A Perspective on Nanoparticle Universal Influenza Vaccines - ACS

Nov 5, 2018 - In addition, the seasonal influenza vaccines are ineffective against an unpredictable pandemic. A universal influenza vaccine would over...
0 downloads 0 Views 1019KB Size
Subscriber access provided by The University of Texas at El Paso (UTEP)

Review

A perspective of nanoparticle universal influenza vaccines Lei Deng, and Bao-Zhong Wang ACS Infect. Dis., Just Accepted Manuscript • DOI: 10.1021/acsinfecdis.8b00206 • Publication Date (Web): 05 Nov 2018 Downloaded from http://pubs.acs.org on November 6, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

A perspective of nanoparticle universal influenza vaccines Authors: Lei Deng 1, Bao-Zhong Wang 1 * Author Affiliations: 1

Center for Inflammation, Immunity & Infection, Georgia State University, 145 Piedmont

Ave SE, Atlanta, GA, USA * Correspondence to Dr. Bao-Zhong Wang (E-mail: [email protected])

Annual influenza infections cause massive economic loss and pose severe threats to public health worldwide. Seasonal influenza vaccines are the most effective means of preventing influenza infections, but they still possess major weaknesses. Seasonal influenza vaccines require annual updating of the vaccine strains and it is difficult to predict future circulating strains accurately. This forecasting of future circulating strains is necessary because of the slow process of producing the vaccine in chicken eggs. When this forecast is inaccurate, the seasonal vaccine provides little protection efficacy against the circulating strains, especially novel and unpredictable pandemic strains. Keywords: prophylaxis; virus like particle; self-assembling; long lasting; headless hemagglutinin, matrix protein 2

A universal influenza vaccine that could be used from year to year would overcome the weaknesses of the seasonal vaccine and abolish the threat of influenza pandemics. One approach under investigation is to design influenza vaccine immunogens based on conserved, type-specific amino acid sequences and conformational epitopes, rather than strain-specific. Such vaccines can elicit broadly reactive humoral and cellular immunity.

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 34

Universal influenza vaccine development has intensively employed nanotechnology because the structural and morphological properties of nanoparticles dramatically improve vaccine immunogenicity and the induced immunity duration. Layered protein nanoparticles can eliminate the possibility of off-target immune responses, fine-tune antigen-recognition and processing, and facilitate comprehensive immune response induction. Herein, we reviewed the designs of effective nanoparticle universal influenza vaccines, the recent discoveries of specific nanoparticle features that contribute to immunogenicity enhancement, and recent progress in clinical trials.

1. An Urgent Need for an Affordable Universal Influenza Vaccine. Seasonal influenza causes 3 to 5 million cases of severe illness and up to 650,000 deaths yearly according to the World Health Organization.

1

Mismatched seasonal influenza

vaccines provide limited protection against circulating strains. For instance, by midOctober in 2017 the influenza vaccine effectiveness against H3N2 was estimated to be only 10%, which was closely associated with the predominant H3N2 activity in the southern hemisphere.

2

Unsurprisingly, vaccination with this vaccine did not counteract

the H3N2-predominant epidemic in the United States during the following months.

3

During the 2017-2018 influenza season, the severity of influenza B outbreaks in some areas of the northern hemisphere was associated with the lack of a vaccine strain from the influenza B Yamagata lineage in the traditional trivalent influenza vaccine. 4, 5

Influenza viruses contain three membrane proteins: hemagglutinin (HA), neuraminidase (NA), and matrix protein 2 (M2) (Figure 1).

6

Induction of protective immunity against

ACS Paragon Plus Environment

Page 3 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

influenza requires recognition of these surface proteins by the host. M2 is relatively genetically stable and possess low immunogenicity. HA and NA are much more immunogenic but also highly antigenically variable. There are 18 HA subtypes and 11 NA subtypes known for influenza A.

7

Antigenic drift (i.e., mutations in HA or NA) results in

new influenza A virus strains over time. Antigenic shift (i.e., a major re-assortment of HA or NA genes) results in new subtypes of influenza A virus. As a result, circulating influenza viruses stay in a continuous state of genetic flux and display different variants of HA and NA on their surfaces, which enables the evasion of preexisting immunity. A non-human influenza virus may also cause a pandemic in human populations by acquiring the capacity for transmission in humans. The recent infection of humans by the highly pathogenic avian H5N1 and the outbreak of a novel avian H7N9 strain has reinforced this concern.

8, 9

The inherent variability of HA and NA surface proteins of influenza virus

creates an intractable problem for the seasonal influenza vaccine approach. For this reason, there is a need for a universal influenza vaccine that will induce broad crossprotection against divergent influenza viruses.

2. Conserved Influenza Sequences as Universal Vaccine Immunogens The suboptimal vaccine effectiveness of conventional influenza vaccines is multifactorial. Lowered vaccine effectiveness is associated with previous vaccinations with seasonal influenza vaccines

10-12

and with increasing age of the recipients.

13

Vaccinologists have

attempted to improve the efficacy of the seasonal vaccine through different means. For example, increasing the dosage of conventional influenza vaccination induced higher vaccine-specific antibody titers and interleukin - 10 levels, but has little positive impact on

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 34

the development of functional T-cell memory in older adults. 14 As well, using insect cellproduced HA in seasonal influenza vaccines can restore the reduced immunogenicity caused by egg-adaptation in influenza viruses.

15

In another approach, researchers

created a novel influenza vaccine strain by introducing eight interferon-sensitive mutations — point mutations identified through quantitative high-throughput genomics analysis — and found improved in vivo immunogenicity and protectivity. 16 Nevertheless, a universal influenza vaccine is still preferable because it can be used without a yearly update of vaccine components.

A universal influenza vaccine would rely on conserved amino acid sequences and epitope conformations from influenza to generate broadly reactive humoral and cellular immunity. M2 is a relatively conserved influenza antigen, but it is small compared to HA and NA and is expressed in low copy numbers on the virion. However, infected host cells abundantly express M2 on their surfaces where it is accessible for antibodies.

17

The protection

against multiple influenza A virus subtypes elicited by M2 ectodomain (M2e)-based vaccines correlates closely with M2e-specific antibody affinity to Fc gamma receptors on target cells. 18-21

The HA stalk domain is more amino acid- and conformation-conserved than the HA head domain and can induce antibodies capable of binding multiple HA subtypes. 22-28 Protein structural analysis has enabled the development of some recombinant HA stalk domain vaccines with effective in vivo immunogenicity. 29-31 HA stalk-directed protective immunity prevents infection by inducing neutralizing antibodies, antibody-dependent cellular

ACS Paragon Plus Environment

Page 5 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

cytotoxicity, and antibody-dependent phagocytosis. 31 T cell responses against influenza structural components correlate with cross-reactive protection and early recovery from infection. 32-34

Nucleoproteins (NP) enclose the viral RNA segments, are necessary for trafficking the viral genome and are highly conserved in comparison to the surface glycoproteins.

35, 36

Blending NP with other conserved influenza components like polymerase basic-2 or matrix protein 1 (M1) in fusion constructs broadened the spectrum of immune protection induced by vaccine candidates. 37-41

Vaccination with multiple conserved target antigens has advantages over using single antigens.

42

A universal vaccine providing long-term protection against heterosubtypic

influenza virus strains will benefit pandemic control and routine vaccination.

3. Different Types of Nanoparticles Accommodating Influenza Conserved Epitopes Benefit for Enhanced Immunogenicity Conserved influenza antigens typically induce weak immune responses. There is a need to develop novel delivery systems and adjuvants that increase the immunogenicity of conserved components for next-generation vaccines. The application of nanotechnology in vaccinology has been increasing dramatically over the past decades.

43

The common

benefits of nanoparticles are prolonged circulation and directed targeting by modification of nanoparticle surfaces with dendritic cell or T cell targeting ligands. 44 Nanoparticles are

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 34

an efficient delivery system which facilitates the enhancement of immune cell uptake and sustained antigen release. 45, 46

Our recent study indicated that gradual intracellular disassembly of protein nanoparticles led to sustained specific immune responses.

47

Physiologically controlled, stimuli-

sensitive release is a major benefit of this nano system. An optimized eudragitS/Trehalose polymer nanoparticle influenza vaccine released influenza antigens during in vitro simulation of the pH conditions of the mouse gastrointestinal tract. Mice experiments indicated that oral immunization with this nanoparticle vaccine-induced protective immune responses.

48

Thermo-sensitive, redox condition-sensitive, and near-

infrared light activated controlled release has recently been developed in the field of anticancer research.

44, 49

These features of different nanoparticle types can benefit

prophylactic universal influenza vaccine designs (Figure 2). One or multiple features can be synergistically integrated into one nanoparticle system for an optimal immune response. Nanoparticle universal influenza vaccines under investigation are summarized in Table 1. i.

Polymer Nanoparticles. Synthetic polymers are widely used to fabricate

nanoparticle vaccines due to their favorable properties such as biocompatibility and biodegradability. US Food and Drug Administration approved poly(D,L-lactic-co-glycolic acid) (PLGA) has been extensively characterized in animal models and is widely used in vaccine development as an encapsulation matrix

50

to co-deliver

51, 52

and sustain

controlled release of antigens. 46, 53 Other synthetic polymers with similar features include

ACS Paragon Plus Environment

Page 7 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

poly(D, L-lactide-co-glycolide),

54

poly(D,L-lactic-co-hydroxymethyl glycolic acid)

55

and

polystyrene. 56

In universal influenza vaccine development, PLGA nanoparticles encapsulating conserved influenza A virus components including norovirus P particle containing M2e and synthetic peptides of HA, NP and polymerase acidic protein, elicited strong prophylactic protection and eliminated influenza symptoms in pigs against swine H1N1 challenge,

57

which significantly improves upon the previously reported immunization

strategy with M2e and NP antigens in pigs.

58

PLGA can be formulated into larger sizes

and facilitate the release of nanometer-sized constructs. Recombinant outer membrane vesicles displaying M2e released from PLGA microparticles over 30 days, induced sustained protective M2e specific immunity in mice. 59

Natural polymers based on polysaccharides have also been used to prepare nanoparticles, such as pullulan, alginate, inulin, and chitosan.

43

Non-toxic chitosan

nanoparticles have been widely studied as vaccine vectors owing to its advantages, biocompatibility, biodegradability, and amenability to size and shape modifications.

60-62

Poly-γ-glutamic acid/chitosan nanoparticles containing truncated M2 and a fusion peptide of influenza HA and mucosal adjuvant cholera toxin subunit A1 acted as an effective mucosal vaccine against diverse influenza A viruses. 63 Nanogel particles encapsulating pandemic H1N1 split vaccine antigen increased cross-protection against homologous and heterosubtypic influenza A viral infection. 64 A diversity of biocompatible polymers for nanoparticles expand our ability to accommodate different immunogens and adjuvants.

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 34

However, because a major portion of the vaccine-loaded nanoparticles is the polymer materials, the immunogen load of the polymer nanoparticles is limited, which could be a limitation when a high dosage is required for a robust immune response.

ii.Virus-like particles. Virus-like particle (VLP) formation is driven by the same mechanisms used during virus assembly — viral self-assembling proteins or domains. The morphological features of VLPs simulate the features of viruses that the host immune system has evolved to combat, imbuing VLPs with high immunogenicity. 65-68

The first VLP vaccine for hepatitis B virus, developed by Glaxo Smith Kline, was commercialized in 1986 69 followed by commercialization of other VLP vaccines including Cervarix (human papillomavirus, Glaxo Smith Kline), Recombivax HB (hepatitis B virus, Merck & Co. Inc) and Gardasil (human papillomavirus, Merck & Co. Inc). Many others are currently in the clinical trial or research stage.

Because VLPs lack genomic components or contain premature termination codons,

70

they are safer compared with other replicating vaccine vectors. Due to viral features like repetitive structures, naturally retained antigen conformation, and virion sizes, VLPs induce innate and adaptive immune responses. 71, 72 Influenza lacks a capsid. Instead, it has a core connected to the viral envelop by the matrix protein M1. VLPs are assembled in the physiological conditions on host membranes and released into the environment via budding. Our influenza VLPs are formed in SF9 insect cells by the same M1-organized budding process that forms the influenza envelope. Enveloped VLPs can be produced by

ACS Paragon Plus Environment

Page 9 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

co-expression of structural proteins in mammalian cells, insect cells, or plants.

73-79

Furthermore, the self-assembling biocompatible viral capsid proteins can be adapted to various cell systems, like mammalian cells, insect cells, plants, yeasts and Escherichia coli (E. coli) for VLP production. 80-82

A vaccine incorporating A/Puerto Rico/8/1934 (PR8) headless HA into human immunodeficiency virus Gag-based VLPs expressed in 293T cells protected mice against influenza viral challenges. 83 M2 VLPs have been produced in insect cells coinfected with recombinant baculoviruses expressing M1 and wild-type M2 protein 84 or multiple M2e. 85 Supplemental M2 VLP immunization with inactivated H1N1 vaccine-enhanced crossprotection against influenza viral challenges.

84, 85

Proper presentation of ligands or

agonists in nanoparticles targeting immune cells make antigens more immunogenic in vivo.

86, 87

Besides appropriate antigen presentation, coadministration of adjuvant

molecules, often targeting specific receptors of immune cells, also improves immune responses. We designed a membrane-anchored fusion protein by replacing the hyperimmunogenic region of Salmonella enterica serovar Typhimurium flagellin with four repeats of M2e (4M2e-tFliC) and fusing it to the membrane anchoring domain of influenza HA. The fusion protein was incorporated into influenza M1-driven VLPs. The combination of VLP antigen presentation and the potent FliC adjuvant greatly enhanced the M2e specific immune responses to the immunizations. 86

Non-enveloped VLPs have been widely developed with chimeric capsid proteins displaying influenza conserved epitopes which provide effective bystander T-helper

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 34

responses and induce protective, specific immune responses. 80-82, 88-94 Immunization with E. coli-produced, hepatitis B-core VLPs displaying M2e cross-protected mice and ferrets against diverse influenza virus challenges and efficiently induced protective M2e-specific immunity in volunteers.

18

VLP displaying system are frequently investigated as a

promising vaccine platform for presenting conserved surface proteins in a highly immunogenic form.

iii.

Self-assembling protein nanoparticles. Naturally occurring, self-assembling

protein nanoparticles have been identified from a wide variety of sources.

95

Self-

assembling motifs can enable fusion proteins to assemble into protein nanoparticles. 98

96-

Ferritin can be self-organized into a nanoscale structure (nanocage) with intracellular

iron storage functionality. It serves as an ideal epitope presentation platform with repetitive symmetrical structure and an ordered matrix. Headless HA trimers and tandem copies of M2e displayed on ferritin nanoparticles retained native conformation and induced protective homosubtypic and heterosubtypic immunity in vivo.

30, 99

Because

most large self-assembling domains are adopted from non-human species, off-target immune responses are a major weakness of such self-assembling protein nanoparticles. 30, 96, 97, 99

Changes in the physicochemical condition of protein solutions can drive the formation of protein nanoparticles. Because changes in physicochemical condition are dynamic processes, nanoparticles assembled this way are generally not homogenous in size but do show a normally distributed range of sizes. We have used ethanol desolvation to

ACS Paragon Plus Environment

Page 11 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

assemble influenza M2e or influenza HA into protein nanoparticles. 31, 87, 100, 101 Because these particles have solid structures without non-antigen components, they have the highest possible antigen-load for protein nanoparticles. Another advantage of these nanoparticles is that they can go through multiple cycles of particle assembly processes with different antigenic proteins to assemble proteins into different layers around the particles. Layered protein nanoparticles are particularly suitable for protein antigens with different stability in solution. We have generated layered protein nanoparticles by ethanol desolvation of M2e or NP peptides into nanoparticle cores and chemically crosslinking structure-stabilized influenza HA stalk antigens or M2e onto the outer layers.

31, 47

The

fabrication process is summarized in figure 3. These physicochemical layered protein nanoparticles can emphasize the different immunological role of the different antigens. 100

The desolvated nanoparticle size is 50 to 300 nm, depending on the desolvent composition 102 and desolvated protein materials. 31 Because this technique is compatible with most proteins, other proteins such as innate signaling initiators (immune stimulators) or immune cell-targeting molecules can be crosslinked onto the outer layers to endow additional beneficial immunological features. 103

The combination of dissolving microneedle patch technology with such protein nanoparticles enables convenient skin vaccination, which is syringe-free, painless, and can be self-administrated. Mouse skin immunization experiments demonstrated that this protein nanoparticle vaccine conferred at least four months of universal immunity against

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 34

diverse influenza virus challenges. Furthermore, this approach allows for cold-chain independent storage and triggered long-lasting protection. 31

iv.

Metal Nanoparticles. Metal nanoparticles are rigid in structure and nearly non-

biodegradable. The shape and growth of metal nanoparticles are controlled by fine-tuning the rates of surface diffusion and deposition. 104 The inorganic nanoparticle is frequently investigated as a vaccine development platform to improve antigen immunogenicity and avoid antibody production against the platform materials. Gold nanoparticles have attracted attention in the nanomedical field due to their unique advantages, their biocompatibility, and easy fabrication regarding size and shape. Gold nanoparticles less than 100 nm in diameter can be synthesized, which is preferentially recognized and engulfed by dendritic cells. Gold nanoparticles can also be formed into different shapes like star, spherical, cube and rod to control the induced, shape-dependent modulation of immune responses. 105 However, the disadvantages of gold nanoparticle vaccines include the expensive manufacture cost and reduced drug loading capacity compared with polymer nanoparticles, VLPs, and desolvated nanoparticles.

A preliminary study reported that M2e-immobilized gold nanoparticles formulated with CpG induced cross-protection against diverse influenza viral challenges.

106, 107

Our

studies have demonstrated that gold nanoparticles can conjugate influenza HA and adjuvant protein flagellin and trigger strong immune responses conferring heterologous protection in mice. 108, 109

ACS Paragon Plus Environment

Page 13 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

4. Important Physical Features Benefit to Enhanced Immunogenicity The properties of nanoparticles, such as size and steric conformation, play an active role in mediating the biological effects and the adaptive immunity induction. 110-113 Ultra-small nanoparticles less than 10 nm in diameter or soluble antigens can rapidly diffuse into and out of lymph organs, which decreases the opportunity for uptake by antigen presenting cells (APCs).

114

Nanoparticles larger than 100 nm can be trapped in the injection site,

require active transport, and are eventually scavenged by tissue-resident APCs (Figure 4).

115

We found that intramuscularly injected fluorescent NP nanoparticles remained at

the injection site much longer than fluorescent soluble NP protein (Figure 5). Smaller nanoparticles optimally sized around 50 - 200 nm in diameter were shown to be engulfed more efficiently by dendritic cells while the larger, micrometer-scale nanoparticles were preferentially internalized by macrophages. 105, 116, 117

The strength of induced immunity is also affected by the efficiency of dendritic cells migration towards the draining lymph nodes (LNs) and the length of the antigen retention period in LNs. 118 We recently found that ~200 nm protein nanoparticles had an increased chance to be presented by APCs due to their efficient drainage into inguinal LNs and spleens and relatively long residence at those sites compared with soluble antigens. The soluble molecules diffused rapidly from the injection sites and disappeared earlier from the LNs. 47

We also found that nanoparticle vaccines delivered by dissolvable microneedle patches conferred mice long-lasting anti-influenza A virus immunity. We speculated that the

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

entrapment in immune tissues and the intracellularly-activated disassembly of nanoparticles within APCs allowed for the sustained processing and releasing of peptides for a longer period versus soluble antigens, which stimulated memory T cells and longterm immunity. Sustained antigen release is a critical characteristic for long-lasting memory immune responses to nanoparticle vaccines. 114

Particle shape also plays important roles in the mediation of immunity induction, especially in the phagocytosis of particles. 119, 120 The local shape at the interface between particles and phagocytes determines the efficiency of particle uptake. 121-123 For example, spherical gold nanoparticles (40 nm) more efficiently induced antibody responses against delivered antigens than did cube- and rod-shaped nanoparticles with similar sizes. 124

A phase I clinical trial of the universal influenza A nanoparticle vaccine, M2eHBc (ACAMFLU-A™) sponsored by Acambis (later acquired by Sanofi Pasteur), generated interesting results. The intramuscularly injected ACAM-FLU-A™, adjuvanted with QS21, was well tolerated and able to stimulate anti-M2e seroconversion in up to 90% of healthy volunteers. However, the induced M2e-specific antibody titers dropped rapidly over ten months.

18

Resolving the short-term persistence issue of the M2e-specific immune

responses is the last piece of the puzzle in M2e-based vaccine designs. We found that desolvated M2e nanoparticles have an order of magnitude higher levels of M2e antigen loading than the M2eHBc construct, potentially inducing stronger and longer sustained protective M2e-specific T cell immune responses in humans.

ACS Paragon Plus Environment

Page 14 of 34

Page 15 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Concluding remarks Even if proof-of-concept has been demonstrated in pre-clinical data, a vaccine product candidate may still be an inviable solution if it is too costly or the manufacturing process is unscalable. The baculovirus expression vector/insect cell (BEVS/IC) system constitutes an optimal expression system for VLP manufactures, through which Glaxo Smith Kline’s Cervarix human papillomavirus VLP cancer vaccine is produced. Insect cell large-scale suspension cultures established in either stirred or rocked bioreactors have been shown to be one of the best VLP production system. Furthermore, improved insect cell lines have been ‘humanized’, which perform mammalian-like post-translation glycosylation modifications. 125 There are no other marketed types of nanoparticle vaccines.

Because of the many beneficial features, nanoparticles have the potential to be developed into affordable universal vaccines. Nanoparticles are safe and biocompatible vaccine platforms that can be designed in a variety of sizes with almost any antigen. The high surface area for immune ligand-receptor ligation-recognition, high capacity for antigens, and long-duration antigen processing and presentation of nanoparticles naturally lend the format to the development of a broadly protective universal influenza vaccine.

Acknowledgment This work is supported by the Institute of Biomedical Science, Georgia State University and by grants R01AI101047 and R01AI116835 (to BZW) from US National Institutes of Health.

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Competing interest statement There are no conflicting interests among all co-authors. References: [1] WHO. (2018) World Health Organization. http://www.who.int/news-room/factsheets/detail/influenza-(seasonal) (Access on June/20/2018). [2] Sullivan, S. G., Chilver, M. B., Carville, K. S., Deng, Y. M., Grant, K. A., Higgins, G., Komadina, N., Leung, V. K., Minney-Smith, C. A., Teng, D., Tran, T., Stocks, N., and Fielding, J. E. (2017) Low interim influenza vaccine effectiveness, Australia, 1 May to 24 September 2017, Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin 22. [3] Garten, R., Blanton, L., Elal, A. I. A., Alabi, N., Barnes, J., Biggerstaff, M., Brammer, L., Budd, A. P., Burns, E., Cummings, C. N., Davis, T., Garg, S., Gubareva, L., Jang, Y., Kniss, K., Kramer, N., Lindstrom, S., Mustaquim, D., O'Halloran, A., Sessions, W., Taylor, C., Xu, X., Dugan, V. G., Fry, A. M., Wentworth, D. E., Katz, J., and Jernigan, D. (2018) Update: Influenza Activity in the United States During the 2017-18 Season and Composition of the 2018-19 Influenza Vaccine, MMWR. Morbidity and mortality weekly report 67, 634-642. [4] Machado, A., Kislaya, I., Nunes, B., Rodrigues, A. P., Guiomar, R., and Euro, E. V. A. p. (2018) Moderate influenza vaccine effectiveness in a B mismatch season: Preliminary results from the 2017/2018 season in Portugal, Pulmonology. [5] CNIC. (2018) Chinese National Influenza Centre, http://www.chinaivdc.cn/cnic/en/ (Access on January/30/2018). [6] Ebrahimi, S. M., and Tebianian, M. Influenza A viruses: why focusing on M2e-based universal vaccines, Virus Genes. [7] Zhang, H., Wang, L., Compans, R. W., and Wang, B. Z. (2014) Universal influenza vaccines, a dream to be realized soon, Viruses 6, 1974-1991. [8] Mei, L., Song, P. P., Tang, Q., Shan, K., Tobe, R. G., Selotlegeng, L., Ali, A. H., Cheng, Y. Y., and Xu, L. Z. (2013) Changes in and shortcomings of control strategies, drug stockpiles, and vaccine development during outbreaks of avian influenza A H5N1, H1N1, and H7N9 among humans, Biosci Trends 7, 64-76. [9] Gao, R., Cao, B., Hu, Y., Feng, Z., Wang, D., Hu, W., Chen, J., Jie, Z., Qiu, H., Xu, K., Xu, X., Lu, H., Zhu, W., Gao, Z., Xiang, N., Shen, Y., He, Z., Gu, Y., Zhang, Z., Yang, Y., Zhao, X., Zhou, L., Li, X., Zou, S., Zhang, Y., Li, X., Yang, L., Guo, J., Dong, J., Li, Q., Dong, L., Zhu, Y., Bai, T., Wang, S., Hao, P., Yang, W., Zhang, Y., Han, J., Yu, H., Li, D., Gao, G. F., Wu, G., Wang, Y., Yuan, Z., and Shu, Y. (2013) Human Infection with a Novel Avian-Origin Influenza A (H7N9) Virus, N Engl J Med. [10] Ohmit, S. E., Petrie, J. G., Malosh, R. E., Fry, A. M., Thompson, M. G., and Monto, A. S. (2015) Influenza vaccine effectiveness in households with children during the 2012-2013 season: assessments of prior vaccination and serologic susceptibility, The Journal of infectious diseases 211, 1519-1528. [11] McLean, H. Q., Thompson, M. G., Sundaram, M. E., Meece, J. K., McClure, D. L., Friedrich, T. C., and Belongia, E. A. (2014) Impact of repeated vaccination on vaccine effectiveness against influenza A(H3N2) and B during 8 seasons, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 59, 1375-1385. [12] Ferdinands, J. M., Fry, A. M., Reynolds, S., Petrie, J., Flannery, B., Jackson, M. L., and Belongia, E. A. (2017) Intraseason waning of influenza vaccine protection: Evidence from the US Influenza

ACS Paragon Plus Environment

Page 16 of 34

Page 17 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Vaccine Effectiveness Network, 2011-12 through 2014-15, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 64, 544-550. [13] Olafsdottir, T. A., Alexandersson, K. F., Sveinbjornsson, G., Lapini, G., Palladino, L., Montomoli, E., Del Giudice, G., Gudbjartsson, D. F., and Jonsdottir, I. (2017) Age and Influenza-Specific PreVaccination Antibodies Strongly Affect Influenza Vaccine Responses in the Icelandic Population whereas Disease and Medication Have Small Effects, Frontiers in immunology 8, 1872. [14] Merani, S., Kuchel, G. A., Kleppinger, A., and McElhaney, J. E. (2018) Influenza vaccine-mediated protection in older adults: Impact of influenza infection, cytomegalovirus serostatus and vaccine dosage, Experimental gerontology 107, 116-125. [15] Zost, S. J., Parkhouse, K., Gumina, M. E., Kim, K., Diaz Perez, S., Wilson, P. C., Treanor, J. J., Sant, A. J., Cobey, S., and Hensley, S. E. (2017) Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains, Proceedings of the National Academy of Sciences of the United States of America 114, 12578-12583. [16] Du, Y., Xin, L., Shi, Y., Zhang, T. H., Wu, N. C., Dai, L., Gong, D., Brar, G., Shu, S., Luo, J., Reiley, W., Tseng, Y. W., Bai, H., Wu, T. T., Wang, J., Shu, Y., and Sun, R. (2018) Genome-wide identification of interferon-sensitive mutations enables influenza vaccine design, Science 359, 290-296. [17] Lamb, R. A., Zebedee, S. L., and Richardson, C. D. (1985) Influenza virus M2 protein is an integral membrane protein expressed on the infected-cell surface, Cell 40, 627-633. [18] Deng, L., Cho, K. J., Fiers, W., and Saelens, X. (2015) M2e-Based Universal Influenza A Vaccines, Vaccines 3, 105-136. [19] Hutchinson, E. C., Charles, P. D., Hester, S. S., Thomas, B., Trudgian, D., Martinez-Alonso, M., and Fodor, E. (2014) Conserved and host-specific features of influenza virion architecture, Nature communications 5, 4816. [20] El Bakkouri, K., Descamps, F., De Filette, M., Smet, A., Festjens, E., Birkett, A., Van Rooijen, N., Verbeek, S., Fiers, W., and Saelens, X. (2011) Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection, J Immunol 186, 1022-1031. [21] Schepens, B., De Vlieger, D., and Saelens, X. (2018) Vaccine options for influenza: thinking small, Current opinion in immunology 53, 22-29. [22] Throsby, M., van den Brink, E., Jongeneelen, M., Poon, L. L., Alard, P., Cornelissen, L., Bakker, A., Cox, F., van Deventer, E., Guan, Y., Cinatl, J., ter Meulen, J., Lasters, I., Carsetti, R., Peiris, M., de Kruif, J., and Goudsmit, J. (2008) Heterosubtypic neutralizing monoclonal antibodies crossprotective against H5N1 and H1N1 recovered from human IgM+ memory B cells, PloS one 3, e3942. [23] Dreyfus, C., Laursen, N. S., Kwaks, T., Zuijdgeest, D., Khayat, R., Ekiert, D. C., Lee, J. H., Metlagel, Z., Bujny, M. V., Jongeneelen, M., van der Vlugt, R., Lamrani, M., Korse, H. J., Geelen, E., Sahin, O., Sieuwerts, M., Brakenhoff, J. P., Vogels, R., Li, O. T., Poon, L. L., Peiris, M., Koudstaal, W., Ward, A. B., Wilson, I. A., Goudsmit, J., and Friesen, R. H. (2012) Highly conserved protective epitopes on influenza B viruses, Science 337, 1343-1348. [24] Corti, D., Voss, J., Gamblin, S. J., Codoni, G., Macagno, A., Jarrossay, D., Vachieri, S. G., Pinna, D., Minola, A., Vanzetta, F., Silacci, C., Fernandez-Rodriguez, B. M., Agatic, G., Bianchi, S., Giacchetto-Sasselli, I., Calder, L., Sallusto, F., Collins, P., Haire, L. F., Temperton, N., Langedijk, J. P., Skehel, J. J., and Lanzavecchia, A. (2011) A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins, Science 333, 850-856. [25] Sui, J., Hwang, W. C., Perez, S., Wei, G., Aird, D., Chen, L. M., Santelli, E., Stec, B., Cadwell, G., Ali, M., Wan, H., Murakami, A., Yammanuru, A., Han, T., Cox, N. J., Bankston, L. A., Donis, R. O., Liddington, R. C., and Marasco, W. A. (2009) Structural and functional bases for broad-spectrum

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

neutralization of avian and human influenza A viruses, Nature structural & molecular biology 16, 265-273. [26] Tan, G. S., Lee, P. S., Hoffman, R. M., Mazel-Sanchez, B., Krammer, F., Leon, P. E., Ward, A. B., Wilson, I. A., and Palese, P. (2014) Characterization of a broadly neutralizing monoclonal antibody that targets the fusion domain of group 2 influenza A virus hemagglutinin, Journal of virology 88, 13580-13592. [27] Friesen, R. H., Lee, P. S., Stoop, E. J., Hoffman, R. M., Ekiert, D. C., Bhabha, G., Yu, W., Juraszek, J., Koudstaal, W., Jongeneelen, M., Korse, H. J., Ophorst, C., Brinkman-van der Linden, E. C., Throsby, M., Kwakkenbos, M. J., Bakker, A. Q., Beaumont, T., Spits, H., Kwaks, T., Vogels, R., Ward, A. B., Goudsmit, J., and Wilson, I. A. (2014) A common solution to group 2 influenza virus neutralization, Proceedings of the National Academy of Sciences of the United States of America 111, 445-450. [28] Tharakaraman, K., Subramanian, V., Viswanathan, K., Sloan, S., Yen, H. L., Barnard, D. L., Leung, Y. H., Szretter, K. J., Koch, T. J., Delaney, J. C., Babcock, G. J., Wogan, G. N., Sasisekharan, R., and Shriver, Z. (2015) A broadly neutralizing human monoclonal antibody is effective against H7N9, Proceedings of the National Academy of Sciences of the United States of America 112, 1089010895. [29] Impagliazzo, A., Milder, F., Kuipers, H., Wagner, M. V., Zhu, X., Hoffman, R. M., van Meersbergen, R., Huizingh, J., Wanningen, P., Verspuij, J., de Man, M., Ding, Z., Apetri, A., Kukrer, B., SneekesVriese, E., Tomkiewicz, D., Laursen, N. S., Lee, P. S., Zakrzewska, A., Dekking, L., Tolboom, J., Tettero, L., van Meerten, S., Yu, W., Koudstaal, W., Goudsmit, J., Ward, A. B., Meijberg, W., Wilson, I. A., and Radosevic, K. (2015) A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen, Science 349, 1301-1306. [30] 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., Lingwood, D., Moin, S. M., Andersen, H., Okuno, Y., Rao, S. S., Harris, A. K., Kwong, P. D., Mascola, J. R., Nabel, G. J., and Graham, B. S. (2015) Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection, Nature medicine 21, 1065-1070. [31] Deng, L., Mohan, T., Chang, T. Z., Gonzalez, G. X., Wang, Y., Kwon, Y. M., Kang, S. M., Compans, R. W., Champion, J. A., and Wang, B. Z. (2018) Double-layered protein nanoparticles induce broad protection against divergent influenza A viruses, Nature communications 9, 359. [32] Zheng, M., Luo, J., and Chen, Z. (2014) Development of universal influenza vaccines based on influenza virus M and NP genes, Infection 42, 251-262. [33] Sridhar, S., Begom, S., Bermingham, A., Hoschler, K., Adamson, W., Carman, W., Bean, T., Barclay, W., Deeks, J. J., and Lalvani, A. (2013) Cellular immune correlates of protection against symptomatic pandemic influenza, Nature medicine 19, 1305-1312. [34] Eliasson, D. G., Omokanye, A., Schon, K., Wenzel, U. A., Bernasconi, V., Bemark, M., Kolpe, A., El Bakkouri, K., Ysenbaert, T., Deng, L., Fiers, W., Saelens, X., and Lycke, N. (2017) M2e-tetramerspecific memory CD4 T cells are broadly protective against influenza infection, Mucosal immunology. [35] Morens, D. M., Taubenberger, J. K., and Fauci, A. S. (2013) Pandemic influenza viruses--hoping for the road not taken, The New England journal of medicine 368, 2345-2348. [36] Chenavas, S., Estrozi, L. F., Slama-Schwok, A., Delmas, B., Di Primo, C., Baudin, F., Li, X., Crepin, T., and Ruigrok, R. W. (2013) Monomeric nucleoprotein of influenza A virus, PLoS pathogens 9, e1003275. [37] Antrobus, R. D., Berthoud, T. K., Mullarkey, C. E., Hoschler, K., Coughlan, L., Zambon, M., Hill, A. V., and Gilbert, S. C. (2014) Coadministration of seasonal influenza vaccine and MVA-NP+M1

ACS Paragon Plus Environment

Page 18 of 34

Page 19 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

simultaneously achieves potent humoral and cell-mediated responses, Molecular therapy : the journal of the American Society of Gene Therapy 22, 233-238. [38] Wang, W., Li, R., Deng, Y., Lu, N., Chen, H., Meng, X., Wang, W., Wang, X., Yan, K., Qi, X., Zhang, X., Xin, W., Lu, Z., Li, X., Bian, T., Gao, Y., Tan, W., and Ruan, L. (2015) Protective Efficacy of the Conserved NP, PB1, and M1 Proteins as Immunogens in DNA- and Vaccinia Virus-Based Universal Influenza A Virus Vaccines in Mice, Clinical and vaccine immunology : CVI 22, 618-630. [39] Lillie, P. J., Berthoud, T. K., Powell, T. J., Lambe, T., Mullarkey, C., Spencer, A. J., Hamill, M., Peng, Y., Blais, M. E., Duncan, C. J., Sheehy, S. H., Havelock, T., Faust, S. N., Williams, R. L., Gilbert, A., Oxford, J., Dong, T., Hill, A. V., and Gilbert, S. C. (2012) Preliminary assessment of the efficacy of a T-cell-based influenza vaccine, MVA-NP+M1, in humans, Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 55, 19-25. [40] Coughlan, L., Sridhar, S., Payne, R., Edmans, M., Milicic, A., Venkatraman, N., Lugonja, B., Clifton, L., Qi, C., Folegatti, P. M., Lawrie, A. M., Roberts, R., de Graaf, H., Sukhtankar, P., Faust, S. N., Lewis, D. J. M., Lambe, T., Hill, A., and Gilbert, S. C. (2018) Heterologous Two-Dose Vaccination with Simian Adenovirus and Poxvirus Vectors Elicits Long-Lasting Cellular Immunity to Influenza Virus A in Healthy Adults, EBioMedicine 29, 146-154. [41] Di Mario, G., Sciaraffia, E., Facchini, M., Gubinelli, F., Soprana, E., Panigada, M., Bernasconi, V., Garulli, B., Siccardi, A., Donatelli, I., and Castrucci, M. R. (2017) Protective immunity against influenza in HLA-A2 transgenic mice by modified vaccinia virus Ankara vectored vaccines containing internal influenza proteins, Pathogens and global health 111, 76-82. [42] Price, G. E., Lo, C. Y., Misplon, J. A., and Epstein, S. L. (2018) Reduction of influenza virus transmission from mice immunized against conserved viral antigens is influenced by route of immunization and choice of vaccine antigen, Vaccine 36, 4910-4918. [43] Zhao, L., Seth, A., Wibowo, N., Zhao, C. X., Mitter, N., Yu, C., and Middelberg, A. P. (2014) Nanoparticle vaccines, Vaccine 32, 327-337. [44] Torchilin, V. P. (2014) Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery, Nature reviews. Drug discovery 13, 813-827. [45] Singh, S. M., Alkie, T. N., Abdelaziz, K. T., Hodgins, D. C., Novy, A., Nagy, E., and Sharif, S. (2016) Characterization of Immune Responses to an Inactivated Avian Influenza Virus Vaccine Adjuvanted with Nanoparticles Containing CpG ODN, Viral immunology 29, 269-275. [46] Wang, Q., Tan, M. T., Keegan, B. P., Barry, M. A., and Heffernan, M. J. (2014) Time course study of the antigen-specific immune response to a PLGA microparticle vaccine formulation, Biomaterials 35, 8385-8393. [47] Deng, L., Chang, T. Z., Wang, Y., Li, S., Wang, S., Matsuyama, S., Yu, G., Compans, R. W., Li, J. D., Prausnitz, M. R., Champion, J. A., and Wang, B. Z. (2018) Heterosubtypic influenza protection elicited by double-layered polypeptide nanoparticles in mice, Proceedings of the National Academy of Sciences of the United States of America. [48] Shastri, P. N., Kim, M. C., Quan, F. S., D'Souza, M. J., and Kang, S. M. (2012) Immunogenicity and protection of oral influenza vaccines formulated into microparticles, Journal of pharmaceutical sciences 101, 3623-3635. [49] Zhang, C., Zhang, J., Shi, G., Song, H., Shi, S., Zhang, X., Huang, P., Wang, Z., Wang, W., Wang, C., Kong, D., and Li, C. (2017) A Light Responsive Nanoparticle-Based Delivery System Using Pheophorbide A Graft Polyethylenimine for Dendritic Cell-Based Cancer Immunotherapy, Molecular pharmaceutics 14, 1760-1770. [50] Dhakal, S., Hiremath, J., Bondra, K., Lakshmanappa, Y. S., Shyu, D. L., Ouyang, K., Kang, K. I., Binjawadagi, B., Goodman, J., Tabynov, K., Krakowka, S., Narasimhan, B., Lee, C. W., and Renukaradhya, G. J. (2017) Biodegradable nanoparticle delivery of inactivated swine influenza

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

virus vaccine provides heterologous cell-mediated immune response in pigs, Journal of controlled release : official journal of the Controlled Release Society 247, 194-205. [51] Rosalia, R. A., Cruz, L. J., van Duikeren, S., Tromp, A. T., Silva, A. L., Jiskoot, W., de Gruijl, T., Lowik, C., Oostendorp, J., van der Burg, S. H., and Ossendorp, F. (2015) CD40-targeted dendritic cell delivery of PLGA-nanoparticle vaccines induce potent anti-tumor responses, Biomaterials 40, 8897. [52] Monkare, J., Pontier, M., van Kampen, E. E. M., Du, G., Leone, M., Romeijn, S., Nejadnik, M. R., O'Mahony, C., Slutter, B., Jiskoot, W., and Bouwstra, J. A. (2018) Development of PLGA nanoparticle loaded dissolving microneedles and comparison with hollow microneedles in intradermal vaccine delivery, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V 129, 111-121. [53] Danhier, F., Ansorena, E., Silva, J. M., Coco, R., Le Breton, A., and Preat, V. (2012) PLGA-based nanoparticles: an overview of biomedical applications, Journal of controlled release : official journal of the Controlled Release Society 161, 505-522. [54] Thomas, C., Rawat, A., Hope-Weeks, L., and Ahsan, F. (2011) Aerosolized PLA and PLGA nanoparticles enhance humoral, mucosal and cytokine responses to hepatitis B vaccine, Molecular pharmaceutics 8, 405-415. [55] Rahimian, S., Fransen, M. F., Kleinovink, J. W., Christensen, J. R., Amidi, M., Hennink, W. E., and Ossendorp, F. (2015) Polymeric nanoparticles for co-delivery of synthetic long peptide antigen and poly IC as therapeutic cancer vaccine formulation, Journal of controlled release : official journal of the Controlled Release Society 203, 16-22. [56] Minigo, G., Scholzen, A., Tang, C. K., Hanley, J. C., Kalkanidis, M., Pietersz, G. A., Apostolopoulos, V., and Plebanski, M. (2007) Poly-L-lysine-coated nanoparticles: a potent delivery system to enhance DNA vaccine efficacy, Vaccine 25, 1316-1327. [57] Hiremath, J., Kang, K. I., Xia, M., Elaish, M., Binjawadagi, B., Ouyang, K., Dhakal, S., Arcos, J., Torrelles, J. B., Jiang, X., Lee, C. W., and Renukaradhya, G. J. (2016) Entrapment of H1N1 Influenza Virus Derived Conserved Peptides in PLGA Nanoparticles Enhances T Cell Response and Vaccine Efficacy in Pigs, PloS one 11, e0151922. [58] Heinen, P. P., Rijsewijk, F. A., de Boer-Luijtze, E. A., and Bianchi, A. T. (2002) Vaccination of pigs with a DNA construct expressing an influenza virus M2-nucleoprotein fusion protein exacerbates disease after challenge with influenza A virus, The Journal of general virology 83, 1851-1859. [59] Watkins, H. C., Pagan, C. L., Childs, H. R., Posada, S., Chau, A., Rios, J., Guarino, C., DeLisa, M. P., Whittaker, G. R., and Putnam, D. (2017) A single dose and long lasting vaccine against pandemic influenza through the controlled release of a heterospecies tandem M2 sequence embedded within detoxified bacterial outer membrane vesicles, Vaccine 35, 5373-5380. [60] Arca, H. C., Gunbeyaz, M., and Senel, S. (2009) Chitosan-based systems for the delivery of vaccine antigens, Expert review of vaccines 8, 937-953. [61] Dhakal, S., Renu, S., Ghimire, S., Shaan Lakshmanappa, Y., Hogshead, B. T., Feliciano-Ruiz, N., Lu, F., HogenEsch, H., Krakowka, S., Lee, C. W., and Renukaradhya, G. J. (2018) Mucosal Immunity and Protective Efficacy of Intranasal Inactivated Influenza Vaccine Is Improved by Chitosan Nanoparticle Delivery in Pigs, Frontiers in immunology 9, 934. [62] Shi, G. N., Zhang, C. N., Xu, R., Niu, J. F., Song, H. J., Zhang, X. Y., Wang, W. W., Wang, Y. M., Li, C., Wei, X. Q., and Kong, D. L. (2017) Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine, Biomaterials 113, 191-202. [63] Chowdhury, M. Y. E., Kim, T. H., Uddin, M. B., Kim, J. H., Hewawaduge, C. Y., Ferdowshi, Z., Sung, M. H., Kim, C. J., and Lee, J. S. (2017) Mucosal vaccination of conserved sM2, HA2 and cholera toxin subunit A1 (CTA1) fusion protein with poly gamma-glutamate/chitosan nanoparticles (PC NPs) induces protection against divergent influenza subtypes, Veterinary microbiology 201, 240-251.

ACS Paragon Plus Environment

Page 20 of 34

Page 21 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

[64] Yang, J., Shim, S. M., Nguyen, T. Q., Kim, E. H., Kim, K., Lim, Y. T., Sung, M. H., Webby, R., and Poo, H. (2017) Poly-gamma-glutamic acid/chitosan nanogel greatly enhances the efficacy and heterosubtypic cross-reactivity of H1N1 pandemic influenza vaccine, Scientific reports 7, 44839. [65] Wang, B. Z., Gill, H. S., He, C., Ou, C., Wang, L., Wang, Y. C., Feng, H., Zhang, H., Prausnitz, M. R., and Compans, R. W. (2014) Microneedle delivery of an M2e-TLR5 ligand fusion protein to skin confers broadly cross-protective influenza immunity, Journal of controlled release : official journal of the Controlled Release Society 178, 1-7. [66] Wang, B. Z., Quan, F. S., Kang, S. M., Bozja, J., Skountzou, I., and Compans, R. W. (2008) Incorporation of membrane-anchored flagellin into influenza virus-like particles enhances the breadth of immune responses, Journal of virology 82, 11813-11823. [67] Wang, B. Z., Xu, R., Quan, F. S., Kang, S. M., Wang, L., and Compans, R. W. (2010) Intranasal immunization with influenza VLPs incorporating membrane-anchored flagellin induces strong heterosubtypic protection, PloS one 5, e13972. [68] Wang, L., Wang, Y. C., Feng, H., Ahmed, T., Compans, R. W., and Wang, B. Z. (2013) Virus-like particles containing the tetrameric ectodomain of influenza matrix protein 2 and flagellin induce heterosubtypic protection in mice, BioMed research international 2013, 686549. [69] Andre, F. E. (1990) Overview of a 5-year clinical experience with a yeast-derived hepatitis B vaccine, Vaccine 8 Suppl, S74-78; discussion S79-80. [70] Si, L., Xu, H., Zhou, X., Zhang, Z., Tian, Z., Wang, Y., Wu, Y., Zhang, B., Niu, Z., Zhang, C., Fu, G., Xiao, S., Xia, Q., Zhang, L., and Zhou, D. (2016) Generation of influenza A viruses as live but replication-incompetent virus vaccines, Science 354, 1170-1173. [71] Zeltins, A. (2013) Construction and characterization of virus-like particles: a review, Molecular biotechnology 53, 92-107. [72] Roldao, A., Mellado, M. C., Castilho, L. R., Carrondo, M. J., and Alves, P. M. (2010) Virus-like particles in vaccine development, Expert review of vaccines 9, 1149-1176. [73] D'Aoust, M. A., Couture, M. M., Charland, N., Trepanier, S., Landry, N., Ors, F., and Vezina, L. P. (2010) The production of hemagglutinin-based virus-like particles in plants: a rapid, efficient and safe response to pandemic influenza, Plant biotechnology journal 8, 607-619. [74] Pillet, S., Aubin, E., Trepanier, S., Bussiere, D., Dargis, M., Poulin, J. F., Yassine-Diab, B., Ward, B. J., and Landry, N. (2016) A plant-derived quadrivalent virus like particle influenza vaccine induces cross-reactive antibody and T cell response in healthy adults, Clinical immunology 168, 72-87. [75] Klausberger, M., Wilde, M., Palmberger, D., Hai, R., Albrecht, R. A., Margine, I., Hirsh, A., GarciaSastre, A., Grabherr, R., and Krammer, F. (2014) One-shot vaccination with an insect cell-derived low-dose influenza A H7 virus-like particle preparation protects mice against H7N9 challenge, Vaccine 32, 355-362. [76] Smith, G. E., Flyer, D. C., Raghunandan, R., Liu, Y., Wei, Z., Wu, Y., Kpamegan, E., Courbron, D., Fries, L. F., 3rd, and Glenn, G. M. (2013) Development of influenza H7N9 virus like particle (VLP) vaccine: homologous A/Anhui/1/2013 (H7N9) protection and heterologous A/chicken/Jalisco/CPA1/2012 (H7N3) cross-protection in vaccinated mice challenged with H7N9 virus, Vaccine 31, 4305-4313. [77] Milian, E., and Kamen, A. A. (2015) Current and emerging cell culture manufacturing technologies for influenza vaccines, BioMed research international 2015, 504831. [78] Beljanski, V., Chiang, C., Kirchenbaum, G. A., Olagnier, D., Bloom, C. E., Wong, T., Haddad, E. K., Trautmann, L., Ross, T. M., and Hiscott, J. (2015) Enhanced Influenza Virus-Like Particle Vaccination with a Structurally Optimized RIG-I Agonist as Adjuvant, Journal of virology 89, 10612-10624. [79] Chia, M. Y., Hu, A. Y., Tseng, Y. F., Weng, T. C., Lai, C. C., Lin, J. Y., Chen, P. L., Wang, Y. F., Chao, S. R., Chang, J. Y., Hwang, Y. S., Yeh, C. T., Yu, C. P., Chen, Y. C., Su, I. J., and Lee, M. S. (2015)

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Evaluation of MDCK cell-derived influenza H7N9 vaccine candidates in ferrets, PloS one 10, e0120793. [80] Neirynck, S., Deroo, T., Saelens, X., Vanlandschoot, P., Jou, W. M., and Fiers, W. (1999) A universal influenza A vaccine based on the extracellular domain of the M2 protein, Nature medicine 5, 1157-1163. [81] Deng, L., Ibanez, L. I., Van den Bossche, V., Roose, K., Youssef, S. A., de Bruin, A., Fiers, W., and Saelens, X. (2015) Protection against Influenza A Virus Challenge with M2e-Displaying Filamentous Escherichia coli Phages, PloS one 10, e0126650. [82] Hashemi, H., Pouyanfard, S., Bandehpour, M., Noroozbabaei, Z., Kazemi, B., Saelens, X., and Mokhtari-Azad, T. (2012) Immunization with M2e-displaying T7 bacteriophage nanoparticles protects against influenza A virus challenge, PloS one 7, e45765. [83] Steel, J., Lowen, A. C., Wang, T. T., Yondola, M., Gao, Q., Haye, K., Garcia-Sastre, A., and Palese, P. (2010) Influenza virus vaccine based on the conserved hemagglutinin stalk domain, mBio 1. [84] Song, J. M., Van Rooijen, N., Bozja, J., Compans, R. W., and Kang, S. M. (2011) Vaccination inducing broad and improved cross protection against multiple subtypes of influenza A virus, Proceedings of the National Academy of Sciences of the United States of America 108, 757-761. [85] Kim, M. C., Song, J. M., O, E., Kwon, Y. M., Lee, Y. J., Compans, R. W., and Kang, S. M. (2013) Viruslike particles containing multiple M2 extracellular domains confer improved cross-protection against various subtypes of influenza virus, Molecular therapy : the journal of the American Society of Gene Therapy 21, 485-492. [86] Wang, B. Z., Gill, H. S., Kang, S. M., Wang, L., Wang, Y. C., Vassilieva, E. V., and Compans, R. W. (2012) Enhanced influenza virus-like particle vaccines containing the extracellular domain of matrix protein 2 and a Toll-like receptor ligand, Clinical and vaccine immunology : CVI 19, 11191125. [87] Wang, L., Hess, A., Chang, T. Z., Wang, Y. C., Champion, J. A., Compans, R. W., and Wang, B. Z. (2014) Nanoclusters self-assembled from conformation-stabilized influenza M2e as broadly cross-protective influenza vaccines, Nanomedicine : nanotechnology, biology, and medicine 10, 473-482. [88] Bessa, J., Schmitz, N., Hinton, H. J., Schwarz, K., Jegerlehner, A., and Bachmann, M. F. (2008) Efficient induction of mucosal and systemic immune responses by virus-like particles administered intranasally: implications for vaccine design, European journal of immunology 38, 114-126. [89] Petukhova, N. V., Gasanova, T. V., Stepanova, L. A., Rusova, O. A., Potapchuk, M. V., Korotkov, A. V., Skurat, E. V., Tsybalova, L. M., Kiselev, O. I., Ivanov, P. A., and Atabekov, J. G. (2013) Immunogenicity and protective efficacy of candidate universal influenza A nanovaccines produced in plants by Tobacco mosaic virus-based vectors, Current pharmaceutical design 19, 5587-5600. [90] Leclerc, D., Rivest, M., Babin, C., Lopez-Macias, C., and Savard, P. (2013) A novel M2e based flu vaccine formulation for dogs, PloS one 8, e77084. [91] Tyulkina, L. G., Skurat, E. V., Frolova, O. Y., Komarova, T. V., Karger, E. M., and Atabekov, I. G. (2011) New viral vector for superproduction of epitopes of vaccine proteins in plants, Acta naturae 3, 73-82. [92] Denis, J., Acosta-Ramirez, E., Zhao, Y., Hamelin, M. E., Koukavica, I., Baz, M., Abed, Y., Savard, C., Pare, C., Lopez Macias, C., Boivin, G., and Leclerc, D. (2008) Development of a universal influenza A vaccine based on the M2e peptide fused to the papaya mosaic virus (PapMV) vaccine platform, Vaccine 26, 3395-3403.

ACS Paragon Plus Environment

Page 22 of 34

Page 23 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

[93] Matic, S., Rinaldi, R., Masenga, V., and Noris, E. (2011) Efficient production of chimeric human papillomavirus 16 L1 protein bearing the M2e influenza epitope in Nicotiana benthamiana plants, BMC biotechnology 11, 106. [94] Ameiss, K., Ashraf, S., Kong, W., Pekosz, A., Wu, W. H., Milich, D., Billaud, J. N., and Curtiss, R., 3rd. (2010) Delivery of woodchuck hepatitis virus-like particle presented influenza M2e by recombinant attenuated Salmonella displaying a delayed lysis phenotype, Vaccine 28, 67046713. [95] Lee, L. A., and Wang, Q. (2006) Adaptations of nanoscale viruses and other protein cages for medical applications, Nanomedicine : nanotechnology, biology, and medicine 2, 137-149. [96] Jardine, J., Julien, J. P., Menis, S., Ota, T., Kalyuzhniy, O., McGuire, A., Sok, D., Huang, P. S., MacPherson, S., Jones, M., Nieusma, T., Mathison, J., Baker, D., Ward, A. B., Burton, D. R., Stamatatos, L., Nemazee, D., Wilson, I. A., and Schief, W. R. (2013) Rational HIV immunogen design to target specific germline B cell receptors, Science 340, 711-716. [97] Kanekiyo, M., Wei, C. J., Yassine, H. M., McTamney, P. M., Boyington, J. C., Whittle, J. R., Rao, S. S., Kong, W. P., Wang, L., and Nabel, G. J. (2013) Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies, Nature 499, 102-106. [98] Georgiev, I. S., Joyce, M. G., Chen, R. E., Leung, K., McKee, K., Druz, A., Van Galen, J. G., Kanekiyo, M., Tsybovsky, Y., Yang, E. S., Yang, Y., Acharya, P., Pancera, M., Thomas, P. V., Wanninger, T., Yassine, H. M., Baxa, U., Doria-Rose, N. A., Cheng, C., Graham, B. S., Mascola, J. R., and Kwong, P. D. (2018) Two-Component Ferritin Nanoparticles for Multimerization of Diverse Trimeric Antigens, ACS infectious diseases 4, 788-796. [99] Qi, M., Zhang, X. E., Sun, X., Zhang, X., Yao, Y., Liu, S., Chen, Z., Li, W., Zhang, Z., Chen, J., and Cui, Z. (2018) Intranasal Nanovaccine Confers Homo- and Hetero-Subtypic Influenza Protection, Small. [100] Wang, L., Chang, T. Z., He, Y., Kim, J. R., Wang, S., Mohan, T., Berman, Z., Tompkins, S. M., Tripp, R. A., Compans, R. W., Champion, J. A., and Wang, B. Z. (2017) Coated protein nanoclusters from influenza H7N9 HA are highly immunogenic and induce robust protective immunity, Nanomedicine : nanotechnology, biology, and medicine 13, 253-262. [101] Deng, L., Kim, J. R., Chang, T. Z., Zhang, H., Mohan, T., Champion, J. A., and Wang, B. Z. (2017) Protein nanoparticle vaccine based on flagellin carrier fused to influenza conserved epitopes confers full protection against influenza A virus challenge, Virology 509, 82-89. [102] Storp, B., Engel, A., Boeker, A., Ploeger, M., and Langer, K. (2012) Albumin nanoparticles with predictable size by desolvation procedure, Journal of microencapsulation 29, 138-146. [103] Chang, T. Z., Diambou, I., Kim, J. R., Wang, B., and Champion, J. A. (2017) Host- and pathogenderived adjuvant coatings on protein nanoparticle vaccines, Bioengineering & translational medicine 2, 120-130. [104] Konuk, M., and Durukanoglu, S. (2016) Shape-controlled growth of metal nanoparticles: an atomistic view, Physical chemistry chemical physics : PCCP 18, 1876-1885. [105] Kumar, S., Anselmo, A. C., Banerjee, A., Zakrewsky, M., and Mitragotri, S. (2015) Shape and sizedependent immune response to antigen-carrying nanoparticles, Journal of controlled release : official journal of the Controlled Release Society 220, 141-148. [106] Tao, W., Ziemer, K. S., and Gill, H. S. (2014) Gold nanoparticle-M2e conjugate coformulated with CpG induces protective immunity against influenza A virus, Nanomedicine (Lond) 9, 237-251. [107] Tao, W., and Gill, H. S. (2015) M2e-immobilized gold nanoparticles as influenza A vaccine: Role of soluble M2e and longevity of protection, Vaccine 33, 2307-2315. [108] Chao, W., Wandi, Z., and W., B. (2017) Dual-linker gold nanoparticles as adjuvanting carriers for multivalent display of recombinant influenza hemagglutinin trimer and flagellin improve the immunological responses in vivo and in vitro, International journal of nanomedicine 12, 47474762.

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

[109] Wang, C., Zhu, W., Luo, Y., and Wang, B. Z. (2018) Gold nanoparticles conjugating recombinant influenza hemagglutinin trimer and flagellin enhanced mucosal cellular immunity, Nanomedicine : nanotechnology, biology, and medicine 14, 1349-1360. [110] Dobrovolskaia, M. A., Aggarwal, P., Hall, J. B., and McNeil, S. E. (2008) Preclinical studies to understand nanoparticle interaction with the immune system and its potential effects on nanoparticle biodistribution, Molecular pharmaceutics 5, 487-495. [111] Toy, R., and Roy, K. (2016) Engineering nanoparticles to overcome barriers to immunotherapy, Bioengineering & translational medicine 1, 47-62. [112] Benne, N., van Duijn, J., Kuiper, J., Jiskoot, W., and Slutter, B. (2016) Orchestrating immune responses: How size, shape and rigidity affect the immunogenicity of particulate vaccines, Journal of controlled release : official journal of the Controlled Release Society 234, 124-134. [113] Hammond, P. T. (2017) Nano Tools Pave the Way to New Solutions in Infectious Disease, ACS infectious diseases 3, 554-558. [114] Demento, S. L., Cui, W., Criscione, J. M., Stern, E., Tulipan, J., Kaech, S. M., and Fahmy, T. M. (2012) Role of sustained antigen release from nanoparticle vaccines in shaping the T cell memory phenotype, Biomaterials 33, 4957-4964. [115] Manolova, V., Flace, A., Bauer, M., Schwarz, K., Saudan, P., and Bachmann, M. F. (2008) Nanoparticles target distinct dendritic cell populations according to their size, European journal of immunology 38, 1404-1413. [116] Xiang, S. D., Scholzen, A., Minigo, G., David, C., Apostolopoulos, V., Mottram, P. L., and Plebanski, M. (2006) Pathogen recognition and development of particulate vaccines: does size matter?, Methods 40, 1-9. [117] Joshi, V. B., Geary, S. M., and Salem, A. K. (2013) Biodegradable particles as vaccine antigen delivery systems for stimulating cellular immune responses, Human vaccines & immunotherapeutics 9, 2584-2590. [118] MartIn-Fontecha, A., Sebastiani, S., Hopken, U. E., Uguccioni, M., Lipp, M., Lanzavecchia, A., and Sallusto, F. (2003) Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming, The Journal of experimental medicine 198, 615-621. [119] Xie, X., Liao, J., Shao, X., Li, Q., and Lin, Y. (2017) The Effect of shape on Cellular Uptake of Gold Nanoparticles in the forms of Stars, Rods, and Triangles, Scientific reports 7, 3827. [120] Gong, T., Xie, J., Liao, J., Zhang, T., Lin, S., and Lin, Y. (2015) Nanomaterials and bone regeneration, Bone research 3, 15029. [121] Anselmo, A. C., Zhang, M., Kumar, S., Vogus, D. R., Menegatti, S., Helgeson, M. E., and Mitragotri, S. (2015) Elasticity of nanoparticles influences their blood circulation, phagocytosis, endocytosis, and targeting, ACS nano 9, 3169-3177. [122] Akei, H., Whitsett, J. A., Buroker, M., Ninomiya, T., Tatsumi, H., Weaver, T. E., and Ikegami, M. (2006) Surface tension influences cell shape and phagocytosis in alveolar macrophages, American journal of physiology. Lung cellular and molecular physiology 291, L572-579. [123] Patel, B., Gupta, N., and Ahsan, F. (2015) Particle engineering to enhance or lessen particle uptake by alveolar macrophages and to influence the therapeutic outcome, European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V 89, 163-174. [124] Niikura, K., Matsunaga, T., Suzuki, T., Kobayashi, S., Yamaguchi, H., Orba, Y., Kawaguchi, A., Hasegawa, H., Kajino, K., Ninomiya, T., Ijiro, K., and Sawa, H. (2013) Gold nanoparticles as a vaccine platform: influence of size and shape on immunological responses in vitro and in vivo, ACS nano 7, 3926-3938. [125] hang, G. D., Chen, C. J., Lin, C. Y., Chen, H. C., and Chen, H. (2003) Improvement of glycosylation in insect cells with mammalian glycosyltransferases, J Biotechnol 102, 61-71.

ACS Paragon Plus Environment

Page 24 of 34

Page 25 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Legends Figure 1. Schematic diagram of influenza A virus. The antigen name encoded by each gene segment is labeled aside. HA, hemagglutinin; NA, neuraminidase; M1, matrix protein 1; M2e, matrix protein 2 ectodomain; vRNP, viral ribonucleoprotein; PB, polymerase basic protein; PA, polymerase acid protein; NP, nucleoprotein; NS, nonstructural protein.

Figure 2. Nanoparticle universal influenza vaccine types. (A) Polymer nanoparticle; (B) Virus like particle; (C) Gold nanoparticle; (D) Chemically assembled double-layered protein nanoparticle; (E) Naturally assembled protein nanoparticle (EMDB EMD-6332).

Figure 3. Schematic diagram of double-layered nanoparticle fabrication process. RT, room temperature; DPBS, Dulbecco’s phosphate-buffered saline.

Figure 4. Nanoparticle transport route changes with nanoparticle sizes. Nanoparticles with larger sizes over 100 nm are captured by dendritic cells from outside of lymphatic epithelia, in comparison, soluble protein antigen and smaller nanoparticles less than 10 nm are able to penetrate through lymphatic epithelia. Once antigens transported into lymph nodes via lymphatic vessels, they are presented to B cells and T cells by follicle dendritic cells. The larger nanoparticles are much more efficiently taken up by dendritic cells than smaller nanoparticles and potentially induce stronger and longterm specific immunity.

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 5. In vivo visualization in mice. The fabrications of fluorescent NP protein and fluorescent NP nanoparticle were described previously. (46) Mice were fed alfalfa-free diets for two weeks to reduce autofluorescence background before intramuscular injection with five micrograms of fluorescent antigen each mouse. Biodistributions of intramuscularly injected Alexa Fluor 700 succinimidyl ester dye-conjugated fluorescent NP nanoparticle and fluorescent soluble NP protein were analyzed in vivo using PerkinElmer IVIS Spectrum In Vivo Imaging System at the following time points: pre-injection, 0-hour post injection (hpi), 24-hpi and 48-hpi. (46) Mice injected with DPBS were used as negative control.

ACS Paragon Plus Environment

Page 26 of 34

Page 27 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 1

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 2

ACS Paragon Plus Environment

Page 28 of 34

Page 29 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 3

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 4

ACS Paragon Plus Environment

Page 30 of 34

Page 31 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

Figure 5

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 34

Table 1. Overview of nanoparticle universal influenza A vaccines. Overview of nanoparticle universal influenza A vaccines Vaccine types Vectors Antigen types Synthetic polymers

PLGA

poly-γ-glutamic acid/chitosan VLPs

Enveloped Hepatitis B core

Self-assembling nanoparticles

Metal nanoparticles

MaMV Tobacco mosaic virus coat protein Papaya mosaic virus F88 bacteriophage Woodchuck hepatitis VLP vectored in Salmonella Typhimurium T7 bacteriophage Q-β Ferritin Self-Antigen Gold nanoparticle

Immunogenicity readout in animal models Pigs, mice,

References

Mice

63, 64

Mice

83, 84, 85, 86, 87 18, 80

M2e M2e

Mice, pigs, ferrets, human Canine Mice

M2e

Mice

92

M2e

Mice

81

M2e

Mice

94

M2e M2e Headless HA; M2e M2e, headless HA; M2e-NA tetramer; NP M2e/CpG

Mice Mice Mice, ferrets Mice

82 88 30, 97, 99 31, 47, 87, 100, 101

Mice

108

Norovirus P particle containing M2e, Peptides of HA, NP and PA; outermembrane vesicles displaying M2e M2, HA fusion peptide; H1N1 split vaccine Headless HA; M2e; M2e/FliC; M2e

ACS Paragon Plus Environment

57, 58, 59

88 89

Page 33 of 34 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Infectious Diseases

For Table of Contents Use Only

ACS Paragon Plus Environment

ACS Infectious Diseases 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

159x80mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 34 of 34