymeric formulations are now being developed. The presented paper shows a mechanism involved in nanovaccine delivery, and a method of administration via a different route, to learn more about their possible effects on immunogenicity, effectiveness, safety, sustained release and dose frequency reduction. The various advanced vaccine delivery methods, with special emphasis on its industrial and regulatory requirements for the higher production of nanovaccine, are also discussed.
Chapter
Infectious diseases have been a significant cause of morbidity and mortality throughout human history. Additionally, superbugs that do not respond to most approved drugs have emerged as a result of the constant rise of antibiotic resistance. Vaccines represent one of the most effective public health initiatives globally. Particularly, during the coronavirus disease 2019 (COVID-19) pandemic, the successful clinical application of the lipid nanoparticle-based mRNA vaccines has highlighted the potential of nanotechnology for vaccine development. Nanovaccines are supposed to be superior to conventional vaccines in terms of lymph node accumulation, antigen assembly, as well as antigen presentation. They also possess the unique characteristic of pathogen biomimicry as a result of the well-organized combination of several immune factors. In the vaccination market, synthetic nanoscale platforms like liposomes and inorganic nano- and microparticles offer many advantages, but they frequently need multiple doses and the addition of synthetic adjuvants like aluminum hydroxide. Contrarily, biologically derived nanoparticles containing native pathogen-associated molecular patterns (PAMPs) can reduce the requirement for artificial adjuvants. Biodegradability, biocompatibility, and the ability to self-assemble are just a few of the extra beneficial qualities that can be built into biological nanoparticles. This makes it easy to scale up production from bench top to large-scale. In this review, we highlight the various types of materials, their advantages and disadvantages, as well as future views while summarizing recent developments in vaccine nanotechnology for the prevention of infectious diseases.
Article
Full-text available
Viruses have threatened human lives for decades, causing both chronic and acute infections accompanied by mild to severe symptoms. During the long journey of confrontation, humans have developed intricate immune systems to combat viral infections. In parallel, vaccines are invented and administrated to induce strong protective immunity while generating few adverse effects. With advancements in biochemistry and biophysics, different kinds of vaccines in versatile forms have been utilized to prevent virus infections, although the safety and effectiveness of these vaccines are diverse from each other. In this review, we first listed and described major pathogenic viruses and their pandemics that emerged in the past two centuries. Furthermore, we summarized the distinctive characteristics of different antiviral vaccines and adjuvants. Subsequently, in the main body, we reviewed recent advances of nanoparticles in the development of next-generation vaccines against influenza viruses, coronaviruses, HIV, hepatitis viruses, and many others. Specifically, we described applications of self-assembling protein polymers, virus-like particles, nano-carriers, and nano-adjuvants in antiviral vaccines. We also discussed the therapeutic potential of nanoparticles in developing safe and effective mucosal vaccines. Nanoparticle techniques could be promising platforms for developing broad-spectrum, preventive, or therapeutic antiviral vaccines.
Article
Full-text available
The impact of vaccination on the world's population is difficult to calculate. For developing different types of vaccines, adjuvants are substances added to vaccines to increase the magnitude and durability of the immune response and the effectiveness of the vaccine. This work explores the potential use of spherical gold nanoparticles (AuNPs) as adjuvants. Thus, we employed docking techniques and molecular mechanics to describe how a AuNP 7.0 nm in diameter interacts with cell signaling pathway proteins. Initially, we used X-ray crystallization data of the proteins ovalbumin, glutathione, LC3, TLR4, ASC PYCARD, PI3K, and NF-Kβ to study the adsorption with an AuNP through molecular docking. Therefore, interaction energies were obtained for the AuNP complexes and individual proteins, as well as the AuNP and OVA complex (AuNP@OVA) with each cellular protein, respectively. Results showed that AuNPs had the highest affinity for OVA individually, followed by glutathione, ASC PYCARD domain, LC3, PI3K, NF-Kβ, and TLR4. Furthermore, when evaluating the AuNP@OVA complex, glutathione showed a greater affinity with more potent interaction energy when compared to the other studied systems.
Article
Full-text available
The development of nanotechnology for the treatment of parasitic diseases is still in its infancy. However, it is expected that this new field can provide a solution to parasitic diseases and compensate for the lack of vaccines to prevent them. It can also provide new treatment options for parasitic diseases resistant to current treatments. Nanomaterials have been developed for antibacterial and anticancer therapies. However, it is important to determine their antiparasitic potential due to the wide variety of their physicochemical properties. When designing metallic nanoparticles (MeNPs) and specialized nanosystems like MeNPs encapsulated within a drug shell, it is essential to consider several key physicochemical properties. Shape, size, surface charge, and type of surfactant control are some of these physicochemical properties. In addition to interacting with parasite cells’ target molecules, shell molecules are also important. By developing antiparasitic drugs using nanotechnology and nanomaterials for diagnostics, new and effective methods of treatment and diagnostic tools for poultry diseases are expected to be available in the future to enhance poultry disease prevention and reduce morbidity and mortality rates.
Article
Full-text available
Monoclonal antibody specific for an epitope of cretory-secretory antigen protein of Opisthorchis felineus (Rivolta, 1884) (Trematoda: Opisthorchiidae) with a molecular weight of 28 kDa was used in a sandwich enzyme-linked immunosorbent assay (ELI-SA) for immobilisation of liver fluke specific antigen to the solid phase. Examination of human sera by this ELISA compared with commercial assays demonstrated that the monoclonal antibody epitope is located within this significant parasite protein. Anti-idiotypic antibody specific for the paratope of this monoclonal antibody was obtained by a hybridoma technique. Mimicking an epitope of excre-tory-secretory antigen of O. felineus, it had the capacity to bind specific antibody and elicit an antibody response. The value of anti-id-iotypic antibody as a substitute for the liver fluke antigen was tested by ELISA using serum samples of infected dogs. Anti-idiotypic antibody proved to be of value in both an indirect-ELISA and a competitive-ELISA for diagnosis of opisthorchiasis. Mature trematodes were isolated from all infected animals. The faecal egg counts were negative in dogs with a relatively small number of parasites, despite finding antibodies in serum by ELISA. Substitution of parasite antigen with anti-idiotype avoids the use of experimental animals and also reduces time-consuming steps of antigen preparation.
Article
Full-text available
Gold nanoparticle conjugates with Vibrio cholerae antigens were synthesized. The animals were immunized with the obtained conjugates. Rabbit polyclonal antibodies to the antigens were obtained, which showed high specific activity. On the model of white laboratory mice, the protective activity of conjugates of cholera antigens with nanoparticles during infection of vaccinated animals was evaluated using a commercial vaccine as a control. It was shown that in terms of immunogenicity, the created prototypes of cholera vaccine using gold nanoparticles as a carrier and adjuvant complied with the production regulations for the Russian national cholera chemical vaccine.
Article
Full-text available
A number of bacterial glycans are specific markers for the detection and the serological identification of microorganisms and are also widely used as antigenic components of vaccines. The use of gold nanoparticles as carriers for glyco-epitopes is becoming an important alternative to the traditional conjugation with proteins and synthetic polymers. In this study, we aimed to prepare and evaluate in vivo glyco-gold nanoparticles (glyco-GNPs) bearing the terminal-branched hexaarabinofuranoside fragment (Ara 6 ) of arabinan domains of lipoarabinomannan and arabinogalactan, which are principal polysaccharides of the cell wall of Mycobacterium tuberculosis , the causative agent of tuberculosis. In particular, we were interested whether the antibodies generated against Ara 6 -GNPs would recognize the natural saccharides on the cell surface of different mycobacterial strains. Two synthetic Ara 6 glycosides with amino-functionalized spacer aglycons differing in length and hydrophilicity were directly conjugated with spherical gold nanoparticles ( d = 15 nm) to give two sets of glyco-GNPs, which were used for the immunization of rabbits. Dot assays revealed cross-reactions between the two obtained antisera with the hexaarabinofuranoside and the 2-aminoethyl aglycon used for the preparation of glyco-GNPs. Both antisera contained high titers of antibodies specific for Mycobacteria as shown by enzyme-linked immunosorbent assay using M. bovis and M. smegmatis cells as antigens while there was only a weak response to M. phlei cells and no interaction with E. coli cells. The results obtained suggest that glyco-GNPs are promising agents for the generation of anti-mycobacterial antibodies.
Article
Full-text available
Objective To create a novel subunit vaccine that used AuNPs as carriers to enhance immune responses in mice against recombinant classical swine fever virus E2 protein (CSFV E2).ResultsGold nanoparticles (AuNPs) were successfully coupled to the E2 protein and formed stable particle complexes called E2 conjugated AuNPs (E2–AuNPs). In vitro studies have shown that the E2–AuNPs complex has the same immunogenicity as the E2 protein, and AuNPs can promote the phagocytosis of the E2 protein by antigen-presenting cells (APCs). In vivo results of BALB/c mice showed that the antibody levels, lymphocyte proliferation index, IFN-γ and IL-10 cytokines induced by E2–AuNPs were relatively higher than those of E2 or AuNPs group.Conclusions This finding demonstrated the potential of using AuNPs as a carrier to enhance the body's immune response for developing CSFV subunit vaccines. This model also contributes to the development of other flavivirus subunit vaccines, such as hepatitis C virus (HCV) and bovine viral diarrhea virus (BVDV).
Article
Full-text available
Aim . Isolation of ribonucleoprotein (RNP) of an attenuated rabies virus, develop schemes for immunizing animals with RNP-based preparations and determine the most effective scheme that allows obtaining serum with a high antibody titer to the RNP. Materials and methods . We used the transplantable cell line Vero, the strain rabies virus «Moscow 3253 Vero», adapted for reproduction on Vero, rabbits of the chinchilla breed. In order to obtain serums containing antibodies to the RNP of the rabies virus, experimental schemes have been proposed for immunizing animals with RNP, including with adjuvants: polyoxidonium and colloidal gold. The dynamics of the accumulation of antibodies to the RNP of the rabies virus in the blood serum of experimental animals was studied by dot-immunoassay. Results . The target component (RNP of the rabies virus) was isolated directly from the cytoplasm of the Vero cell culture infected with the rabies virus according to the modified M. Dastkhosh (2014) method, lyophilized and used in the development of preparations for immunizing experimental animals. In the study of the dynamics of the formation of antibodies to RNP of the rabies virus by the method of dot-immunoassay, the effectiveness of an adjuvant is established — colloidal gold nanoparticles ranging in size from 15 to 17 nm, the use of which makes it possible to increase the antibody titer by 2 times. Conclusion . The results obtained are of interest for further research related to the design of diagnostic products and the development of methodological techniques using such preparations.
Article
Full-text available
The spike (S) protein of coronavirus, which binds to cellular receptors and mediates membrane fusion for cell entry, is a candidate vaccine target for blocking coronavirus infection. However, some animal studies have suggested that inadequate immunization against severe acute respiratory syndrome coronavirus (SARS‐CoV) induces a lung eosinophilic immunopathology upon infection. In this study, we evaluated two kinds of vaccine adjuvants for use with recombinant S protein: gold nanoparticles (AuNPs), which are expected to function as both an antigen carrier and an adjuvant in immunization; and Toll‐like receptor (TLR) agonists, which have been previously shown to be an effective adjuvant in a ultraviolet‐inactivated SARS‐CoV vaccine. All of the mice immunized with more than 0.5 µg S protein without adjuvant escaped from SARS after infection with mouse‐adapted SARS‐CoV; however, eosinophilic infiltrations were observed in the lungs of almost all of the immunized mice. The AuNP‐adjuvanted protein induced a strong IgG response but failed to improve vaccine efficacy or reduce eosinophilic infiltration because of highly allergic inflammatory responses. Whereas similar virus titers were observed in the control animals and the animals immunized with S protein with or without AuNPs, type 1 interferon and pro‐inflammatory responses were moderate in the mice treated with S protein with and without AuNPs. On the other hand, the TLR agonist‐adjuvanted vaccine induced highly protective antibodies without eosinophilic infiltrations, as well as Th1/17 cytokine responses. The findings of this study will support the development of vaccines against severe pneumonia‐associated coronaviruses. This article is protected by copyright. All rights reserved.
Article
Full-text available
The development of vaccines plays a vital role in the effective control of several fatal diseases. However, effective prophylactic and therapeutic vaccines have yet to be developed for completely curing deadly diseases, such as cancer, malaria, HIV, and serious microbial infections. Thus, suitable vaccine candidates need to be designed to elicit appropriate immune responses. Nanotechnology has been found to play a unique role in the design of vaccines, providing them with enhanced specificity and potency. Nano-scaled materials, such as virus-like particles, liposomes, polymeric nanoparticles (NPs), and protein NPs, have received considerable attention over the past decade as potential carriers for the delivery of vaccine antigens and adjuvants, due to their beneficial advantages, like improved antigen stability, targeted delivery, and long-time release, for which antigens/adjuvants are either encapsulated within, or decorated on, the NP surface. Flexibility in the design of nanomedicine allows for the programming of immune responses, thereby addressing the many challenges encountered in vaccine development. Biomimetic NPs have emerged as innovative natural mimicking biosystems that can be used for a wide range of biomedical applications. In this review, we discuss the recent advances in biomimetic nanovaccines, and their use in anti-bacterial therapy, anti-HIV therapy, anti-malarial therapy, anti-melittin therapy, and anti-tumor immunity.
Article
Full-text available
Background: Influenza virus infection causes significant morbidity and mortality worldwide. Humans fail to make a universally protective memory response to influenza A because of high mutation rates in the immune-dominant influenza epitopes. We seek the development of a universal influenza A vaccine. The extracellular domain of the M2-ion channel (M2e) is an ideal antigenic target, as it is highly conserved, has a low mutation rate, and is essential for viral entry and replication. Considering the potential of a universal influenza vaccine for lifelong protection, we aimed to examine this potential using a recently published gold nanoparticle M2e vaccine with CpG as an adjuvant (AuNP-M2e + sCpG). Intranasal vaccination induces an M2e-specific memory response, which is protective against lethal infection with H1N1, H3N2, and H5N1 serotypes, in young BALB/c mice. Protection with AuNP-M2e + sCpG has been published up to 8 months after vaccination. However, the highest risk population during most influenza seasons is adults over 65 years old. Additionally, the efficacy of many vaccines decrease after aging and requiring booster vaccinations to remain effective. Results: To determine if the AuNP-M2e + sCpG vaccine is a viable option as a universal vaccination capable of protection through geriatric age, we tested if the AuNP-M2e + sCpG vaccination loses efficacy after aging mice to geriatric age (over 18 months). Our data shows that mice aged 15 months after vaccination (~ 18-21 months old) retain significant M2e-specific antibody titers in total IgG, IgG1, IgG2a, and IgG2b. These mice are significantly protected from lethal influenza challenge (H1N1, 8.3 PFU). Further, these antibody titers increase upon infection with influenza A and remain elevated for 3 months, suggesting the elderly mice retain effective M2e-specific memory B cells. Conclusions: Our results demonstrate that protective M2e-specific memory in mice developed at a young age can persist until geriatric age. Additionally, this memory is protective and M2e-specific B cells produced by vaccination with AuNP-M2e + sCpG are maintained and functional. If the results of this study persist in humans, they suggest that a universal influenza A vaccine could be administered early in life and maintain lifelong protection into geriatric age.
Article
Full-text available
Here we exploit the natural properties of a synthetic nanoparticle (NP) scaffold as a subunit vaccine against enterohemorrhagic Escherichia coli (EHEC). Two EHEC-specific immunogenic antigens, namely, LomW and EscC, either alone or in combination, were covalently linked on the surface of gold nanoparticles (AuNPs) and used to immunize mice prior to challenge with EHEC O157:H7 strain 86-24. LomW is a putative outer membrane protein encoded in bacteriophage BP-933W, while EscC is a structural type III secretion system protein which forms a ring in the outer membrane. The resulting AuNP preparations, AuNP-LomW and AuNP-EscC, showed that the nanoparticles were able to incorporate the antigens, forming stable formulations that retained robust immunogenicity in vivo after subcutaneous immunization. When administered subcutaneously, AuNP-LomW or AuNP-EscC or a combination containing equivalent amounts of both candidates resulted in higher IgG titers in serum and secretory IgA titers in feces. The serum IgG titers correlated with a significant reduction in EHEC intestinal colonization after 3 days postinoculation. In addition, we showed that serum from antigen-coated AuNP-immunized mice resulted in a reduction of adherence to human intestinal epithelial cells for EHEC, as well as for two other E. coli pathotypes (enteropathogenic E. coli [EPEC], encoding EscC, and enteroaggregative E. coli [EAEC], encoding LomW). Further, the serum had antigen-specific bactericidal properties, engaging the classical complement pathway. Overall, our results demonstrate the immunogenicity and stability of a novel nanovaccine against EHEC. These results also strengthen the prospect of development of a synthetic nanoparticle vaccine conjugated to E. coli antigens as a promising platform against other enteric pathogens.
Article
Vaccinations are the most effective preventive methods against infectious diseases and represent one of the most relevant successes of medicine. Vaccine development is constantly evolving; therefore, the number of vaccine candidates is progressively increasing. However, most of new potential vaccines are characterized by a lower immunogenicity, with the inability to stimulate powerful and long-lasting immune responses. Hence, to get modern and effective vaccines, we need adjuvants and innovative delivery systems that increase their immunogenicity. The use of nanotechnology in vaccinology is providing the opportunity to contrast these difficulties and develop effective vaccines. Particularly, nanoparticles used as vehicles of vaccine components, are able to increase the host's immune responses and, due to their size, to reach specific cellular districts. To date, a certain number of nanovaccines has been approved for human health and many are studied in clinical or pre-clinical trials. There are several types of nanoparticles considered as possible delivers of vaccine antigens. These nanoparticles-based synthetic delivery systems, in the size range of 20-200 nm, protect antigen from degradation, enhance its presentation and facilitate its uptake by professional antigen-presenting cells. Virus-like particles, self-assembled proteins, micelles, liposomes, inorganic nanoparticles, and polymers are the most studied of these systems. In this review, we provide a general overview of different types, methods of synthesis, characterizations, properties and applications of nanoparticles in vaccine production.