Next Article in Journal
Antibiotic Resistance and Biofilm-Forming Ability in Enterococcal Isolates from Red Meat and Poultry Preparations
Next Article in Special Issue
Spread of Influenza Viruses in Poland and Neighboring Countries in Seasonal Terms
Previous Article in Journal
NcPuf1 Is a Key Virulence Factor in Neospora caninum
Previous Article in Special Issue
Organization of the Influenza A Virus Genomic RNA in the Viral Replication Cycle—Structure, Interactions, and Implications for the Emergence of New Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anti-Influenza Strategies Based on Nanoparticle Applications

by
Klaudia Wieczorek
1,2,
Barbara Szutkowska
1 and
Elzbieta Kierzek
1,*
1
Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
2
NanoBioMedical Centre, Adam Mickiewicz University, 61-704 Poznan, Poland
*
Author to whom correspondence should be addressed.
Pathogens 2020, 9(12), 1020; https://doi.org/10.3390/pathogens9121020
Submission received: 30 October 2020 / Revised: 30 November 2020 / Accepted: 1 December 2020 / Published: 3 December 2020
(This article belongs to the Special Issue Advance in Influenza A Virus)

Abstract

:
Influenza virus has the potential for being one of the deadliest viruses, as we know from the pandemic’s history. The influenza virus, with a constantly mutating genome, is becoming resistant to existing antiviral drugs and vaccines. For that reason, there is an urgent need for developing new therapeutics and therapies. Despite the fact that a new generation of universal vaccines or anti-influenza drugs are being developed, the perfect remedy has still not been found. In this review, various strategies for using nanoparticles (NPs) to defeat influenza virus infections are presented. Several categories of NP applications are highlighted: NPs as immuno-inducing vaccines, NPs used in gene silencing approaches, bare NPs influencing influenza virus life cycle and the use of NPs for drug delivery. This rapidly growing field of anti-influenza methods based on nanotechnology is very promising. Although profound research must be conducted to fully understand and control the potential side effects of the new generation of antivirals, the presented and discussed studies show that nanotechnology methods can effectively induce the immune responses or inhibit influenza virus activity both in vitro and in vivo. Moreover, with its variety of modification possibilities, nanotechnology has great potential for applications and may be helpful not only in anti-influenza but also in the general antiviral approaches.

1. Introduction

Influenza virus is a member of the Orthomyxovirade family of viruses [1]. Four influenza viruses can be distinguished: A, B, C (known for being human-infectious) and D (so far unconfirmed for being human-threatening) [2]. Among these, influenza A virus (IAV) and influenza B virus (IBV) have the potential to cause annual epidemics, known also as a seasonal flu. Moreover, IAV has a zoonotic potential, which means it can be easily transmitted from animals into humans. As a consequence, it has strong potential to evolve into a deadly pandemic strain and therefore is considered as one of the most human-threatening viruses [3].
The IAV genome consists of eight single-stranded, negative-sense RNAs ((-)RNA or vRNA) segments. RNAs along with viral proteins form eight viral ribonucleoprotein (vRNP) complexes. Each vRNP consists of the viral RNA segment bound to viral heterotrimeric RNA-dependent RNA polymerase (RdRp) and coated with nucleoproteins (NP). The influenza virus’ life cycle is entirely dependent on both RNA and viral polymerase. vRNA is a template for messenger RNA (mRNA) and complementary RNA (cRNA). The cRNA serves as a template for vRNA replication. The eight segments encode at least 16 proteins, including through alternative splicing and translation initiation [4]. These proteins include nucleoprotein, hemagglutinin (HA), neuraminidase (NA), matrix protein 1 and 2 (M1, M2), nuclear export protein (NEP), subunits of RNA-dependent RNA polymerase complex (PB1, PB2 and PA), as well as two non-structural proteins: PB1-F2 and NS1 (Figure 1).
The IAV genome undergoes two phenomena, antigenic shift and drift, that lead to the occurrence of new strains [5,6,7,8]. Different strains of the IAVs circulate annually as seasonal flus, and some of them can evolve into epidemic or pandemic strains [9]. Pandemics, written on the pages of modern history, showed that the influenza virus can be one of the most deadliest viruses [10]. The current SARS-CoV-2 pandemics is taking its toll around the world [11,12,13], recalling how deadly and severe for human health and devastating for the economy and daily life RNA viruses are. Moreover, many co-infections of influenza virus and SARS-CoV-2 have been observed worldwide and it seems that co-infections tends to strengthen the negative symptoms of the infection [14,15,16,17]. This broadens the already dangerous effects of influenza spreading and illness.
Vaccination is considered to be the most effective influenza prevention strategy [17]. The composition of the influenza vaccines is carefully estimated and published by the WHO every year, based on previous circulating influenza strains [18]. The vaccination’s effectiveness, monitored and published by the CDC, has ranged over the years from only 10% (season 2004–2005) to merely 60% (2010–2011) [19]. It is worth noting that the effectiveness varies between the particular subtypes, and in the previous 2018/19 season, it was significantly higher in H3N2 strains, as reported by Kissling et al. [20,21]. Although new generations of the universal vaccines have been developed, the perfect one has still not been found [22].
On the other hand, the constantly evolving genome of the influenza virus makes it resistant to existing antivirals. For that reason, there is an urgent need for developing new therapeutics and therapies. Most anti-influenza drugs target the viral proteins, so their effectiveness could be changed along with future viral mutations and might be only strain-specific effective [23,24]. Taking this together, the pandemic potential of the influenza virus should be studied even more closely. Many studies have applied different targets in the influenza virus, such as viral RNA or proteins, disrupting different viral processes including viral entry, replication or splicing as well as influencing cellular immune responses [25,26,27,28]. Developing new prevention methods and an effective treatment against the virus, based on up-to-date knowledge, is crucial.
Nanotechnology has been with us for a very long time, although its use in science and medicine has grown rapidly over the last few years [29,30]. According to International Union of Pure and Applied Chemistry’s (IUPAC) definition recommendations, nanoparticles (NPs) are particles of any shape with approximately one of the dimensions in the 1 × 10−9 to 1 × 10−7 m range [31]. Usage of the NPs in life sciences gave life to new fields of science—nanomedicine and nanobiology [32]. These scientific fields complement each other and have opened up a broad range of applications in modern medicine—from diagnostic, molecular therapies, drugs, and their delivery to imaging biological structures on a molecular scale [33,34,35,36,37]. The possibility of choosing any size, morphology, porosity, and the possibility of any surface functionalization make NPs very attractive. It has been reported that the choice of particular NPs’ parameters is crucial while targeting specific cells or tissues is a major determinant to mitigate cytotoxic effects [38,39].
As the nanotechnology field grows, its applications in anti-influenza therapies and prevention methods are expanding. Intensive research on nanotechnology-based antivirals and their influence on the viral life cycle and cell itself broadens the knowledge concerning virus biology, and contributes to the development of nanomedicines. This review focuses on nanotechnology trends towards the facilitation of existing anti-influenza strategies and new methodologies applying NPs against influenza viruses.

2. Influenza Virus Inhibition Strategies Based on the Use of Nanoparticles

The nanosize and numerous possibilities of NPs’ functionalization make them excellent carriers of various antiviral agents. Here we present different strategies of using NPs to fight influenza virus infection: NPs as immunity-inducing vaccines, NPs used in gene silencing approaches, the use of NPs for drug delivery, and bare NPs which also exhibit anti-influenza properties (Table 1).

2.1. Nanotechnology in Vaccination Strategies

The IAV is highly variable due to genetic drift and shift, and therefore the organism does not gain life-long immunity against the virus [5]. For that reason, new vaccine formulation must be developed and manufactured annually, which is a labor-extensive, time-consuming, and expensive process [69,70]. The WHO’s recommendation for northern and southern hemisphere influenza vaccine composition is predicted and introduced every year independently [71,72]. However, the vaccine’s effectiveness is variable between seasons and depends on prediction accuracy [73]. Hence, it is important to create new and faster methods for vaccine production to increase vaccination efficiency. Vaccination strategies rely on the delivery of the viral antigens immobilized on NPs (Figure 2 (A)), mRNA fragments coding the antigens (Figure 2 (B)) or inactivated virus (Figure 2 (C)) encapsulated in NPs into the cells as it is taking place during the viral infection (Table 2).

2.1.1. Viral Antigen Vaccination Strategy Based on NPs Delivery

Commercial vaccines usually contain inactivated subunits or live attenuated influenza to induce immunity and produce antibodies (Abs) against HA and NA. However, vaccine production is a long process and antigenic drift can lead to a decrease in the vaccination’s effectiveness. Hence, it is very important to create new, faster to produce, and more effective vaccination methods.
When the viral infection occurs for the first time, the immune system is stimulated to produce specific Abs to gain immunity for the next viral encounter. Two types of Abs are produced during the infection: neutralizing Abs directly inhibiting the virion cell entry by interacting near the receptor binding site (RBS) of HA and non-neutralizing Abs that act indirectly by supporting other mechanisms such as phagocytosis, activating, complementing, and promoting antibody-dependent cellular cytotoxicity (Figure 2) [74]. Anti-influenza Abs can be further divided into Abs against external and internal proteins. There are three types of anti-external protein Abs: anti-HA, anti-NA and anti-M2. Moreover, organism can produce Abs against internal proteins: anti-nucleoprotein, anti-M1 and anti-PB1-F2 [74]. The viral antigen strategy relies on delivery of the viral antigens into the cells to produce anti-external Abs—anti-NA, anti-HA and anti-M2—as takes place during the viral infection.

Hemagglutinin Protein as a Viral Antigen in Vaccine Based on Nanoparticles

Hemagglutinin is a viral surface glycoprotein engaged in initiating the infection by binding the α-sialic acid residues presented on the hosts’ cell membrane [75]. It is a major determinant of pathogenicity and the main target of neutralizing Abs. Binding the HA to the sialic acid presented on the host’s cell surface is followed by the fusion of the viral envelope with the endosomal membrane and viral entry, allowing delivery of the virus to the cytoplasm [69,76].
Wang et al. designed dual-linker Au NPs conjugated with both recombinant trimetric HA from influenza A/Aichi/2/68 (H3N2) and flagellin (FliC) as an adjuvant [40]. FliC is a bacterial flagella protein, which can prompt the host’s innate signaling pathways through recognition by a TLR5-expressed antigen. The results of the in vivo study performed in mice show that levels of antigen-specific Abs are significantly increased after the nanovaccination. In the in vitro study, the Au NPs-HA/FliC conjugate was capable to trigger the TLR5 signaling pathways. As a result, antigenic epitopes were effectively processed and cross-presented by the antigen-presenting cells to induce the T cell-mediated immunity. In continuation of this study, Wang et al. investigated co-delivery of separately functionalized Au NPs-FliC and Au NPs-HA in an in vivo mice model. Although the IgG Abs levels were similar to the previous strategy, co-delivery stimulated CD8+ T cell response more efficiently [41].
Knuschke et al. synthesized biodegradable calcium phosphate (CaP) NPs encapsulating immunoactive TLR9 ligand (CpG) functionalized with the HA. Nanoconjugates were taken up by the dendritic cells in vivo and resulting in a strong T cell-mediated immune responses in immunized mice. Moreover, both methods of immunization (intraperitoneal and intranasal injections) were effective [42,43].
In a similar study, chitosan NPs were ionic cross-linked with sodium tripolyphosphate (CS/TPP) to deliver encapsulated HA antigen. The nanovaccine, tested in the mouse model, showed that HA encapsulated in CS/TPP could induce high numbers of IFN-γ-secreting cells in spleens while the naked HA vaccine could not. HA encapsulated in CS/TPP reduced the influenza morbidity and led to 100% protection against a lethal influenza virus dose in infected mice [44].
Kanekiyo et al. fused HA protein to ferritin, which naturally forms NPs composed of 24 identical units, leading to the assembly of the HA-generated eight trimeric HA spikes on its surface. Immunization of mice and ferrets elicited the reduction in HA Abs titers and induced neutralizing Abs against the HA structures: the stem and the RBS situated on the head. Nanovaccination protected against a broad spectrum of H1N1 IAV [45]. In a subsequent study, Kenekiyo et al. focused on the more conserved HA stem domain. Using HA-NPs elicited broadly cross-reactive Abs that fully protected mice and also partially protected ferrets against heterosubtypic H5N1 IAV [46].
The encapsulation method was also used to create recombinant H5 hemagglutinin trimer (H53) trapped into polyanhydride NPs. The H53 antigen works as a strong immunogen which induces high neutralizing Abs titers and enhances CD4+ T cell immune responses after the mice immunization. Moreover, H53-polyanhydride NPs induce higher CD4+ T cell memory than H53 antigen alone. However, the best results were obtained when H53-polyanhydride NPs were used along with immunostimmulant (poly I:C). Such combination enhanced proliferative T cell responses in mice and inducted of Abs titers at earlier time [47]. In the continuation of this study, Ross et al. examined encapsulated H53 in combined polyanhydride NPs and pentablock copolymer-based hydrogel to induce immunity against H5N1 IAV. Immunization in mice has induced high neutralizing Abs titers that were maintained for at least 70 days. While receiving the combination of nanovaccines, the mice had lower weight loss and reduced viral loads in the lung than mice vaccinated with each platform separately [48].
The subsequent research examined the immunogenicity stimulation with nanovaccine consisting of polyanhydride NPs encapsulating HA (H5M) antigen. The study confirmed the immunogenic potential of the nanovaccine, as level of immune responses against the H5N1 and H5N2 strains was significantly increased [49].

External Domain of the Matrix Protein 2 as a Viral Antigen in Vaccine Based on Nanoparticles

Antigenic drift and shift are mechanisms providing mutational potential of the influenza viruses [5]. In the antigenic drift, new types of HA and NA glycoproteins can evolve, while shifting can lead a new subtype to arise. For that reason, using a different viral antigen such as M2 protein might be very promising. The M2 is an integral membrane protein presented on the surface of nucleocapsid and the surface of virus-infected cells. It is engaged in processes such as cell entry and virion maturation [77,78]. The M2 protein is classified as a class I viroporin and consists of three domains: the extracellular N-terminal, internal hydrophobic, and C-terminal cytoplasmic domain [78]. The detailed study of the M2 protein showed that the Abs bind to the extracellular N-terminal region of the M2 protein (M2e) [79]. The natural immunity against the M2 protein in response to infection or induced by the conventional influenza vaccines is very weak [80,81]. However, combining the M2e domain with a protein carrier makes it highly immunogenic [82]. Therefore, using the M2e external domain of the M2 as a vaccine antigen aims to induce an adaptive immune response, which cooperates with the humoral anti-HA and anti-NA immunity. Moreover, the M2e antigen vaccine could overcome the shift and drift mechanisms and confront the virus with new immune responses. For that reason, the M2e is considered a universal vaccine candidate against the IAV [83].
In 2014, Tao et al. used a nanovaccine combining the highly conserved region of M2 protein attached to gold NPs (Au NPs). Mice were immunized intranasally with or without the addition of an adjuvant: soluble synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine rich oligonucleotide motifs (CpG-ODN). The CpG-ODN has been used as a mucosal adjuvant to stimulate Th1-biased cytokines and a high response of IgG2a Abs level. The study confirmed that M2e-Au NPs conjugates with the addition of CpG-ODN enabled stimulation of M2e-specific IgG Abs, which could recognize both M2e and native M2 resulting in full protection of mice against the lethal dose of A/PR/8/1934 IAV [50]. The subsequent study showed that the molar ratio of M2e covering the Au NPs surface completely has significant effects on immune responses and overall protection against the lethal influenza dose. The group of mice vaccinated with the highest amount of free M2e exhibited 100% protection against influenza A/PR/8/1934 (H1N1) virus and showed the least weight loss [51]. The same mice vaccination schedule showed that the A/California/04/2009 (H1N1), A/Victoria/3/1975 (H3N2) and A/Vietnam/1203/2004 (H5N1) influenza virus strains led to 100%, 92%, and 100% protection, respectively [52].
M2e proteins also have been used to create nanosized virus-like particles (VLP). For this purpose, one to five copies of M2e were fused to the recombinant nodavirus capsid proteins to form the VLP. Mice immunization developed Abs specifically against M2e and the Abs titer was proportional to the copy numbers of M2e displayed on the VLP [53]. Similar results have also been obtained by De Filette et al., where they fused up to three copies of M2e to the hepatitis B core antigen, which self-assembled into a VLP [54,55].

Mix of Influenza Membrane Proteins as Viral Antigens Vaccines Based on Nanoparticles

The localization of the antigen seems to be important while inducing an immune response. Interesting comparison of conjugated and encapsulated antigens (HA and NA from A/California/7/2009 (H1N1)) with trimethylaminoethylmethacrylate chitosan (TMC-chitosan) NPs was published by Liu et al. The group evaluated the antigen-specific immune responses in mice after nanovaccine nasal immunization. They concluded that antigen conjunct on TMC-chitosan NPs was more efficient to increase anti-H1N1 IgG level than antigen conjunct on pure TMC solution. The antigens conjunct on the surfaces of NPs were better recognized by APC than the antigens encapsulated into NPs. Functionalized NPs have been shown to stimulate macrophages and spleen lymphocytes to produce interleukins and interferons [56].
Hu et al. placed HA, NA and M1 antigens derived from A/Taiwan/S02076/2013 (H7N9) strain on the surface of the VLPs. VLPs were generated by co-infecting insect cells with three respective recombinant baculoviruses. In the experiments performed on mice and chickens, VLP immunization showed elevated Abs levels against NA and M1 proteins and increased antigen-specific cytokine production compared to the VLP-free protein formulation [57].
Further research focused on the combination of the recombinant HA and NA antigens. In detail, nanoadjuvants such as polyanhydride NPs and pentablock copolymer micelles were tested to check antiviral activity in the infected mice. The study indicates that two nanoadjuvants induced Abs titers significantly, resulting in reduced viral load and 100% protection in young mice [58].
Another interesting approach used biodegradable polylactic-co-glycolic acid (PLGA) NPs entrapping four conserved IAV H1N1 peptides: HA, nucleoprotein, PA and chimeric construct that express M2e on the surface loop of norovirus P particle (M2e-PP). Pigs vaccinated intranasally with PLGA NPs-peptides did not demonstrate any fever or flu symptoms. Vaccination without adjuvant also induced the virus-specific T cell responses in the lungs and reduced the viral load in the airways of pigs [59].

2.1.2. Inactivated Influenza Virus Strategy Based on NPs Delivery

Interesting research into the incorporation into chitosan NPs of the inactivated virus along with immunostimulants such as CpG ODN or Quillaja saponin was performed by Deghan et al. Rabbit immunostimulation with CpG ODN showed better results in the induction of the humoral and Th1 responses compared to immunostimulation with Quillaja saponin [60,61].
Dhakal et al. evaluated the effectiveness of a polyanhydride NP encapsulating the inactivated swine IAV in pigs. The nanovaccine induces virus-specific lymphocyte proliferation and increases the frequency of CD4+CD8αα+ T helper and CD8+ cytotoxic T cells. The animals have lower viral antigens in the lungs and six to eight-fold reduction in nasal shedding of IAV compared to control animals [62]. Moreover, the nanovaccine prevented influenza-related symptoms, including fever.
Morçöl et al. used calcium phosphate NPs (CaP NPs) carrying inactivated IAV and tested them in mice. The CaP NPs as adjuvants induced higher hemagglutination inhibition, virus neutralization, and IgG Abs titers compared to the non-adjuvanted vaccine [63].
Alkie et al. investigated different types of inactivated IAV encapsulated in PLGA in chickens. They tested PLGA NPs encapsulating IAV alone or co-encapsulating CpG ODN adjuvant. The PLGA NPs co-encapsulating IAV and CpG ODN were also surfaced modified with mannan or chitosan. The authors reported that all tested nanovaccines induced an immune responses [64].

2.1.3. mRNA Vaccines Strategy Based on NPs Delivery

mRNA vaccines are based on imitating the naturally occurring defective interfering particles (DIPs). Viral genes encoding structural proteins are replaced by mRNA fragments coding the antigens and deriving them in the viral-like NPs to the cells [84]. Thus, the mRNAs are replicated inside the vaccinated hosts’ cells, leading to the antigens’ expression, but without generating the infectious viral particles. Because the DIPs can activate the immune system by stimulating pathogen receptors—the pattern recognition receptors—the DIP-like NPs also have the same facility [84,85]. These kinds of vaccines are considered very promising to fight against infectious diseases, although they have some limitations. In comparison to standard vaccination strategy, this technology overcomes the antivector immunity responses and is egg-independent. Although the technology generates overall lower immunogenicity, it requires high doses and is considered as less stable [86].
One of the first studies using mRNA vaccines, was liposome-based NPs entrapping the mRNA encoding the nucleoprotein of A/Northern Territory/60/1968 (H3N2). The research performed in mice confirmed induced cytotoxic T lymphocytes, which were identical to those of the natural viral infection [65]. A similar approach has been tested by Bahl et al., where lipid NPs contained mRNA encoding HA protein of A/Jiangxi-Donghu/346/2013 (H10N8) or A/Anhui/1/2013 (H7N9) IAV strain. Immune responses have been studied in mice, ferrets, and nonhuman primates and measured by hemagglutination inhibition and microneutralization assays. Moreover, initial data from the first-in-human trial seem to confirm a robust immune response [66]. Another study used the self-amplifying mRNA (SAM® technology) in lipid nanoparticles (LNP) vectors expressing nucleoprotein and M1 IAV proteins. Nanovaccines tested in mice confirmed the stimulation of nucleoprotein, M1, and HA-specific T-cells and HA-specific Abs [67]. The very recent study allowed the authors to select the perfect candidate for the universal, multi-targeting anti-influenza vaccine construction. The multi-targeting mRNA-LNPs vaccine has been confirmed to induce broad immune responses in mice using single and low dosage [68].

2.2. Nanotechnology in Gene Silencing Strategies

Gene silencing strategies are based on the natural mechanisms of gene inactivation. Silencers are synthetic nucleic acids able to repress gene expression by targeting particular fragments of the genome or transcriptome. The group of silencers includes antisense oligonucleotides (ASO), ribozymes, DNAzymes, short interfering RNAs (siRNA), microRNAs (miRNAs), short hairpin RNA (shRNAs), or peptide nucleic acids (PNAs). The evidence for their effectiveness in vivo has been provided in many, including those performed in our group [87,88,89,90,91,92]. However, the use of synthetic nucleic acids is limited due to imperfect systems of their introduction into cells. As for today, a few biochemical and physical transfection methods are available on the market [92,93]. However, they often influence the cellular condition, thus the search for new ones with reduced cytotoxicity and improved transfection efficiency is still in the spotlight [94]. In answer to the need, the NPs seem to be very promising as a tool for evaluation of new delivery systems. The multiple possibilities of the modification and functionalization, as well as the diversity of NPs types, making them very attractive potential transfection reagents (Figure 3).

2.2.1. Antisense Oligonucleotides Strategy Based on NPs Delivery

The ASOs are single-stranded DNA oligonucleotides or RNA-modified oligonucleotides, generally 12–30 nt in length, which interact with the RNA of interest. There are several mechanisms of the ASO action: the activation of the RNase H cleavage of mRNA in DNA/RNA duplexes, creating a steric hindrance of the ribosome by binding ASO near the translational initiation site, destabilization and degradation of the transcript by ASO targeted near to the cap or the poly(A) of the mRNA, and destabilization or blocking the functional RNA structure (Figure 3 (A)). Other strategies include the inhibition of mRNA maturation and the induction of the mRNA precursor alternative splicing [95,96].
An interesting approach has been presented by Levina et al., who used ASO DNAs, targeting the noncoding 3′-terminal region of segment 5 (−)RNA. The ASOs were noncovalently immobilized on titanium dioxide NPs (TiO2 NPs) through the polylysine linker (PL). Nanocomposites used for the inhibition of H3N2 IAV replication in MDCK cells have shown a high antiviral activity simultaneously with the low cytotoxicity level. Nearly 50% of the virus inhibition concentration (IC50) was evaluated at 30 nM concentration for DNA (1.5 µg/mL of TiO2). Meanwhile, the 50% toxic concertation (TC50) was estimated to be 1800 µg/mL [97]. The subsequent study showed that the same ASO sequence covalently bound to PL and exhibited even higher antiviral activity than those associated via electrostatic interactions [98]. Next, they used the same nanocomposites to investigate the inhibition of the H1N1 and H5N1 strains. The antiviral activity of nanocomposites was very high (99,9%), probably due to targeting the conserved region of segment 5 IAV. The insertion of the mismatches has shown that the inhibition level is lower when the number of mismatches is higher [99]. In a subsequent study, using the ASOs targeting the most conservative regions of segment 5, both the negative (−)RNA and the positive (+)RNA sense strands of IAV were designed. Nanocomposites containing DNA targeting (−)RNA had higher antiviral activity than those targeting (+)RNA [100]. Moreover, Amirkhanov et al. studied the antiviral activity of TiO2-PL nanocomposites with DNA/peptide nucleic acids (PNA) heteroduplex. PNA targeting segment 5 of IAV RNA inhibited the IAV reproduction by 99.8% [101].
Levina et al. also presented ASO targeting the 3′-noncoding regions of viral (−)RNA of IAV segment 5 bound by electrostatic interactions to silicon–organic (Si-NH2) NPs. These NPs selectively and effectively inhibited the replication of the IAV in cell culture [102].

2.2.2. siRNA Strategy Based on NPs Delivery

RNA interference (RNAi) is a post-transcriptional gene regulation process in which siRNA or miRNA, incorporated in the RNA-induced silencing complex (RISC), targets the mRNA in a sequence-specific manner (Figure 3 (B)). Interestingly, the RNAi is one of the naturally occurring antiviral mechanisms in mammalian cells, although its specificity is still not fully understood [106]. The RNAi mechanism has been used as an antiviral strategy against many viruses, including influenza [107]. Moreover, currently used methods for siRNA design, the simplicity of synthesis procedures, and the development of the new nanotechnology-based delivery methods make it an even more promising anti-influenza strategy.
Frede et al. demonstrated multi-shell NPs consisting of a CaP core, covered with siRNAs encapsulated into PLGA (siRNA-loaded CaP/PLGA NPs) and coated with a final outer layer of polyethylenimine (PEI). They used siRNAs directly against the CCL-2, IP-10 and IFN-γ to reduce the gene expression in inflamed lung tissue. Mice treated with siRNA-loaded CaP/PLGA NPs revealed lower expression of targeted genes, resulting in reduced lung inflammation [103]. In the subsequent study, performed by Jamali et al., the chitosan NPs encapsulating siRNA targeting viral nucleoprotein were used. These siRNA-chitosan NP complexes inhibited IAV growth in cells and protected mice against a lethal viral dose [104].

2.2.3. DNAzymes Strategy Based on NPs Delivery

DNAzymes, so far not widely explored, might be also attractive for nanoparticle-based antiviral application. DNAzymes offer another opportunity for NP application, so far not widely explored. DNAzymes are DNA fragments with catalytic capability to cleave complementary RNA (Figure 3 (C)). Although there are several artificial DNAzymes, so far natural ones have not been detected. Repkova et al. demonstrated the anti-influenza activity of DNAzymes immobilized on TiO2 NPs precovered with PL. The DNAzymes targeted the coding region of (+)RNA of IAV segment 5. The ability of TiO2 DNAzymes to cleave the target RNA has resulted in lower IAV replication by 2700-fold in MDCK cells [105].

2.3. Bare Nanoparticles Antiviral Strategy

Nanocarriers are not only a very useful tool for delivering drugs or nucleic acids, but bare NPs might be used as antivirals as well (Table 3). Nanotechnology is a very fast-growing field of science and has an infinite number of NP synthesis methods. As for today, the only limitation is an accurate idea of what effect is wished to be achieved. The nano-scale particles’ and materials’ main feature is that they exhibit different properties than at a greater scale. Moreover, different shapes, sizes and surface modifications of NPs are available, making them even more welcome for wider use in biomedical sciences [108].
Mehrbod et al. published the first research concerning the antiviral properties of the silver NPs (Ag NPs). The in vivo exposure of Ag NPs before and after the viral infection reduced the titer by 78.42% (at concentration 0.5 µg/mL) [109]. Next, the antiviral potential of Ag NPs against the H1N1 and H3N2 strains in both in vitro and in vivo was investigated by Xiang et al. The authors concluded that the best antiviral effect, which was nearly 98% of the survival rate in MDCK cells, was achieved after 72 and 96 h post-infection at 50 µg/mL Ag NP concentration [110]. The subsequent in vivo studies showed that 75% of mice treated with Ag NPs survived, while the IAV titer in the lungs was reduced by 2 log10 [111]. Different sizes of Ag NPs embedded into the chitosan matrix were tested as IAV inhibitors as well. Smaller Ag NPs were more effective than the larger one at similar Ag NP concentrations [112]. Fatima et al. synthesized Ag NPs (AgNO3) using Cinnamomum cassia extract and evaluated their activity against the H7N3 strain. A more significant inhibition level was observed when Ag NPs were incubated with the virus before infection, where the concentration of 200 µg/mL led to an inhibition level of 76% [113]. In a similar approach, the ultra-sonication method was used to synthesize Ag NPs with an aqueous extract from Panax ginseng roots. At concentrations of 0.02 and 0.25 M of Ag NPs, inhibition levels were obtained at very low levels: 7.10% and 15.12%, respectively, while the cytotoxicity was at 11.19% and 19.45%, respectively [114]. These results suggest that bare Ag NPs have stronger antiviral effects than NPs prepared by green synthesis. The reduced effectiveness of the NPs might be a result of the non-specific surface functionalization with naturally occurring compounds in the extract. Liang et al. reported that montmorillonite clay-based nano silicate platelets (NSP) surface-modified by anionic sodium dodecyl sulfate have antiviral properties against the IAV. Moreover, the NSP modified by Ag NPs did not suppress the infection of IAV at a concentration 10 µg/mL [115].
Kumar et al. studied the influence of size, shape, and surface properties of the pristine and polymer coated magnetite iron oxide (Fe3O4; IO) NPs. The cytoplasmic presence of IO NPs in the MDCK cells after 12 h has been confirmed by TEM. The pristine IO NPs did not reduce the IAV titer, although the polyethylene glycol (PEG) and polyvinyl pyrolidine (PVP) coated NPs showed 2.5 and 3.5 log of virus reduction with a concentration of 30 µg/mL [116]. Next, they tested glycine coated IO NPs against the H1N1 strain. They observed an 8-fold lower expression of the viral transcripts after the 24 h of viral infection, when 2 pg/mL concentration of IO NPs was used [117].
Ag NPs and IO NPs are not the only ones reported for their antiviral properties. Zinc oxide NPs (ZnO NPs) were also used against IAV in the post-exposure experiment. Bare and PEGylated ZnO NPs at the highest non-toxic concentration (75 µg/mL) reduced the viral titer to 1.2 and 2.8 log10 TCID50, respectively. Likewise, the cells exposed to ZnO NPs were tested before, during and after IAV exposure. The inhibitory effects were observed only when ZnO NPs were added 1 h after infection [118]. Sametband et al. tested anionic gold NPs (Au NPs) against several influenza A and B strains. The Au NPs were functionalized with mercaptosuccinic acid (Au NPs-MSA) or with mercaptoethanesulfonate (Au-MES NPs) molecules bound to the NPs’ surface through the thiol groups. Both particles inhibited influenza activity in the MDCK tissue culture; however, Au-MES NPs were more potent inhibitors in the lower concentrations. At the highest used Au-MES NPs concentration (0.5 mg/mL), the inhibition level was decreased by 97%. The virus-sulfonate interaction may play a unique role in blocking the virus activity [119]. Papp et al. designed Au NPs functionalized by sialic-acid-terminated glycerol dendron. They showed that sialic-acid linked to 14 nm Au NPs effectively bound to HA protein and inhibited viral replication. In that case, 30 min of preincubation of sialic-acid@Au NPs with IAV reduced the viral titer by 40%. Moreover, the cytotoxicity assay did not show any impact on cells’ viability [120]. TiO2 NPs in anatase phase also showed inhibition of the H9N2 strain. Moreover, the TiO2 NPs activated by UV light were shown to enhance the antiviral activity compared to controls [121]. In another study conducted by Thammakarn et al., calcium oxide NPs (CaO NPs) were prepared by grinding scallop shell powder into 500 nm NPs. The CaO NPs inactivated H7N1 IAV within 5 s of incubation [122]. The antiviral effects of zirconium dioxide (ZrO2) NPs were tested in H5N1-infected mice. The most effective NPs were selected from a variety of different sizes, surface charges, and compositions. The most successful NPs were 200 nm positively-charged ZrO2 NPs, which effectively protected mice against the IAV at a concentration of 100 mg/kg [123].
The exact mechanism of the antiviral effects of bare NPs is not fully understood. Xiang et al. suggested that Ag NPs may damage the viral envelope and thus inhibit the virus attachment to the host membrane. Ag NPs may also react with HA and block the bond formation between HA-cell membrane [111]. The mechanism of IO NPs antiviral activity in cells is still unidentified, although it has been shown that IO NPs in contact with IAV exhibit lipid peroxidase activities that catalyze viral lipid envelope peroxidation [124].

2.4. Nanotechnology in Delivery of Drugs Targeting Proteins

Some of the first anti-influenza drugs approved by the FDA were M2 ion channel inhibitors—amantadine and rimantadine (Table 4). A few decades later, it became clear that new resistant strains had evolved [125]. Today, they are classified as “historically used drugs” and are no longer approved to use by FDA [126]. Over the years, oseltamivir- and zanamivir-resistant (neuraminidase inhibitors, NAIs) strains have been described [127,128,129]. Strains resistant to NAIs are less frequent and for that reason oseltamivir and zanamivir are widely used, and new methods of their synthesis continue to be developed [130,131]. Another used NAI is peramivir, formerly region-restricted to China, Japan, South Korea, and the USA [25]. It is already authorized in European countries [132]. The last NAI, authorized in Japan only, is laninamivir [25,133]. Most recently, another drug, called baloxavir marboxil, was approved to use. The drug is an inhibitor of endonuclease activity of viral polymerase complex [134,135]. Considering the mutational nature of the IAV, known drugs might be insufficient in the not-so-distant future [24]. For that reason, development of the new anti-influenza strategies, in particular based on new generation technologies, is desirable.
Li et al. and Lin et al. presented an interesting application of selenium NPs (Se NPs) to deliver anti-influenza drugs. Se NPs (70–200 nm) have been shown to be excellent carriers of oseltamivir, zanamivir, ribavirin, amantadine, and arbidol. These anti-influenza drugs successfully inhibited IAV replication [136,137,138,139,140]. The same authors tested Ag NPs as nanocarriers of amantadine, oseltamivir and zanamivir. The research showed effective inhibition of the IAV [141,142,143]. Figure 4 presents the influence of the particular drugs on the influenza life cycle.
NPs can also be used to deliver poorly soluble drugs. One of them is saliphenylhalamide (SaliPhe), which inhibits endosome acidification, an essential process for successful IAV infection (Figure 3 (C)). Bimbo et al. used thermally hydrocarbonized porous silicon NPs (THCPSi NPs) as a drug carrier to increase the solubility of SaliPhe. As a result, THCPSi@SaliPhe NPs inhibit the infection of IAV, and exhibit in vitro stability and low cytotoxicity [144].

3. Conclusions

The world of nanotechnology is rapidly expanding and showing new possibilities for the use of NPs in anti-influenza research and flu prevention. So far, we have been able to create NPs with a multitude of shapes, sizes, porosities, surface functionalizations, and other properties. Moreover, modifications of NPs can significantly alter their antiviral efficacy [108,112,116,123]. In this review, we presented various strategies for using NPs to fight influenza virus infection. NPs were used as advanced delivery vehicles for different antiviral agents, adding new functionality for known strategies. Additionally, NPs themselves appear to have anti-influenza properties. Besides the facilitation of potential drug development, the other category of nanotechnology application is usage in the delivery of immuno-inducing flu vaccines. Although the nanotechnology-based strategies and their antiviral application are very promising, the constantly mutating IAV genome is challenging and nanotechnology cannot overcome mutation problems. However, most of the strategies may be used for a wide range of IAV strains or can be easily adapted to new emerging strains. The NPs are excellent delivery agents of proteins, nucleic acids, anti-proteins drugs and vaccines. However, there is a need to ensure the safety of used NPs not only for human organisms but also for the environment in a long-term perspective. Possible cytotoxicity effects, drug–drug interactions or variable properties of each NP need to be carefully evaluated to take full benefit from the nanotechnology [145]. It is important to study the influence of particular NPs in in vivo systems. The good news is that there are already many NP-based antivirals that have shown minimal or no cytotoxic effects and might be considered as the therapeutics of the near future [118,146,147]. The safety of the nanotechnological products used in medicine is monitored and approved by the FDA [34,148]. There are already several cases of FDA-approved NP-based antivirals against different viruses, including influenza [145,149,150].

Author Contributions

Conceptualization, K.W. and E.K.; writing—original draft preparation, K.W.; writing—review and editing, K.W., B.S. and E.K.; supervision, E.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Science Centre grants UMO-2020/01/0/NZ6/00137 to E.K.

Acknowledgments

K.W. would like to acknowledge the project “Srodowiskowe interdyscyplinarne studia doktoranckie w zakresie nanotechnologii” No. POWR.03.02.00-00-I032/16 under the European Social Fund-Operational Programme Knowledge Education Development, Axis III Higher Education for Economy and Development, Action 3.2 PhD Programme.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Woolhouse, M.E.J.; Brierley, L. Epidemiological characteristics of human-infective RNA viruses. Sci. Data 2018, 5, 1–6. [Google Scholar] [CrossRef]
  2. Gounder, A.P.; Boon, A.C.M. Influenza Pathogenesis: The Effect of Host Factors on Severity of Disease. J. Immunol. 2019, 202, 341–350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fukuyama, S.; Kawaoka, Y. The pathogenesis of influenza virus infections: The contributions of virus and host factors. Curr. Opin. Immunol. 2011, 23, 481–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Bouvier, N.M.; Palese, P. The biology of influenza viruses. Vaccine 2008, 26, 49–53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kim, H.; Webster, R.G.; Webby, R.J. Influenza Virus: Dealing with a Drifting and Shifting Pathogen. Viral Immunol. 2018, 31, 174–183. [Google Scholar] [CrossRef] [PubMed]
  6. Rejmanek, D.; Hosseini, P.R.; Mazet, J.A.K.; Daszak, P.; Goldstein, T. Evolutionary Dynamics and Global Diversity of Influenza A Virus. J. Virol. 2015, 89, 10993–11001. [Google Scholar] [CrossRef] [Green Version]
  7. Shao, W.; Li, X.; Goraya, M.U.; Wang, S.; Chen, J.L. Evolution of influenza a virus by mutation and re-assortment. Int. J. Mol. Sci. 2017, 18, 1650. [Google Scholar] [CrossRef] [Green Version]
  8. Asadi, S.; Gaaloul, N.; Barre, R.S.; Ristenpart, W.D.; Bouvier, N.M.; Wexler, A.S. Influenza A virus is transmissible via aerosolized fomites. Nat. Commun. 2020, 11, 1–9. [Google Scholar] [CrossRef]
  9. Dunning, J.; Thwaites, R.S.; Openshaw, P.J.M. Seasonal and pandemic influenza: 100 years of progress, still much to learn. Mucosal Immunol. 2020, 13, 566–573. [Google Scholar] [CrossRef] [PubMed]
  10. Saunders-Hastings, P.R.; Krewski, D. Reviewing the history of pandemic influenza: Understanding patterns of emergence and transmission. Pathogens 2016, 5, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Martellucci, C.A.; Flacco, M.E.; Cappadona, R.; Bravi, F.; Mantovani, L.; Manzoli, L. SARS-CoV-2 pandemic: An overview. Adv. Biol. Regul. 2020, 77, 1–11. [Google Scholar] [CrossRef] [PubMed]
  12. Triggle, C.R.; Bansal, D.; Farag, E.A.B.A.; Ding, H.; Sultan, A.A. COVID-19: Learning from Lessons To Guide Treatment and Prevention Interventions. Am. Soc. Microbiol. 2020, 5, e00317-20. [Google Scholar] [CrossRef] [PubMed]
  13. Chang, T.; Wu, J.; Chang, L. Co-infection of influenza B virus and SARS-CoV-2: A case report from Taiwan. J. Formos. Med. Assoc. 2020, 19–20. [Google Scholar] [CrossRef]
  14. Hashemi, S.A.; Safamanesh, S.; Ghasemzadeh-moghaddam, H.; Ghafouri, M.; Amir, A. High prevalence of SARS-CoV-2 and influenza A virus (H1N1) co-infection in dead patients in Northeastern Iran. J. Med. Virol. 2020, 1–5. [Google Scholar] [CrossRef]
  15. Kondo, Y.; Miyazaki, S.; Yamashita, R.; Ikeda, T. Coinfection with SARS-CoV-2 and influenza A virus. BMJ Case Rep. 2020, 13. [Google Scholar] [CrossRef]
  16. Singh, B.; Kaur, P.; Reid, R.-J.; Shamoon, F.; Bikkina, M. COVID-19 and Influenza Co-Infection: Report of Three Cases. Cureus 2020, 12, e9852. [Google Scholar] [CrossRef]
  17. Centers for Disease Control and Prevention, National Center for Immunization and Respiratory Diseases (NCIRD) How to Prevent Flu. Available online: https://www.cdc.gov/flu/prevent/prevention.htm (accessed on 28 September 2020).
  18. World Health Organization. WHO Rcommendations on the Composition of Influenza Virus Vaccines. Available online: https://www.who.int/influenza/vaccines/virus/recommendations/en/ (accessed on 28 September 2020).
  19. Centers for Disease Control and Prevention, National Center for Immunization and Respiratory Diseases (NCIRD) Past Seasons Vaccine Effectiveness Estimates. Available online: https://www.cdc.gov/flu/vaccines-work/past-seasons-estimates.html (accessed on 28 September 2020).
  20. Kissling, E.; Pozo, F.; Buda, S.; Gherasim, A.; Brytting, M.; Domegan, L.; Gómez, V.; Meijer, A.; Lazar, M.; Vučina, V.V.; et al. Low 2018/19 vaccine effectiveness against influenza A(H3N2) among 15–64-year-olds in Europe: Exploration by birth cohort. Eurosurveillance 2019, 24, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Kissling, E.; Rose, A.; Embor, H.A.; Gherasim, A.; Pebody, R.; Pozo, F.; Trebbien, R.; Mazagatos, C.; Whitaker, H.; Valenciano, M.; et al. Interim 2018/19 influenza vaccine effectiveness: Six European studies, October 2018 to January 2019. Eurosurveillance 2019, 24, 1900121. [Google Scholar] [CrossRef]
  22. Estrada Leonardo, D.; Shultz-Cherry, S. Development of a universal influenza vaccine. J. Immunol. 2019, 202, 392–398. [Google Scholar] [CrossRef] [Green Version]
  23. Eshaghi, A.; Shalhoub, S.; Rosenfeld, P.; Li, A.; Higgins, R.R.; Stogios, P.J.; Savchenko, A.; Bastien, N.; Li, Y.; Rotstein, C.; et al. Multiple influenza a (H3N2) mutations conferring resistance to neuraminidase inhibitors in a bone marrow transplant recipient. Antimicrob. Agents Chemother. 2014, 58, 7188–7197. [Google Scholar] [CrossRef] [Green Version]
  24. Dobrovolny, H.M.; Beauchemin, C.A.A. Modelling the emergence of influenza drug resistance: The roles of surface proteins, the immune response and antiviral mechanisms. PLoS ONE 2017, 12, e18058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Bassetti, M.; Castaldo, N.; Carnelutti, A. Neuraminidase inhibitors as a strategy for influenza treatment: Pros, cons and future perspectives. Expert Opin. Pharmacother. 2019, 20, 1711–1718. [Google Scholar] [CrossRef] [PubMed]
  26. Wu, X.; Wu, X.; Sun, Q.; Zhang, C.; Yang, S.Y.; Li, L.; Jia, Z. Progress of small molecular inhibitors in the development of anti-influenza virus agents. Theranostics 2017, 7, 826–845. [Google Scholar] [CrossRef]
  27. Davidson, S. Treating influenza infection, from now and into the future. Front. Immunol. 2018, 9, 1946. [Google Scholar] [CrossRef] [PubMed]
  28. Keshavarz, M.; Dianat-Moghadam, H.; Sofiani, V.H.; Karimzadeh, M.; Zargar, M.; Moghoofei, M.; Biglari, H.; Ghorbani, S.; Nahand, J.S.; Mirzaei, H. MiRNA-based strategy for modulation of influenza A virus infection. Epigenomics 2018, 10, 829–844. [Google Scholar] [CrossRef] [PubMed]
  29. Bhagyaraj, S.M.; Oluwafemi, O.S. Synthesis of Inorganic Nanomaterials, 1st ed.; Elsevier Ltd.: Amsterdam, The Netherlands, 2018; ISBN 978008019764. [Google Scholar]
  30. Lungu, M.; Neculae, A.; Bunoiu, M.; Biris, C. Nanoparticles’ Promises and Risks; Springer Nature: Basel, Switzerland, 2015; ISBN 9783319117287. [Google Scholar]
  31. Vert, M.; Doi, Y.; Hellwich, K.A.; Hess, M.; Hodge, P.; Kubisa, P.; Rinaudo, M.; Schué, F. Terminology for biorelated polymers applications (IUPACRecommendations, 2.0.1.2.). Pure Appl. Chem. 2012, 84, 377–410. [Google Scholar] [CrossRef]
  32. Kroll, A. Nanobiology-convergence of disciplines inspires great applications. Cell. Mol. Life Sci. 2012, 69, 335–336. [Google Scholar] [CrossRef] [Green Version]
  33. Schmidt, C.; Storsberg, J. Nanomaterials-tools, technology and methodology of nanotechnology based biomedical systems for diagnostics and therapy. Biomedicines 2015, 3, 203–223. [Google Scholar] [CrossRef] [Green Version]
  34. Bobo, D.; Robinson, K.J.; Islam, J.; Thurecht, K.J.; Corrie, S.R. Nanoparticle-Based Medicines: A Review of FDA-Approved Materials and Clinical Trials to Date. Pharm. Res. 2016, 33, 2373–2387. [Google Scholar] [CrossRef]
  35. Sanna, V.; Sechi, M. Therapeutic Potential of Targeted Nanoparticles and Perspective on Nanotherapies. ACS Med. Chem. Lett. 2020, 11, 1069–1073. [Google Scholar] [CrossRef] [Green Version]
  36. Wang, Y.; Yu, L.; Kong, X.; Sun, L. Application of nanodiagnostics in point-of-care tests for infectious diseases. Int. J. Nanomed. 2017, 12, 4789–4803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Gamage, S.; Howard, M.; Makita, H.; Cross, B.; Hastings, G.; Luo, M.; Abate, Y. Probing structural changes in single enveloped virus particles using nano-infrared spectroscopic imaging. PLoS ONE 2018, 13, e0199112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Toy, R.; Roy, K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng. Transl. Med. 2016, 1, 47–62. [Google Scholar] [CrossRef] [PubMed]
  39. Chen, T.; Shukoor, M.I.; Chen, Y.; Yuan, Q.; Zhu, Z.; Zhao, Z.; Gulbakan, B.; Tan, W. Aptamer-conjugated nanomaterials for bioanalysis and biotechnology applications. Nanoscale 2011, 3, 546–556. [Google Scholar] [CrossRef]
  40. Wang, C.; Zhu, W.; Wang, B.Z. Dual-linker gold nanoparticles as adjuvanting carriers for multivalent display of recombinant influenza hemagglutinin trimers and flagellin improve the immunological responses in vivo and in vitro. Int. J. Nanomed. 2017, 12, 4747–4762. [Google Scholar] [CrossRef] [Green Version]
  41. Wang, C.; Zhu, W.; Luo, Y.; Wang, B.Z. Gold nanoparticles conjugating recombinant influenza hemagglutinin trimers and flagellin enhanced mucosal cellular immunity. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1349–1360. [Google Scholar] [CrossRef]
  42. Knuschke, T.; Sokolova, V.; Rotan, O.; Wadwa, M.; Tenbusch, M.; Hansen, W.; Staeheli, P.; Epple, M.; Buer, J.; Westendorf, A.M. Immunization with Biodegradable Nanoparticles Efficiently Induces Cellular Immunity and Protects against Influenza Virus Infection. J. Immunol. 2013, 190, 6221–6229. [Google Scholar] [CrossRef] [Green Version]
  43. Sokolova, V.; Knuschke, T.; Kovtun, A.; Buer, J.; Epple, M.; Westendorf, A.M. The use of calcium phosphate nanoparticles encapsulating Toll-like receptor ligands and the antigen hemagglutinin to induce dendritic cell maturation and T cell activation. Biomaterials 2010, 31, 5627–5633. [Google Scholar] [CrossRef]
  44. Sawaengsak, C.; Mori, Y.; Yamanishi, K.; Mitrevej, A.; Sinchaipanid, N. Chitosan nanoparticle encapsulated hemagglutinin-split influenza virus mucosal vaccine. AAPS PharmSciTech 2014, 15, 317–325. [Google Scholar] [CrossRef] [Green Version]
  45. Kanekiyo, M.; Wei, C.J.; Yassine, H.M.; McTamney, P.M.; Boyington, J.C.; Whittle, J.R.R.; Rao, S.S.; Kong, W.P.; Wang, L.; Nabel, G.J. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 2013, 499, 102–106. [Google Scholar] [CrossRef]
  46. Yassine, H.M.; Boyington, J.C.; McTamney, P.M.; Wei, C.J.; Kanekiyo, M.; Kong, W.P.; Gallagher, J.R.; Wang, L.; Zhang, Y.; Joyce, M.G.; et al. Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection. Nat. Med. 2015, 21, 1065–1070. [Google Scholar] [CrossRef] [PubMed]
  47. Ross, K.A.; Lyod, H.; Wu, W.; Huntimer, L.; Ahmed, S.; Sambol, A.; Broderick, S.; Flickinger, Z.; Rajan, K.; Bronich, T.; et al. Hemagglutinin-based polyanhydride nanovaccines against H5N1 infuenza elicit protective virus neutralizing titers and cell-mediated immunity. Int. J. Nanomed. 2015, 10, 229–243. [Google Scholar] [CrossRef] [Green Version]
  48. Ross, K.; Adams, J.; Loyd, H.; Ahmed, S.; Sambol, A.; Broderick, S.; Rajan, K.; Kohut, M.; Bronich, T.; Wannemuehler, M.J.; et al. Combination Nanovaccine Demonstrates Synergistic Enhancement in Efficacy against Influenza. ACS Biomater. Sci. Eng. 2016, 2, 368–374. [Google Scholar] [CrossRef]
  49. Kingstad-Bakke, B.A.; Chandrasekar, S.S.; Phanse, Y.; Ross, K.A.; Hatta, M.; Suresh, M.; Kawaoka, Y.; Osorio, J.E.; Narasimhan, B.; Talaat, A.M. Effective mosaic-based nanovaccines against avian influenza in poultry. Vaccine 2019, 37, 5051–5058. [Google Scholar] [CrossRef]
  50. Tao, W.; Ziemer, K.S.; Gill, H.S. Gold nanoparticle–M2e conjugate coformulated with CpG induces protective immunity against influenza A virus. Nanomedicine (Lond.) 2014, 9, 237–251. [Google Scholar] [CrossRef] [Green Version]
  51. Tao, W.; Gill, H.S. M2e-immobilized gold nanoparticles as influenza A vaccine: Role of soluble M2e and longevity of protection. Vaccine 2015, 33, 2307–2315. [Google Scholar] [CrossRef] [Green Version]
  52. Tao, W.; Hurst, B.; Shakya, A.K.; Uddin, M.J.; Ingrole, R.S.; Hernandez-Sanabria, M.; Arya, R.; Bimler, L.; Paust, S.; Tarbet, E.B.; et al. Consensus M2e peptide conjugated to gold nanoparticles confers protection against H1N1, H3N2 and H5N1 influenza A viruses. Antivir. Res. 2017, 141, 62–72. [Google Scholar] [CrossRef] [Green Version]
  53. Yong, C.Y.; Yeap, S.K.; Ho, K.L.; Omar, A.R.; Tan, W.S. Potential recombinant vaccine against influenza A virus based on M2e displayed on nodaviral capsid nanoparticles. Int. J. Nanomed. 2015, 10, 2751–2763. [Google Scholar] [CrossRef] [Green Version]
  54. de Filette, M.; Jou, W.M.; Birkett, A.; Lyons, K.; Schultz, B.; Tonkyro, A.; Resch, S.; Fiers, W. Universal influenza A vaccine: Optimization of M2-based constructs. Virology 2005, 337, 149–161. [Google Scholar] [CrossRef] [Green Version]
  55. de Filette, M.; Martens, W.; Smet, A.; Schotsaert, M.; Birkett, A.; Londoño-Arcila, P.; Fiers, W.; Saelens, X. Universal influenza A M2e-HBc vaccine protects against disease even in the presence of pre-existing anti-HBc antibodies. Vaccine 2008, 26, 6503–6507. [Google Scholar] [CrossRef]
  56. Liu, Q.; Zheng, X.; Zhang, C.; Shao, X.; Zhang, X.; Zhang, Q.; Jiang, X. Conjugating Influenza A (H1N1) Antigen to N-Trimethylaminoethylmethacrylate Chitosan Nanoparticles Improves the Immunogenicity of the Antigen After Nasal Administration. J. Med. Virol. 2015, 87, 1807–1815. [Google Scholar] [CrossRef] [PubMed]
  57. Hu, C.M.J.; Chien, C.Y.; Liu, M.T.; Fang, Z.S.; Chang, S.Y.; Juang, R.H.; Chang, S.C.; Chen, H.W. Multi-antigen avian influenza a (H7N9) virus-like particles: Particulate characterizations and immunogenicity evaluation in murine and avian models. BMC Biotechnol. 2017, 17, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Ross, K.; Senapati, S.; Alley, J.; Darling, R.; Goodman, J.; Jefferson, M.; Uz, M.; Guo, B.; Yoon, K.J.; Verhoeven, D.; et al. Single dose combination nanovaccine provides protection against influenza A virus in young and aged mice. Biomater. Sci. 2019, 7, 809–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Hiremath, J.; Kang, K.I.; Xia, M.; Elaish, M.; Binjawadagi, B.; Ouyang, K.; Dhakal, S.; Arcos, J.; Torrelles, J.B.; Jiang, X.; et al. Entrapment of H1N1 influenza virus derived conserved peptides in PLGA nanoparticles enhances T cell response and vaccine efficacy in pigs. PLoS ONE 2016, 11, e0151922. [Google Scholar] [CrossRef] [Green Version]
  60. Dehghan, S.; Kheiri, M.T.; Tabatabaiean, M.; Darzi, S.; Tafaghodi, M. Dry-powder form of chitosan nanospheres containing influenza virus and adjuvants for nasal immunization. Arch. Pharm. Res. 2013, 36, 981–992. [Google Scholar] [CrossRef]
  61. Dehghan, S.; Tafaghodi, M.; Bolourieh, T.; Mazaheri, V.; Torabi, A.; Abnous, K.; Tavassoti Kheiri, M. Rabbit nasal immunization against influenza by dry-powder form of chitosan nanospheres encapsulated with influenza whole virus and adjuvants. Int. J. Pharm. 2014, 475, 1–8. [Google Scholar] [CrossRef]
  62. Dhakal, S.; Goodman, J.; Bondra, K.; Lakshmanappa, Y.S.; Hiremath, J.; Shyu, D.L.; Ouyang, K.; Kang, K.I.; Krakowka, S.; Wannemuehler, M.J.; et al. Polyanhydride nanovaccine against swine influenza virus in pigs. Vaccine 2017, 35, 1124–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Morçöl, T.; Hurst, B.L.; Tarbet, E.B. Calcium phosphate nanoparticle (CaPNP) for dose-sparing of inactivated whole virus pandemic influenza A (H1N1) 2009 vaccine in mice. Vaccine 2017, 35, 4569–4577. [Google Scholar] [CrossRef] [PubMed]
  64. Alkie, T.N.; Yitbarek, A.; Taha-Abdelaziz, K.; Astill, J.; Sharif, S. Characterization of immunogenicity of avian influenza antigens encapsulated in PLGA nanoparticles following mucosal and subcutaneous delivery in chickens. PLoS ONE 2018, 13, e0206324. [Google Scholar] [CrossRef] [PubMed]
  65. Martinon, F.; Krishnan, S.; Lenzen, G.; Magné, R.; Gomard, E.; Guillet, J.-G.; Lévy, J.-P.; Meulien, P. Induction of virus-specific cytotoxic T lymphocytes in vivo by liposome-entrapped mRNA. Eur. J. Immunol. 1993, 23, 1719–1722. [Google Scholar] [CrossRef]
  66. Bahl, K.; Senn, J.J.; Yuzhakov, O.; Bulychev, A.; Brito, L.A.; Hassett, K.J.; Laska, M.E.; Smith, M.; Almarsson, Ö.; Thompson, J.; et al. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol. Ther. 2017, 25, 1316–1327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Magini, D.; Giovani, C.; Mangiavacchi, S.; MacCari, S.; Cecchi, R.; Ulmer, J.B.; De Gregorio, E.; Geall, A.J.; Brazzoli, M.; Bertholet, S. Self-amplifying mRNA vaccines expressing multiple conserved influenza antigens confer protection against homologous and heterosubtypic viral challenge. PLoS ONE 2016, 11, e0161193. [Google Scholar] [CrossRef] [PubMed]
  68. Freyn, A.W.; Ramos da Silva, J.; Rosado, V.C.; Bliss, C.M.; Pine, M.; Mui, B.L.; Tam, Y.K.; Madden, T.D.; de Souza Ferreira, L.C.; Weissman, D.; et al. A Multi-Targeting, Nucleoside-Modified mRNA Influenza Virus Vaccine Provides Broad Protection in Mice. Mol. Ther. 2020, 28, 1569–1584. [Google Scholar] [CrossRef] [PubMed]
  69. Stöhr, K.; Bucher, D.; Colgate, T.; Wood, J. Influenza Virus Surveillance, Vaccine Strain Selection, and Manufacture. Methods Mol. Biol. 2012, 865, 147–162. [Google Scholar] [CrossRef] [PubMed]
  70. Plotkin, S.; Robinson, J.M.; Cunningham, G.; Iqbal, R.; Larsen, S. The complexity and cost of vaccine manufacturing—An overview. Vaccine 2017, 35, 4064–4071. [Google Scholar] [CrossRef] [PubMed]
  71. Global, W.H.O.; Programme, I.; September, B.; Europe, E. Recommended composition of influenza virus vaccines for use in the 2020 southern hemisphere influenza season. Wkly. Epidemiol. Rec. 2019, 94, 473–484. [Google Scholar]
  72. World Health Organization. Recommended Composition of Influenza Virus Vaccines for Use in the 2018–2019 Northern Hemisphere Influenza Season. Available online: https://www.who.int/influenza/vaccines/virus/recommendations/201802_recommendation.pdf (accessed on 28 September 2020).
  73. Hollingsworth, R.; El Guerche-Séblain, C.; Tsai, T.; Vasiliev, Y.; Lee, S.; Bright, H.; Barbosa, P. Assessment of the benefits of seasonal influenza vaccination: Elements of a framework to interpret estimates of vaccine effectiveness estimates and support robust decision-making and communication. Influenza Other Respir. Viruses 2020. [Google Scholar] [CrossRef]
  74. Padilla-Quirarte, H.O.; Lopez-Guerrero, D.V.; Gutierrez-Xicotencatl, L.; Esquivel-Guadarrama, F. Protective antibodies against influenza proteins. Front. Immunol. 2019, 10, 1677. [Google Scholar] [CrossRef] [Green Version]
  75. Kosik, I.; Yewdell, J.W. Influenza hemagglutinin and neuraminidase: Yin–yang proteins coevolving to thwart immunity. Viruses 2019, 11, 346. [Google Scholar] [CrossRef] [Green Version]
  76. Garten, W.; Klenk, H. Understanding influenza virus pathogenicity Septic shock caused by Gram-positive bacteria. Trends Microbiol. 1999, 7, 99–100. [Google Scholar] [CrossRef]
  77. Pinto, L.H.; Lamb, R.A. Influenza virus proton channels. Photochem. Photobiol. Sci. 2006, 5, 629–632. [Google Scholar] [CrossRef] [PubMed]
  78. Manzoor, R.; Igarashi, M.; Takada, A. Influenza A virus M2 protein: Roles from ingress to egress. Int. J. Mol. Sci. 2017, 18, 2649. [Google Scholar] [CrossRef] [Green Version]
  79. Zebedee, S.L.; Lamb, R.A. Influenza A virus M2 protein: Monoclonal antibody restriction of virus growth and detection of M2 in virions. J. Virol. 1988, 62, 2762–2772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Drummond, J.E.; Shaw, E.E.; Antonello, J.M.; Green, T.; Page, G.J.; Motley, C.O.; Wilson, K.A.; Finnefrock, A.C.; Liang, X.; Casimiro, D.R. Design and optimization of a multiplex anti-influenza peptide immunoassay. J. Immunol. Methods 2008, 334, 11–20. [Google Scholar] [CrossRef] [PubMed]
  81. Kitikoon, P.; Strait, E.L.; Thacker, E.L. The antibody responses to swine influenza virus (SIV) recombinant matrix 1 (rM1), matrix 2 (M2), and hemagglutinin (HA) proteins in pigs with different SIV exposure. Vet. Microbiol. 2008, 126, 51–62. [Google Scholar] [CrossRef] [PubMed]
  82. Schepens, B.; De Vlieger, D.; Saelens, X. Vaccine options for influenza: Thinking small. Curr. Opin. Immunol. 2018, 53, 22–29. [Google Scholar] [CrossRef]
  83. Mezhenskaya, D.; Isakova-Sivak, I.; Rudenko, L. M2e-based universal influenza vaccines: A historical overview and new approaches to development. J. Biomed. Sci. 2019, 26, 1–15. [Google Scholar] [CrossRef]
  84. Vasou, A.; Sultanoglu, N.; Goodbourn, S.; Randall, R.E.; Kostrikis, L.G. Targeting pattern recognition receptors (PRR) for vaccine adjuvantation: From synthetic PRR agonists to the potential of defective interfering particles of viruses. Viruses 2017, 9, 186. [Google Scholar] [CrossRef] [Green Version]
  85. Demento, S.L.; Eisenbarth, S.C.; Foellmer, H.G.; Platt, C.; Caplan, J.; Saltzman, W.M.; Mellman, I.; Ledizet, M.; Fikrig, E.; Richard, A.; et al. Inflammasome-activating nanoparticles as modulac systems for optimizing vaccine efficacy. Vaccine 2010, 27, 3013–3021. [Google Scholar] [CrossRef] [Green Version]
  86. Zhang, C.; Maruggi, G.; Shan, H.; Li, J. Advances in mRNA vaccines for infectious diseases. Front. Immunol. 2019, 10, 564. [Google Scholar] [CrossRef] [Green Version]
  87. Piasecka, J.; Lenartowicz, E.; Soszynska-Jozwiak, M.; Szutkowska, B.; Kierzek, R.; Kierzek, E. RNA Secondary Structure Motifs of the Influenza A Virus as Targets for siRNA-Mediated RNA Interference. Mol. Ther. Nucleic Acids 2020, 19, 627–642. [Google Scholar] [CrossRef]
  88. Huang, D.T.-N.; Lu, C.-Y.; Shao, P.-L.; Chang, L.-Y.; Wang, J.-Y.; Chang, Y.-H.; Lai, M.-J.; Chi, Y.-H.; Huang, L.-M. In vivo inhibition of influenza A virus replication by RNA interference targeting the PB2 subunit via intratracheal delivery. PLoS ONE 2017, 12, e0174523. [Google Scholar] [CrossRef] [PubMed]
  89. Kesy, J.; Patil, K.M.; Kumar, S.R.; Shu, Z.; Yong, H.Y.; Zimmermann, L.; Ong, A.A.L.; Toh, D.F.K.; Krishna, M.S.; Yang, L.; et al. A Short Chemically Modified dsRNA-Binding PNA (dbPNA) Inhibits Influenza Viral Replication by Targeting Viral RNA Panhandle Structure. Bioconjug. Chem. 2019, 30, 931–943. [Google Scholar] [CrossRef] [PubMed]
  90. Lenartowicz, E.; Nogales, A.; Kierzek, E.; Kierzek, R.; Martínez-Sobrido, L.; Turner, D.H. Antisense Oligonucleotides Targeting Influenza A Segment 8 Genomic RNA Inhibit Viral Replication. Nucleic Acid Ther. 2016, 26, 277–285. [Google Scholar] [CrossRef] [Green Version]
  91. Michalak, P.; Soszynska-Jozwiak, M.; Biala, E.; Moss, W.N.; Kesy, J.; Szutkowska, B.; Lenartowicz, E.; Kierzek, R.; Kierzek, E. Secondary structure of the segment 5 genomic RNA of influenza A virus and its application for designing antisense oligonucleotides. Sci. Rep. 2019, 9, 3801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Reed, C.A.; Peralta, E.R.; Wenrich, L.M.; Wong, C.A.; Bennett, C.F.; Freier, S.; Lollo, B. Transfection protocol for antisense oligonucleotides affects uniformity of transfection in cell culture and efficiency of mRNA target reduction. Oligonucleotides 2005, 15, 12–22. [Google Scholar] [CrossRef] [PubMed]
  93. Pavlin, M.; Kandušer, M.; Reberšek, M.; Pucihar, G.; Hart, F.X.; Magjarević, R.; Miklavčič, D. Effect of cell electroporation on the conductivity of a cell suspension. Biophys. J. 2005, 88, 4378–4390. [Google Scholar] [CrossRef] [Green Version]
  94. Kim, T.K.; Eberwine, J.H. Mammalian cell transfection: The present and the future. Anal. Bioanal. Chem. 2010, 397, 3173–3178. [Google Scholar] [CrossRef] [Green Version]
  95. Mescalchin, A.; Restle, T. Oligomeric nucleic acids as antivirals. Molecules 2011, 16, 1271–1296. [Google Scholar] [CrossRef]
  96. Frazier, K.S. Antisense Oligonucleotide Therapies:The Promise and the Challenges from a Toxicologic Pathologist’s Perspective. Toxicol. Pathol. 2015, 43, 78–89. [Google Scholar] [CrossRef]
  97. Levina, A.S.; Repkova, M.N.; Ismagilov, Z.R.; Shikina, N.V.; Malygin, E.G.; Mazurkova, N.A.; Zinov’ev, V.V.; Evdokimov, A.A.; Baiborodin, S.I.; Zarytova, V.F. High-performance method for specific effect on nucleic acids in cells using TiO2~DNA nanocomposites. Sci. Rep. 2012, 2, 756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Levina, A.S.; Repkova, M.N.; Ismagilov, Z.R.; Shikina, N.V.; Mazurkova, N.A.; Zarytova, V.F. Efficient inhibition of human influenza a virus by oligonucleotides electrostatically fixed on polylysine-containing TiO2 nanoparticles. Russ. J. Bioorg. Chem. 2014, 40, 179–184. [Google Scholar] [CrossRef] [PubMed]
  99. Levina, A.S.; Repkova, M.N.; Mazurkova, N.A.; Makarevich, E.V.; Ismagilov, Z.R.; Zarytova, V.F. Knockdown of different influenza A virus subtypes in cell culture by a single antisense oligodeoxyribonucleotide. Int. J. Antimicrob. Agents 2015, 46, 125–128. [Google Scholar] [CrossRef] [PubMed]
  100. Levina, A.S.; Repkova, M.N.; Bessudnova, E.V.; Filippova, E.I.; Mazurkova, N.A.; Zarytova, V.F. High antiviral effect of TiO2·PL-DNA nanocomposites targeted to conservative regions of (-)RNA and (+)RNA of influenza A virus in cell culture. Beilstein J. Nanotechnol. 2016, 7, 1166–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Amirkhanov, R.N.; Mazurkova, N.A.; Amirkhanov, N.V.; Zarytova, V.F. Composites of peptide nucleic acids with titanium dioxide nanoparticles. IV. Antiviral activity of nanocomposites containing DNA/PNA duplexes. Russ. J. Bioorg. Chem. 2015, 41, 162–169. [Google Scholar] [CrossRef] [PubMed]
  102. Levina, A.S.; Repkova, M.N.; Shikina, N.V.; Ismagilov, Z.R.; Yashnik, S.A.; Semenov, D.V.; Savinovskaya, Y.I.; Mazurkova, N.A.; Pyshnaya, I.A.; Zarytova, V.F. Non-agglomerated silicon-organic nanoparticles and their nanocomplexes with oligonucleotides: Synthesis and properties. Beilstein J. Nanotechnol. 2018, 9, 2516–2525. [Google Scholar] [CrossRef] [Green Version]
  103. Frede, A.; Neuhaus, B.; Knuschke, T.; Wadwa, M.; Kollenda, S.; Klopfleisch, R.; Hansen, W.; Buer, J.; Bruder, D.; Epple, M.; et al. Local delivery of siRNA-loaded calcium phosphate nanoparticles abates pulmonary inflammation. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2395–2403. [Google Scholar] [CrossRef]
  104. Jamali, A.; Mottaghitalab, F.; Abdoli, A.; Dinarvand, M.; Esmailie, A.; Kheiri, M.T.; Atyabi, F. Inhibiting influenza virus replication and inducing protection against lethal influenza virus challenge through chitosan nanoparticles loaded by siRNA. Drug Deliv. Transl. Res. 2018, 8, 12–20. [Google Scholar] [CrossRef]
  105. Repkova, M.; Levina, A.; Chelobanov, B.; Ismagilov, Z.; Shatskaya, N.; Baiborodin, S.; Filippova, E.; Mazurkova, N.; Zarytova, V. Efficient inhibition of influenza A viral replication in cells by deoxyribozymes delivered by nanocomposites. Int. J. Antimicrob. Agents 2017, 49, 703–708. [Google Scholar] [CrossRef]
  106. Ding, S.-W.; Han, Q.; Wang, J.; Li, W.-X. Antiviral RNA interference in mammals. Curr. Opin. Immunol. 2019, 54, 109–114. [Google Scholar] [CrossRef]
  107. Qureshi, A.; Tantray, V.G.; Kirmani, A.R.; Ahangar, A.G. A review on current status of antiviral siRNA. Rev. Med. Virol. 2018, 28, e1976. [Google Scholar] [CrossRef] [PubMed]
  108. Brandelli, A.; Ritter, A.C.; Veras, F.F. Antimicrobial Activities of Metal Nanoparticles. In Metal Nanoparticles in Pharma; Springer: Cham, Switzerland, 2017; pp. 337–363. [Google Scholar] [CrossRef]
  109. Mehrbod, P.; Motamed, N.; Tabatabaian, M.; Soleimani Estyar, R.; Amini, E.; Shahidi, M.; Kheiri, M.T. In vitro antiviral effect of “nanosilver” on influenza virus. Daru 2009, 17, 88–93. [Google Scholar]
  110. Xiang, D.X.; Chen, Q.; Pang, L.; Zheng, C.L. Inhibitory effects of silver nanoparticles on H1N1 influenza A virus in vitro. J. Virol. Methods 2011, 178, 137–142. [Google Scholar] [CrossRef] [PubMed]
  111. Xiang, D.; Zheng, Y.; Duan, W.; Li, X.; Yin, J.; Shigdar, S.; O’Connor, M.L.; Marappan, M.; Zhao, X.; Miao, Y.; et al. Inhibition of A/Human/Hubei/3/2005 (H3N2) influenza virus infection by silver nanoparticles in vitro and in vivo. Int. J. Nanomed. 2013, 8, 4103–4114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Mori, Y.; Ono, T.; Miyahira, Y.; Nguyen, V.Q.; Matsui, T.; Ishihara, M. Antiviral activity of silver nanoparticle/chitosan composites against H1N1 influenza A virus. Nanoscale Res. Lett. 2013, 8, 93. [Google Scholar] [CrossRef] [Green Version]
  113. Fatima, M.; Zaidi, N.u.S.S.; Amraiz, D.; Afzal, F. In vitro antiviral activity of Cinnamomum cassia and its nanoparticles against H7N3 influenza a virus. J. Microbiol. Biotechnol. 2015, 26, 151–159. [Google Scholar] [CrossRef]
  114. Sreekanth, T.V.M.; Nagajyothi, P.C.; Muthuraman, P.; Enkhtaivan, G.; Vattikuti, S.V.P.; Tettey, C.O.; Kim, D.H.; Shim, J.; Yoo, K. Ultra-sonication-assisted silver nanoparticles using Panax ginseng root extract and their anti-cancer and antiviral activities. J. Photochem. Photobiol. B Biol. 2018, 188, 6–11. [Google Scholar] [CrossRef]
  115. Liang, J.-J.; Wei, J.-C.; Lee, Y.-L.; Hsu, S.-H.; Lin, J.-J.; Lin, Y.-L. Surfactant-Modified Nanoclay Exhibits an Antiviral Activity with High Potency and Broad Spectrum. J. Virol. 2014, 88, 4218–4228. [Google Scholar] [CrossRef] [Green Version]
  116. Kumar, S.R.; Paulpandi, M.; Manivelraja, M.; Mangalaraj, D.; Viswanathan, C.; Kannan, S.; Ponpandian, N. An in vitro analysis of H1N1 viral inhibition using polymer coated superparamagnetic Fe3O4 nanoparticles. RSC Adv. 2014, 4, 13409–13418. [Google Scholar] [CrossRef]
  117. Kumar, R.; Nayak, M.; Sahoo, G.C.; Pandey, K.; Sarkar, M.C.; Ansari, Y.; Das, V.N.R.; Topno, R.K.; Bhawna; Madhukar, M.; et al. Iron oxide nanoparticles based antiviral activity of H1N1 influenza A virus. J. Infect. Chemother. 2019, 25, 325–329. [Google Scholar] [CrossRef]
  118. Ghaffari, H.; Tavakoli, A.; Moradi, A.; Tabarraei, A.; Bokharaei-Salim, F.; Zahmatkeshan, M.; Farahmand, M.; Javanmard, D.; Kiani, S.J.; Esghaei, M.; et al. Inhibition of H1N1 influenza virus infection by zinc oxide nanoparticles: Another emerging application of nanomedicine. J. Biomed. Sci. 2019, 26, 1–10. [Google Scholar] [CrossRef] [PubMed]
  119. Sametband, M.; Shukla, S.; Meningher, T.; Hirsh, S.; Mendelson, E.; Sarid, R.; Gedanken, A.; Mandelboim, M. Effective multi-strain inhibition of influenza virus by anionic gold nanoparticles. Med. Chem. Commun. 2011, 2, 421–423. [Google Scholar] [CrossRef]
  120. Papp, I.; Sieben, C.; Ludwig, K.; Roskamp, M.; Böttcher, C.; Schlecht, S.; Herrmann, A.; Haag, R. Inhibition of influenza virus infection by multivalent sialic-acid-functionalized gold nanoparticles. Small 2010, 6, 2900–2906. [Google Scholar] [CrossRef] [PubMed]
  121. Cui, H.; Jiang, J.; Gu, W.; Sun, C.; Wu, D.; Yang, T.; Yang, G. Photocatalytic inactivation efficiency of anatase nano-TiO2 sol on the H9N2 avian influenza virus. Photochem. Photobiol. 2010, 86, 1135–1139. [Google Scholar] [CrossRef] [PubMed]
  122. Thammakarn, C.; Satoh, K.; Suguro, A.; Hakim, H.; Ruenphet, S.; Takehara, K. Inactivation of avian influenza virus, Newcastle disease virus and goose parvovirus using solution of nano-sized scallop shell powder. J. Vet. Med. Sci. 2014, 76, 1277–1280. [Google Scholar] [CrossRef] [Green Version]
  123. Huo, C.; Xiao, J.; Xiao, K.; Zou, S.; Wang, M.; Qi, P.; Liu, T.; Hu, Y. Pre-treatment with zirconia nanoparticles reduces inflammation induced by the pathogenic H5N1 influenza virus. Int. J. Nanomed. 2020, 15, 661–674. [Google Scholar] [CrossRef] [Green Version]
  124. Qin, T.; Ma, R.; Yin, Y.; Miao, X.; Chen, S.; Fan, K.; Xi, J.; Liu, Q.; Gu, Y.; Yin, Y.; et al. Catalytic inactivation of influenza virus by iron oxide nanozyme. Theranostics 2019, 9, 6920–6935. [Google Scholar] [CrossRef]
  125. Belshe, R.B.; Burk, B.; Newman, F.; Cerrutie, R.L.; Sim, I.S. Resistance of influenza A virus to amantadine and rimantadine: Results of the one decade of surveillance. J. Infect. Dis. 1989, 159, 430–435. [Google Scholar] [CrossRef]
  126. Influenza (Flu) Antiviral Drugs and Related Information—Get Information on Medicines for the Flu. Available online: https://www.fda.gov/drugs/information-drug-class/influenza-flu-antiviral-drugs-and-related-information (accessed on 28 September 2020).
  127. Cheng, P.K.C.; Leung, T.W.C.; Ho, E.C.M.; Leung, P.C.K.; Ng, A.Y.Y.; Lai, M.Y.Y.; Lim, W.W.L. Oseltamivir-and amantadine-resistant influenza viruses A (H1N1). Emerg. Infect. Dis. 2009, 15, 966–968. [Google Scholar] [CrossRef]
  128. Zaraket, H.; Saito, R.; Suzuki, Y.; Baranovich, T.; Dapat, C.; Caperig-Dapat, I.; Suzuki, H. Genetic makeup of amantadine-resistant and oseltamivir-resistant human influenza A/H1N1 viruses. J. Clin. Microbiol. 2010, 48, 1085–1092. [Google Scholar] [CrossRef] [Green Version]
  129. Mishin, V.P.; Hayden, F.G.; Gubareva, L.V. Susceptibilities of antiviral-resistant influenza viruses to novel neuraminidase inhibitors. Antimicrob. Agents Chemother. 2005, 49, 4515–4520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Sagandira, C.R.; Mathe, F.M.; Guyo, U.; Watts, P. The evolution of Tamiflu synthesis, 20 years on: Advent of enabling technologies the last piece of the puzzle? Tetrahedron 2020, 76, 131440. [Google Scholar] [CrossRef] [PubMed]
  131. Cheng, L.P.; Wang, T.C.; Yu, R.; Li, M.; Huang, J.W. Design, synthesis and biological evaluation of novel zanamivir derivatives as potent neuraminidase inhibitors. Bioorg. Med. Chem. Lett. 2018, 28, 3622–3629. [Google Scholar] [CrossRef] [PubMed]
  132. European Medicines Agency. Alpivab (Peramivir). Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/alpivab (accessed on 28 September 2020).
  133. Ison, M.G.; Hayden, F.G. Antiviral Agents Against Respiratory Viruses, 4th ed.; Elsevier Ltd.: Amsterdam, The Netherlands, 2017. [Google Scholar]
  134. Heo, Y.A. Baloxavir: First Global Approval. Drugs 2018, 78, 693–697. [Google Scholar] [CrossRef] [PubMed]
  135. O’Hanlon, R.; Shaw, M.L. Baloxavir marboxil: The new influenza drug on the market. Curr. Opin. Virol. 2019, 35, 14–18. [Google Scholar] [CrossRef] [PubMed]
  136. Li, Y.; Lin, Z.; Guo, M.; Xia, Y.; Zhao, M.; Wang, C.; Xu, T.; Chen, T.; Zhu, B. Inhibitory activity of selenium nanoparticles functionalized with oseltamivir on H1N1 influenza virus. Int. J. Nanomed. 2017, 12, 5733–5743. [Google Scholar] [CrossRef] [Green Version]
  137. Lin, Z.; Li, Y.; Guo, M.; Xiao, M.; Wang, C.; Zhao, M.; Xu, T.; Xia, Y.; Zhu, B. Inhibition of H1N1 influenza virus by selenium nanoparticles loaded with zanamivir through p38 and JNK signaling pathways. RSC Adv. 2017, 7, 35290–35296. [Google Scholar] [CrossRef] [Green Version]
  138. Li, Y.; Lin, Z.; Guo, M.; Zhao, M.; Xia, Y.; Wang, C.; Xu, T.; Zhu, B. Inhibition of H1N1 influenza virus-induced apoptosis by functionalized selenium nanoparticles with amantadine through ROS-mediated AKT signaling pathways. Int. J. Nanomed. 2018, 13, 2005–2016. [Google Scholar] [CrossRef] [Green Version]
  139. Lin, Z.; Li, Y.; Gong, G.; Xia, Y.; Wang, C.; Chen, Y.; Hua, L.; Zhong, J.; Tang, Y.; Liu, X.; et al. Restriction of H1N1 influenza virus infection by selenium nanoparticles loaded with ribavirin via resisting caspase-3 apoptotic pathway. Int. J. Nanomed. 2018, 13, 5787–5797. [Google Scholar] [CrossRef] [Green Version]
  140. Li, Y.; Lin, Z.; Gong, G.; Guo, M.; Xu, T.; Wang, C.; Zhao, M.; Xia, Y.; Tang, Y.; Zhong, J.; et al. Inhibition of H1N1 influenza virus-induced apoptosis by selenium nanoparticles functionalized with arbidol through ROS-mediated signaling pathways. J. Mater. Chem. B 2019, 7, 4252–4262. [Google Scholar] [CrossRef]
  141. Li, Y.; Lin, Z.; Zhao, M.; Guo, M.; Xu, T.; Wang, C.; Xia, H.; Zhu, B. The reversal of H1N1 influenza virus-induced apoptosis by silver nanoparticles functionalized with Amantadine. RSC Adv. 2016, 6, 89679–89686. [Google Scholar] [CrossRef]
  142. Li, Y.; Lin, Z.; Zhao, M.; Xu, T.; Wang, C.; Hua, L.; Wang, H.; Xia, H.; Zhu, B. Silver Nanoparticle Based Codelivery of Oseltamivir to Inhibit the Activity of the H1N1 Influenza Virus through ROS-Mediated Signaling Pathways. ACS Appl. Mater. Interfaces 2016, 8, 24385–24393. [Google Scholar] [CrossRef] [PubMed]
  143. Lin, Z.; Li, Y.; Guo, M.; Xu, T.; Wang, C.; Zhao, M.; Wang, H.; Chen, T.; Zhu, B. The inhibition of H1N1 influenza virus-induced apoptosis by silver nanoparticles functionalized with zanamivir. RSC Adv. 2017, 7, 742–750. [Google Scholar] [CrossRef] [Green Version]
  144. Bimbo, L.M.; Denisova, O.V.; Makila, E.; Kaasalainen, M.; De Brabander, J.K.; Hirvonen, J.; Salonen, J.; Kakkola, L.; Kainov, D.; Santos, A.H. Inhibition of Influenza A Virus Infection in Vitro by Saliphenylhalamide-Loaded Porous Silicon Nanoparticles. ACS Nano 2013, 7, 6884–6893. [Google Scholar] [CrossRef]
  145. Chakravarty, M.; Vora, A. Nanotechnology-based antiviral therapeutics. Drug Deliv. Transl. Res. 2020. [Google Scholar] [CrossRef]
  146. Rai, M.; Deshmukh, S.D.; Ingle, A.P.; Gupta, I.R.; Galdiero, M.; Galdiero, S. Metal nanoparticles: The protective nanoshield against virus infection. Crit. Rev. Microbiol. 2016, 42, 46–56. [Google Scholar] [CrossRef]
  147. Cagno, V.; Andreozzi, P.; D’Alicarnasso, M.; Silva, P.J.; Mueller, M.; Galloux, M.; Goffic, R.L.; Jones, S.T.; Vallino, M.; Hodek, J.; et al. Broad-spectrum non-toxic antiviral nanoparticles with a virucidal inhibition mechanism. Nat. Mater. 2018, 17, 195–203. [Google Scholar] [CrossRef]
  148. Jones, A.A.D.; Mi, G.; Webster, T.J. A Status Report on FDA Approval of Medical Devices Containing Nanostructured Materials. Trends Biotechnol. 2019, 37, 117–120. [Google Scholar] [CrossRef] [Green Version]
  149. Cojocaru, F.D.; Botezat, D.; Gardikiotis, I.; Uritu, C.M.; Dodi, G.; Trandafir, L.; Rezus, C.; Rezus, E.; Tamba, B.I.; Mihai, C.T. Nanomaterials designed for antiviral drug delivery transport across biological barriers. Pharmaceutics 2020, 12, 171. [Google Scholar] [CrossRef] [Green Version]
  150. Abo-Zeid, Y.; Ismail, N.S.M.; McLean, G.R.; Hamdy, N.M. A molecular docking study repurposes FDA approved iron oxide nanoparticles to treat and control COVID-19 infection. Eur. J. Pharm. Sci. 2020, 153, 105465. [Google Scholar] [CrossRef]
Figure 1. Scheme of influenza A virion structure. The virion surface is encrusted with the membrane proteins (hemagglutinin and nucleoprotein) and M2 proton channel proteins. The inner side of the virion is overlaid with M1 matrix protein. The 8 vRNP complexes and multiple copies of nuclear export protein are located in the virion interior.
Figure 1. Scheme of influenza A virion structure. The virion surface is encrusted with the membrane proteins (hemagglutinin and nucleoprotein) and M2 proton channel proteins. The inner side of the virion is overlaid with M1 matrix protein. The 8 vRNP complexes and multiple copies of nuclear export protein are located in the virion interior.
Pathogens 09 01020 g001
Figure 2. Schematic representation of different nanotechnology applications in vaccination strategies. The main goals are the stimulation of antibody (Ab) production and lymphocyte (T cell) activation. For this purpose, different vaccination strategies were used: viral antigens immobilized on nanoparticles’ (NPs) surfaces (A); viral self-amplifying mRNA encapsulated in NPs, which replicate inside cells, leading to antigen expression (B); or inactivated virus encapsulated in NPs (C).
Figure 2. Schematic representation of different nanotechnology applications in vaccination strategies. The main goals are the stimulation of antibody (Ab) production and lymphocyte (T cell) activation. For this purpose, different vaccination strategies were used: viral antigens immobilized on nanoparticles’ (NPs) surfaces (A); viral self-amplifying mRNA encapsulated in NPs, which replicate inside cells, leading to antigen expression (B); or inactivated virus encapsulated in NPs (C).
Pathogens 09 01020 g002
Figure 3. Schematic view of nanoparticle (NP) applications in gene silencing strategies. The NPs functionalized with oligonucleotides are used as a delivery system to the specific target locations in cells. Different oligonucleotide strategies are used to inhibit virus replication: blocking of accessible ssRNA regions with antisense oligonucleotides (ASOs) complementary to viral RNA (A); targeted degradation of viral mRNA with siRNAs (B); catalytic hydrolysis of viral RNA with DNAzymes designed to target viral RNA (C).
Figure 3. Schematic view of nanoparticle (NP) applications in gene silencing strategies. The NPs functionalized with oligonucleotides are used as a delivery system to the specific target locations in cells. Different oligonucleotide strategies are used to inhibit virus replication: blocking of accessible ssRNA regions with antisense oligonucleotides (ASOs) complementary to viral RNA (A); targeted degradation of viral mRNA with siRNAs (B); catalytic hydrolysis of viral RNA with DNAzymes designed to target viral RNA (C).
Pathogens 09 01020 g003
Figure 4. Schematic representation of anti-influenza drugs delivery via nanoparticles (A–F). Arbidol inhibits the formation of hemagglutinin-sialic acid bonding, and thereby blocks the viral entry (A). Amantadine blocks the viral M2 ion channel in order to disturb the uncoating process (B). Saliphenylhalamide blocks the acidification of endosomes and vRNP releasing (C). Ribavirin inhibits RdRp, thus disturbing vRNP formation and mRNA maturation processes (D). Oseltamivir and zanamivir are neuraminidase inhibitors, which blocks the release of the progeny virions (E and F, respectively). Influenza virus life cycle (1–11). Interaction between HA and sialic-acid followed by membrane fusion and viral entry to the cell (1). Endosome formation (2). Uncoating and vRNP releasing to the cytoplasm after pH changes catalyzed by M2 ion channel (3). vRNP transport to the nucleus (4). Genome replication and mRNA synthesis (5). mRNA maturation catalyzed by RdRp (6). Translation of viral proteins by ER and cytosolic ribosomes (7). Maturation of viral proteins at Golgi (8) followed by transport of the proteins to the nucleus for vRNP formation (9). Transport of vRNP and viral proteins to the cellular membrane for assembly and budding (10). Releasing of the progeny virions (11).
Figure 4. Schematic representation of anti-influenza drugs delivery via nanoparticles (A–F). Arbidol inhibits the formation of hemagglutinin-sialic acid bonding, and thereby blocks the viral entry (A). Amantadine blocks the viral M2 ion channel in order to disturb the uncoating process (B). Saliphenylhalamide blocks the acidification of endosomes and vRNP releasing (C). Ribavirin inhibits RdRp, thus disturbing vRNP formation and mRNA maturation processes (D). Oseltamivir and zanamivir are neuraminidase inhibitors, which blocks the release of the progeny virions (E and F, respectively). Influenza virus life cycle (1–11). Interaction between HA and sialic-acid followed by membrane fusion and viral entry to the cell (1). Endosome formation (2). Uncoating and vRNP releasing to the cytoplasm after pH changes catalyzed by M2 ion channel (3). vRNP transport to the nucleus (4). Genome replication and mRNA synthesis (5). mRNA maturation catalyzed by RdRp (6). Translation of viral proteins by ER and cytosolic ribosomes (7). Maturation of viral proteins at Golgi (8) followed by transport of the proteins to the nucleus for vRNP formation (9). Transport of vRNP and viral proteins to the cellular membrane for assembly and budding (10). Releasing of the progeny virions (11).
Pathogens 09 01020 g004
Table 1. Nanotechnology-based anti-influenza vaccines.
Table 1. Nanotechnology-based anti-influenza vaccines.
StrategyNPs Material/SizeInfluenza Virus Strain (Subtype)In Vitro/in Vivo StudiesVaccineRef.
HA as an viral antigenAu NPs/18 nmA/Aichi/2/1968 (H3N2)JAWS II, BMDC/miceIN[40]
A/Aichi/2/1968 (H3N2)HEK 293/miceIN[41]
CaP NPs/250 nmA/Puerto Rico/8/1934 (H1N1)Splenocytes, dendritic cells,
T cells/mice
SC, IN[42]
-Splenocytes, dendritic cells,
T cells/-
-[43]
chitosan NPs/300 nmA/Brisbane/59/2007(H1N1)-/miceIN[44]
ferritin NPs/37 nmA/New Caledonia/20/1999 (H1N1), A/Puerto Rico/8/1934 (H1N1), A/Singapore/6/1986 (H1N1), A/Brisbane/59/2007 (H1N1), A/California/04/2009 (H1N1), A/Beijing/262/1995 (H1N1),
A/Solomon Islands/3/2006 (H1N1),
A/Perth/16/2009 (H3N2), B/Florida/04/2006
-/mice, ferretsSC[45]
ferritin NPs/no dataA/New Caledonia/20/1999 (H1N1), A/South Carolina/1/1918 (H1N1), A/California/04/2009 (H1N1), A/Singapore/6/1986 (H1N1), A/Canada/720/2005 (H2N2), A/Vietnam/1203/2004 (H5N1), A/Indonesia/5/2005 (H5N1), A/Anhui/1/2013 (H7N9),
A/Hong Kong/1074/1999 (H9N2)
-/mice, ferretsSC[46]
polyanhydride NPs/200 nmA/Whooper Swan/Mongolia/244/2005 (H5N1), A/Vietnam/1203/2004 (H5N1) -/miceSC, IN[47]
polyanhydride NPs/200 nmVNH5N1-PR8/CDC-RG (a reassortant virus strain containing the HA gene with a modified basic amino acid cleavage site) segment, the neuraminidase gene segment from the A/Vietnam/1203/2004 (H5N1) virus and the six internal gene segments of A/Puerto Rico/8/1934 (H1N1) virus generated by reverse genetics technology)-/miceSC[48]
polyanhydride NPs/200 nmA/Vietnam/1203/2004 (H5N1), A/Indonesia/5/2005 (H5N1), A/Hong Kong/482/1997 (H5N1),
A/Chicken/Qalubia-Egypt/1/2008 (H5N1),
A/northern pintail/Alaska/622/2012 (H5N2),
A/northern pintail/Alaska/622/2012 (H5N2),
A/American green-winged teal/Alaska/472/2014 (H5N2), recombinant A/Puerto Rico/8/1934 (H1N1), recombinant A/chicken/Vietnam/NCVD5/2003 (H5N1)
-/chickensSC[49]
Au NPs/12 nmA/Puerto Rico/8/1934 (H1N1)-/miceIN[50]
IN[51]
A/California/04/2009 (H1N1), A/Victoria/3/1975 (H3N2), A/Vietnam/1203/2004 (H5N1)MDCK/miceIN[52]
VLPs/30 nmno data-/miceSC[53]
VLPs/no datamouse adapted recombinant X47virus (H3N2) (A/Victoria/3/1975 (H3N2) x A/PuertoRico/8/1934 (H1N1))-/miceIN[54]
mouse adapted recombinant X47virus(H3N2) (A/Victoria/3/1975 (H3N2) x A/PuertoRico/8/1934 (H1N1))-/miceSC[55]
mix of virus membrane proteins as a viral antigenchitosan NPs/140 nmA/California/7/2009 (H1N1)RAW 264.7/
mice
IN[56]
VLPs/114 nmA/Taiwan/S02076/2013 (H7N9)-/mice, chickensSC[57]
polyanhydride NPs/200 nm A/Puerto Rico/8/1934 (H1N1)-/miceSC[58]
PLGA NPs/260 nmA/South Carolina/1/1918 (H1N1),
A/New Caledonia/20/1999 (H1N1)
-/pigsIN, IT[59]
inactivated virus strategychitosan NPs/581 nmA/New Caledonia/20/1999 (H1N1)--[60]
A/New Caledonia/20/1999 (H1N1)-/rabbitsSC, IN[61]
polyanhydride NPs/200 nmA/Swine/Ohio/24366/2007 (H1N1), A/Swine/Ohio/FAH10-1/2010 (H1N2)-/pigsIN[62]
CaP NPs/450–500 nmA/California/7/2009 (H1N1)MDCK/miceIM[63]
PLGA NPs/624 nm
chitosan@PLGA NPs/819 nm mannan@PLGA NPs/719 nm
A/Duck/Czech/1956 (H4N6)chickenMC, SC[64]
mRNA strategylipid NPs/
no data
A/Northern Territory/60/1968 (H3N2)-/miceIJ[65]
lipid NPs/80–100 nmA/Anhui/1/2013 (H7N9),
A/Jiangxi-Donghu/346/2013 (H10N8)
-/mice, ferrets, cynomolgus monkeys (cynos)IJ[66]
lipid NPs/130–142 nmA/Puerto Rico/8/1934 (H1N1),
mouse-adapted A/HongKong/1/1968 (H3N2), A/California/7/2009 (H1N1)
BHK/miceIM[67]
lipid NPs/~80 nmRecombinant IAV with: the HA and NA from A/Singapore/GP1908/2015 (H1N1) and remaining proteins from A/Texas/1/1977 (H3N2), A/Michigan/45/2015 (H1N1), A/New Caledonia/20/1999 (H1N1) and A/Puerto Rico/8/1934 (H1N1). Recombinant chimeric IAV with: HA head domain from A/mallard/Sweden/81/2002 (H6N1), HA stalk domain from A/California/04/2009 (H1N1), NA from A/mallard/Sweden/86/2003 (H12N5) and remaining proteins from A/Puerto Rico/8/1934 (H1N1).
Recombinant IAV with the polybasic cleavage site removed from the A/Vietnam/1203/2004 (H5N1), the N8 from A/mallard/Sweden/50/2002 (H3N8) and remaining proteins from A/Puerto Rico/8/1934 (H1N1). Recombinant IAV with: the HA and NA from A/chicken/Hong Kong/G9/1997 (H9N2) and remaining proteins from A/Puerto Rico/8/1934 (H1N1) and A/Hong Kong/4801/2014 (H3N2).
NIH-3T3, MDCK, HEK293T/
mice
IJ[68]
BMDCs: bone marrow-derived dendritic cells; HEK: human embryonic kidney 293 cells; “-”: not tested; MDCK: Madin-Darby canine kidney cells; BHK: baby hamster kidney cells; ID: intradermal; IM: intramuscular; IJ: injections; IN: intranasal; IT: intratracheal; MC: mucosal; SC: subcutaneous.
Table 2. Anti-influenza gene silencing strategies based on nanotechnology.
Table 2. Anti-influenza gene silencing strategies based on nanotechnology.
StrategyNPs Material/SizeInfluenza Virus Strain (Subtype)In Vitro/In Vivo StudiesVaccineRef.
antisense
oligonucleotides
TiO2 NPs/
5 nm
A/Aichi/2/1968 (H3N2)MDCK/--[97]
A/Aichi/2/1968 (H3N2)MDCK/--[98]
A/Salekhard/01/2009 (H1N1), Aichi/2/68 (H3N2),
A/chicken/Kurgan/05/2005(H5N1)
MDCK/--[99]
A/Salekhard/01/2009 (H1N1), Aichi/2/68 (H3N2),
A/chicken/Kurgan/05/2005(H5N1)
MDCK/--[100]
TiO2 NPs/
4–6 nm
Aichi/2/1968 (H3N2)MDCK/--[101]
Si-NH2 NPs/
1.2–1.5 nm
A/chicken/Kurgan/05/2005 (H5N1)MDCK/--[102]
siRNACaP NPs/
145 nm
A/Puerto Rico/8/1934 (H1N1)-/miceIN[103]
chitosan NPs/278 nmA/Puerto Rico/8/1934 (H1N1)Vero/miceIN[104]
DNAzymesTiO2 NPs/5 nmA/chicken/Kurgan/05/2005 (H5N1) MDCK, HeLa/--[105]
MDCK: Madin-Darby canine kidney cells; “-”: not tested; IN: intranasal.
Table 3. Bare nanoparticles as new anti-influenza therapeutics.
Table 3. Bare nanoparticles as new anti-influenza therapeutics.
StrategyNPs Material/SizeInfluenza Virus Strain (Subtype)In Vitro/In Vivo StudiesVaccineRef.
silver NPsAg NPs/no dataA/New Caledonia/20/1999 (H1N1)MDCK/--[109]
Ag NPs/10 nmA/Puerto Rico/8/1934 (H1N1)MDCK/--[110]
Ag NPs/10 nmA/Human/Hubei/3/2005 (H3N2)MDCK/miceIN[111]
Ag NPs/4–13 nmA/Puerto Rico/8/1934 (H1N1)MDCK/--[112]
Ag NPs/42 nmno data (H7N3)Vero/--[113]
Ag NPs/
5–15 nm
A/Puerto Rico/8/1934 (H1N1)MDCK/--[114]
silicate platelet@Ag NPs/
80 × 80 × 1 nm
A/WSN/1933 (H1N1)MDCK, A549/--[115]
iron oxide NPsFe3O4 NPs/8–10 nmA/2009 (H1N1)MDCK/--[116]
Fe3O4 NPs/8–10 nmA/Puerto Rico/8/1934 (H1N1)MA104/--[117]
zinc oxide NPsZnO NPs/16–50 nmA/Puerto Rico/8/1934 (H1N1)MDCK/--[118]
gold NPsAu NPs/5 nmA/Israel/119/2009 (H1N1), A/Brisbane/10/2007 (H3N2), A/Puerto Rico/8/1934 (H1N1), B/Brisbane/60/2008, B/Shandong/7/1997MDCK/--[119]
sialic-acid@Au NPs/2 and 14 nmA/X-31 (H3N2)MDCK, CHO/--[120]
titanium dioxide NPsTiO2 NPs/50 nmno data (H9N2)MDCK/--[121]
calcium oxide NPsCaO NPs/500 nmA/duck/Aomori/395/2004 (H7N1)MDCK/--[122]
zirconium dioxide NPsZrO2 NPs/200 nmA/chicken/Henan/1/2004 (H5N1)-/miceIN[123]
MDCK: Madin-Darby canine kidney cells; “-”: not tested; CHO: chinese hamster ovary; IN: intranasal.
Table 4. Delivery of anti-influenza drugs by nanocarriers.
Table 4. Delivery of anti-influenza drugs by nanocarriers.
DrugNPs Material/SizeInfluenza Virus Strain (Subtype)In Vitro/In Vivo StudiesVaccineRef.
oseltamivirSe NPs/
70–200 nm
no data (H1N1)MDCK/--[136]
zanamivirA/Hubei/74/2009 (H1N1)MDCK/--[137]
ribavirinno data (H1N1)MDCK/--[138]
amantadineA/Guangdong/1/2009 (H1N1)MDCK/miceIN[139]
arbidolno data (H1N1)MDCK/miceIN[140]
amantadineAg NPs/
2–3 nm
no data (H1N1)MDCK/--[141]
oseltamivirno data (H1N1)MDCK/--[142]
zanamivirA/Hubei/74/2009 (H1N1)MDCK/--[143]
saliphenylhalamideTHCPSi NPs/129 nmA/WSN/1933 (H1N1),
A/Puerto Rico/8/1934-NS116-GFP (H1N1)
MDCK, monkey Vero-E6/--[144]
MDCK: Madin-Darby canine kidney cells; “-”: not tested; IN: intranasal.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wieczorek, K.; Szutkowska, B.; Kierzek, E. Anti-Influenza Strategies Based on Nanoparticle Applications. Pathogens 2020, 9, 1020. https://doi.org/10.3390/pathogens9121020

AMA Style

Wieczorek K, Szutkowska B, Kierzek E. Anti-Influenza Strategies Based on Nanoparticle Applications. Pathogens. 2020; 9(12):1020. https://doi.org/10.3390/pathogens9121020

Chicago/Turabian Style

Wieczorek, Klaudia, Barbara Szutkowska, and Elzbieta Kierzek. 2020. "Anti-Influenza Strategies Based on Nanoparticle Applications" Pathogens 9, no. 12: 1020. https://doi.org/10.3390/pathogens9121020

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop