Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Biochemical and Biophysical Research Communications 554 (2021) 166e172

Contents lists available at ScienceDirect

Biochemical and Biophysical Research Communications


journal homepage: www.elsevier.com/locate/ybbrc

Nasalesubcutaneous primeeboost regimen for inactivated whole-


virus influenza vaccine efficiently protects mice against both upper
and lower respiratory tract infections
Meito Shibuya a, b, c, Shigeyuki Tamiya a, b, c, Atsushi Kawai a, b, c, Yasuo Yoshioka a, b, c, d, e, *
a
Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka,
565-0871, Japan
b
Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka
University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
c
Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1
Yamadaoka, Suita, Osaka, 565-0871, Japan
d
The Research Foundation for Microbial Diseases of Osaka University (BIKEN), 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
e
Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan

a r t i c l e i n f o a b s t r a c t

Article history: Although influenza vaccines are effective for reducing viral transmission and the severity of clinical
Received 17 February 2021 symptoms, influenza viruses still induce considerable morbidity and mortality worldwide. Seasonal
Accepted 18 March 2021 influenza viruses infect the upper respiratory tract initially but then often induce severe pulmonary
Available online 30 March 2021
complications in the lower respiratory tract. Therefore, influenza vaccines that prevent viral infection at
both the upper and lower respiratory tracts are highly anticipated. Here, we examined whether using
Keywords:
different vaccination routes for priming and boosting achieved protection in both regions of the respi-
IgA
ratory tract. To this end, we used inactivated whole-virion influenza vaccines to immunize mice either
IgG
Influenza virus
subcutaneously or intranasally for both priming and boosting. Regardless of the route used for boosting,
Respiratory tract the levels of virus-specific IgG in plasma were higher in mice primed subcutaneously than those in
Vaccine control mice, which received PBS only. In addition, intranasal priming followed by subcutaneous
boosting induced higher levels of virus-specific IgG in plasma than those in control mice. The levels of
virus-specific nasal IgA were higher in mice that were primed intranasally than in control mice or in mice
primed subcutaneously. Furthermore, intranasal priming but not subcutaneous priming provided pro-
tection against viral challenge in the upper respiratory tract. In addition, when coupled with subcu-
taneous boosting, both subcutaneous and intranasal priming protected against viral challenge in the
lower respiratory tract. These results indicate that intranasal priming followed by subcutaneous boosting
induces both virus-specific IgG in plasma and IgA in nasal washes and protects against virus challenge in
both the upper and lower respiratory tracts. Our results will help to develop novel vaccines against
influenza viruses and other respiratory viruses.
© 2021 Elsevier Inc. All rights reserved.

1. Introduction

Vaccines for influenza viruses are an effective way to reduce the


severity of clinical symptoms and to limit infection and trans-
mission [1,2]. To prevent viral infection of cells and virus release
Abbreviations: Cal7, A/California/07/2009; HA, hemagglutinin; NA, neuramini- from infected cells, influenza vaccines must induce neutralizing
dase; PBS, phosphate-buffered saline; SD, standard deviation; SV, split vaccine; Th,
antibodies specific for hemagglutinin (HA) and neuraminidase
CD4þ T helper; TLR, Toll-like receptor; WV, inactivated whole-virion vaccine.
* Corresponding author. Vaccine Creation Group, BIKEN Innovative Vaccine (NA), the major surface glycoproteins of influenza virus [1,2].
Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Several types of influenza vaccines, such as split vaccines (SVs) and
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan. inactivated whole-virion vaccines (WVs), are available for use in
E-mail address: y-yoshioka@biken.osaka-u.ac.jp (Y. Yoshioka).

https://doi.org/10.1016/j.bbrc.2021.03.099
0006-291X/© 2021 Elsevier Inc. All rights reserved.
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

Fig. 1. Antibody responses after using different antigens for priming and boosting. Mice were immunized twice subcutaneously with SV or WV for priming and boosting.
Control mice were treated with PBS subcutaneously only. After the second immunization, levels of HA- and NA-specific total IgG in plasma were evaluated by using ELISA. We used
160- ((C), 800- (A), and 4,000- (:) foldediluted plasma samples. n ¼ 5 per group. Data are shown as means ± 1 SD. ##P < 0.01, ####P < 0.0001 vs. control group; **P < 0.01,
***P < 0.001, ****P < 0.0001 between indicated groups according to Tukey’s test. Significance of differences was analyzed for the 160-fold-diluted plasma samples only.

humans [1,2]. SVs are generated by treating purified viruses with mucosal administration, for priming and boosting reportedly in-
ether or detergents (or both) to remove the viral lipid envelope; the duces antigen-specific antibody and T-cell responses both sys-
HA and NA proteins are then isolated and used as SVs [2]. For WVs, temically and mucosally, compared with either parenteral or
purified viruses are inactivated by treating with formalin or b- mucosal vaccination only [13e18]. Therefore, to induce strong
propiolactone (or both) [2,3]. In Japan, a WV with adjuvant is virus-specific antibody responses in both the systemic and nasal
already licensed for use in adults as a vaccine against H5N1 influ- compartments, combining parenteral and nasal vaccination for
enza virus [4]. WVs retain the original viral structure and have priming and boosting is a rational strategy. In this study, we used an
single-stranded viral RNA, which is a natural ligand for Toll-like influenza WV as an antigen to examine the efficacy of combining
receptor 7 (TLR7) [5,6]. Therefore, due to their stimulation of different routes of immunization to induce virus-specific anti-
TLR7, WVs generally induce stronger viral-specific antibody and T- bodies in both the nasal cavity and in blood. We demonstrated that
cell responses than do SVs. the vaccination regimen combining intranasal priming with sub-
Despite the development of several types of influenza vaccines, cutaneous boosting achieves antibody responses in both com-
influenza viruses still are a threat to humans and a major infectious partments and provides strong protection against virus challenge
source of morbidity and mortality worldwide, indicating the need in both the upper and lower respiratory tracts.
for influenza vaccines with improved efficacy [1,2]. In humans, the
upper respiratory tract is the initiation site for infection by seasonal 2. Materials and methods
influenza viruses. Although infection of the upper respiratory tract
induces relatively mild illness, this infection is crucial for human- 2.1. Mice
to-human transmission of virus via coughing, talking, or sneezing
[1,7]. Infection of the lower respiratory tract due to influenza virus Male C57BL/6J mice (age, 6e7 weeks; SLC, Kyoto, Japan) were
from the upper respiratory tract often induces severe clinical housed in a room with a 12:12-h light:dark cycle and had unre-
complications, including pneumonia [1]. Therefore, one ideal stricted access to food and water. All animal experiments were
strategy is to develop vaccines that can block virus infection at both conducted in accordance with the guidelines of Osaka University
the upper and lower respiratory tracts. for the ethical treatment of animals and were approved by the
The major mechanism of defense against viral infections in the Animal Care and Use Committee of the Research Institute for Mi-
upper respiratory tract is the production of virus-specific IgA an- crobial Diseases, Osaka University, Japan (protocol number, BIKEN-
tibodies at the nasal surface [8,9]. However, most current influenza AP-H26-11-0).
vaccines, which are administered parenterally through subcu-
taneous or intramuscular injection, fail to stimulate the production
of virus-specific IgA in the upper respiratory tract, despite inducing 2.2. Vaccines and influenza viruses
high levels of virus-specific IgG in blood [8,9]. Therefore, these
vaccines generally do not prevent viral infection at the upper res- An ether-treated HA-antigeneenriched virion-free SV and a
piratory tract, although the leakage of virus-specific IgG into the formalin-inactivated WV from an H1N1 influenza virus (strain: A/
lower respiratory tract can prevent the aggravation of illness. In California/07/2009 [Cal7]) were kindly provided by Dr. Yasuyuki
contrast, intranasal vaccination can induce not only virus-specific Gomi (The Research Foundation for Microbial Diseases of Osaka
IgG in blood but also virus-specific IgA in the upper respiratory University (BIKEN), Kagawa, Japan). H1N1 influenza virus (strain:
tract [8,9]. In fact, many studies have demonstrated that nasal Cal7) was generously provided by Dr. Hideki Asanuma (National
influenza vaccines prevent upper respiratory tract infection more Institute of Infectious Diseases, Tokyo, Japan).
efficiently than do parenteral vaccines [10e12]. However, nasal
vaccines do not induce virus-specific IgG in blood as strongly as 2.3. Recombinant HA and NA proteins
parenteral vaccines [8,9]. In this regard, novel vaccination strategies
might overcome this drawback of nasal vaccines and increase the Recombinant HA and NA proteins derived from Cal7 were
levels of virus-specific IgG in blood. generated as described previously [19]. Briefly, the ectodomain of
Combining different vaccination routes, such as parenteral and HA, which carried a C-terminal hexahistidine tag and the foldon of
fibritin from bacteriophage T4, and the ectodomain of NA, which
167
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

Fig. 2. Level of IgG in plasma according to vaccination regimen. Mice were immunized twice with WV subcutaneously (s.c.) or intranasally (i.n.). Control mice were treated with
PBS subcutaneously only. Levels of (a) WV-specific total IgG, IgG1, IgG2b, and IgG2c in plasma and (b) HA-specific total IgG in plasma were evaluated after the second vaccination.
We used (a) 4,000- ((C), 20,000- (A), and 100,000- (:) foldediluted plasma samples and (b) 160- ((C), 800- (A), and 4,000- (:) foldediluted plasma samples. n ¼ 5 per group.
Data are shown as means ± 1 SD. #P < 0.05, ##P < 0.01, ####P < 0.0001 vs. control group; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 between indicated groups according to
Tukey’s test. Significance of differences was analyzed for the (a) 4,000-foldediluted and (b) 160-foldediluted plasma samples only.

had a C-terminal hexahistidine tag and an N-terminal tetrabrachion subcutaneously with phosphate-buffered saline (PBS) only. At 7
tetramerization domain from the bacterium Staphylothermus days after the second dose, samples of plasma and nasal wash were
marinus, were expressed by using the Expi293 Expression System collected and stored at 30  C until use. Nasal wash samples were
(Thermo Fisher Scientific, Hampton, NH, USA). collected by gently flushing the nasal cavity with 400 mL of PBS.

2.4. Vaccination 2.5. Antibody responses

For subcutaneous vaccination, mice were immunized at the base The levels of WV-, recombinant HA-, and recombinant NA-
of the tail by using SV (1 mg/mouse) or WV (1 mg/mouse) in a total specific antibodies in the plasma and nasal wash samples were
volume of 50 mL. For nasal vaccination, anesthetized mice were analyzed by using ELISA, as described previously [20]. Briefly, 10 mg/
immunized with WV (1 mg/mouse) in total volume of 5 mL (2.5 mL to mL WV in PBS, 1 mg/mL recombinant HA in carbonate buffer, or
each nostril). For both routes, mice were immunized by using two 1 mg/mL recombinant NA in carbonate buffer were added to the
doses at a 21-day interval. Control mice were treated wells of 96-well plates, which were incubated overnight at 4  C. The
168
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

Fig. 3. Level of IgA in nasal wash according to vaccination regimen. Mice were immunized twice with WV subcutaneously or intranasally, as described in Fig. 2. Levels of WV-
specific IgA and HA-specific IgA in nasal wash were evaluated after the second vaccination. We used 2- (C), 10- (A), and 50- (:) foldediluted nasal wash samples. n ¼ 5 per
group. Data are shown as means ± 1 SD. #P < 0.05, ####P < 0.0001 vs. control group; *P < 0.05, ***P < 0.001, ****P < 0.0001 between indicated groups according to Tukey’s test.
Significance of differences was analyzed for the 2-foldediluted nasal wash samples only.

antigen-coated plates were then incubated with Block Ace (DS to assess the benefit of combining different antigens for priming
Pharma Biomedical, Osaka, Japan), and diluted plasma and nasal and boosting to induce antibody responses. Mice were immunized
wash samples were added. The plates were then washed three subcutaneously with SV or WV twice, after which we measured the
times with PBS containing 0.05% Tween 20 and incubated with levels of recombinant HA- and NA-specific total IgG in plasma
horseradish peroxidaseeconjugated goat anti-mouse IgG (Merck (Fig. 1). The levels of HA- and NA-specific total IgG were signifi-
Millipore, Darmstadt, Germany), IgG1 (SouthernBiotech, Birming- cantly higher in mice primed with WV than in mice primed with SV
ham, AL, USA), IgG2b (SouthernBiotech), or IgG2c (South- but did not depend on the antigen used for boosting (Fig. 1). In
ernBiotech) or horseradish peroxidaseeconjugated goat anti- particular, mice primed with WV and boosted with SV showed
mouse IgA (SouthernBiotech). Plates were washed again, tetra- significantly higher levels of HA- and NA-specific total IgG than
methyl benzidine (Nacalai Tesque, Kyoto, Japan) was added, the mice primed with SV followed by WV (Fig. 1). These results suggest
color reaction was stopped by adding 2 N H2SO4, and the absor- the importance of the priming antigen for the strength of immune
bance was measured at OD450e570 in a microplate reader (Power responses. In addition, consistent with previous results, WV is su-
Wave HT, BioTek, Winooski, VT, USA). perior to induce virus-specific antibody responses compared with
SV [3,21]. Therefore, we focused on WV as the antigen in subse-
2.6. Infection quent experiments.
Next, to examine the effect of varying the administration routes
At 10 days after the second immunization, mice were anes- for priming and boosting on antibody responses, we allocated mice
thetized and intranasally challenged with Cal7. For upper respira- into 4 groups. Group 1 was immunized subcutaneously for both
tory tract infection, mice were challenged by using 3.0  105 TCID50 priming and boosting. Group 2 was immunized subcutaneously for
of Cal7 in a total volume of 5 mL of PBS (2.5 mL to each nostril). After priming and intranasally for boosting. Group 3 was immunized
3 days, nasal washes were collected by using 400 mL of PBS, and the intranasally for priming and subcutaneously for boosting. Group 4
virus titers in nasal wash were evaluated by using MDCK cells, as was immunized intranasally for both priming and boosting. WV
described previously [20]. For lower respiratory tract infection, from Cal7 was used as an antigen.
mice were challenged by using 3.0  105 TCID50 of Cal7 in 30 mL of Nasal vaccination in mice often involves the administration of
PBS (15 mL to each nostril). The body weights and survival of the large volumes of fluid (e.g., 20e40 mL). However, excess vaccine
challenged mice were monitored for 15 days after challenge. We solution can flow from the upper respiratory tract to the lower
defined the day on which mice weighed less than 75% of their initial respiratory tract, thus stimulating immune responses at both sites.
body weight as the day of death. Because this experimental condition might not reflect the actual
situation for nasal vaccination in humans, we instead used only 5 mL
2.7. Statistical analyses (2.5 mL for each nostril) as the inoculation volume for intranasal
vaccination. Mice were immunized twice with WV, and we
Statistical analyses were performed by using Prism (GraphPad measured the levels of WV- and HA-specific total IgG, IgG1, IgG2b,
Software, San Diego, CA, USA). All data are presented as means with and IgG2c in plasma (Fig. 2) and of WV- and HA-specific IgA in nasal
standard deviations. Significant differences were determined by wash (Fig. 3). The levels of WV-specific total IgG, IgG2b, and IgG2c
Tukey’s test. Significant differences in survival rates were deter- and of HA-specific total IgG all showed the same tendency (Fig. 2).
mined by using the log-rank test to compare KaplaneMeier curves. Mice in group 1, group 2, and group 3 had significantly higher levels
A P value less than 0.05 was considered to indicate statistical of WV-specific total IgG, IgG2b, and IgG2c and HA-specific total IgG
significance. than those in the control group (which received PBS subcutane-
ously only), but these antibody levels did not differ between control
3. Results and discussion mice and those in group 4 (Fig. 2). In particular, group 1 and group 3
had significantly higher levels of WV-specific total IgG, IgG2b, and
First, we used SV or WV from H1N1 influenza A virus (strain: A/ IgG2c and HA-specific total IgG than group 4 (Fig. 2). The levels of
California/07/2009 [Cal7]) as an antigen for priming and boosting WV-specific IgG1 in group 1 and group 2 were significantly higher

169
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

than in controls and group 4, whereas the amounts of WV-specific the post-challenge virus titer in nasal wash as the indicator of
IgG1 were similar between group 3 and control mice (Fig. 2a). vaccine efficacy (Fig. 4a). The virus titers in mice primed subcuta-
In contrast, the quantities of WV- and HA-specific IgA in nasal neously, namely group 1 and group 2, did not differ from that in
wash were significantly higher in mice that were primed intrana- control mice (Fig. 4a). In contrast, virus titers in nasal wash were
sally, that is, group 3 and group 4, than in control mice and those significantly lower in mice immunized intranasally for priming,
primed subcutaneously, namely group 1 and group 2 (Fig. 3). In that is, group 3 and group 4, compared with mice in group 1, group
particular, group 4 had the highest levels of WV- and HA-specific 2, and the control group (Fig. 4a). These results suggest that the
IgA in nasal wash among all groups (Fig. 3). Collectively, these re- preventive effect in the upper respiratory tract is correlated with
sults suggest that the priming site plays a crucial role in defining the ability to stimulate the production of virus-specific IgA in the
the strength of the IgG response in blood and of the IgA response in upper respiratory tract, thus indicating the usefulness of nasal
nasal wash, and the vaccination regimen for group 3 induced both vaccine for priming, regardless of the route used for boosting.
virus-specific IgG in plasma and IgA in nasal wash, albeit at mod- We next investigated the protective effects of vaccination in the
erate levels. lower respiratory tract, instilling 30 mL of Cal7 virus in the nares as a
To examine the protective effects of vaccination in the upper post-vaccination challenge dose. In this model, we assessed body
respiratory tract, we used a 5-mL volume of Cal7 virus as an intra- weight loss and survival as indicators of vaccine efficacy (Fig. 4b
nasal challenge dose after vaccination. In this model, we measured and c). All of the control mice died within 10 days after challenge

Fig. 4. Protective effects against influenza virus. (a) At 10 days after the second vaccination, mice were challenged with Cal7 to cause upper respiratory tract infection. At 3 days
after challenge, virus titers in nasal wash samples were evaluated. (b, c) At 10 days after the second vaccination, mice were challenged with Cal7 to cause lower respiratory tract
infection. Percentage change in (b) initial body weight and (c) survival were monitored after viral challenge. n ¼ 4 or 5 per group. Data are shown as means ± 1 SD. (a) ##P < 0.01,
###
P < 0.001 vs. control group; ***P < 0.001, ****P < 0.0001 between indicated groups according to Tukey’s test. (c) ##P < 0.01 vs. control group according to comparison of
KaplaneMeier curves by using the log-rank test.

170
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

(Fig. 4b and c). Mice in group 4 rapidly lost weight, and only 50% References
survived (Fig. 4b and c). In contrast, all of the mice in group 1, group
2, and group 3 survived after viral challenge, and none of them lost [1] F. Krammer, G.J.D. Smith, R.A.M. Fouchier, M. Peiris, K. Kedzierska,
P.C. Doherty, P. Palese, M.L. Shaw, J. Treanor, R.G. Webster, A. Garcia-Sastre,
weight (Fig. 4b and c). These results imply that the preventive effect Influenza Nat. Rev. Dis. Primers 4 (2018) 3.
in the lower respiratory tract is correlated with the ability to induce [2] C.J. Wei, M.C. Crank, J. Shiver, B.S. Graham, J.R. Mascola, G.J. Nabel, Next-
virus-specific IgG in blood and that both subcutaneous and nasal generation influenza vaccines: opportunities and challenges, Nat. Rev. Drug
Discov. 19 (2020) 239e252.
vaccination for priming followed by subcutaneous boosting protect [3] T. Sekiya, E.J. Mifsud, M. Ohno, N. Nomura, M. Sasada, D. Fujikura, T. Daito,
against virus challenge in the lower respiratory tract. Collectively, M. Shingai, Y. Ohara, T. Nishimura, M. Endo, R. Mitsumata, T. Ikeda,
our results indicate that intranasal administration for priming fol- H. Hatanaka, H. Kitayama, K. Motokawa, T. Sobue, S. Suzuki, Y. Itoh, L.E. Brown,
K. Ogasawara, Y. Kino, H. Kida, Inactivated whole virus particle vaccine with
lowed by subcutaneous inoculation for boosting protects against potent immunogenicity and limited IL-6 induction is ideal for influenza,
virus challenge in both the upper and lower respiratory tracts. Vaccine 37 (2019) 2158e2166.
Our data showed that the protection against virus challenge in [4] T. Nakayama, T. Kumagai, K.J. Ishii, T. Ihara, Alum-adjuvanted H5N1 whole
virion inactivated vaccine (WIV) induced IgG1 and IgG4 antibody responses in
the lower respiratory tract accomplished through nasal vaccination
young children, Vaccine 30 (2012) 7662e7666.
for priming followed by subcutaneous vaccination (group 3) is [5] S. Koyama, T. Aoshi, T. Tanimoto, Y. Kumagai, K. Kobiyama, T. Tougan,
comparable to that of the traditional use of subcutaneous vacci- K. Sakurai, C. Coban, T. Horii, S. Akira, K.J. Ishii, Plasmacytoid dendritic cells
delineate immunogenicity of influenza vaccine subtypes, Sci. Transl. Med. 2
nation for both priming and boosting (group 1) (Fig. 4b and c).
(2010) 25ra24.
However, the levels of WV-specific total IgG and IgG1 were [6] T. Onodera, A. Hosono, T. Odagiri, M. Tashiro, S. Kaminogawa, Y. Okuno,
significantly lower in group 3 than in group 1, despite the equiva- T. Kurosaki, M. Ato, K. Kobayashi, Y. Takahashi, Whole-virion influenza vac-
lent levels of WV-specific IgG2b and IgG2c in these groups (Fig. 2). cine recalls an early burst of high-affinity memory B cell response through TLR
signaling, J. Immunol. 196 (2016) 4172e4184.
Previous reports have shown that, in mice, virus-specific IgG2 are [7] M. Richard, J.M.A. van den Brand, T.M. Bestebroer, P. Lexmond, D. de Meulder,
more protective against influenza virus challenge than IgG1 [22]. R.A.M. Fouchier, A.C. Lowen, S. Herfst, Influenza A viruses are transmitted via
Thus, the strong protection against virus challenge in the lower the air from the nasal respiratory epithelium of ferrets, Nat. Commun. 11
(2020) 766.
respiratory tract of group 3 might result from virus-specific IgG2b [8] T. Azegami, Y. Yuki, H. Kiyono, Challenges in mucosal vaccines for the control
and IgG2c. of infectious diseases, Int. Immunol. 26 (2014) 517e528.
Although the protection achieved in the upper respiratory tract [9] H. Yusuf, V. Kett, Current prospects and future challenges for nasal vaccine
delivery, Hum. Vaccines Immunother. 13 (2017) 34e45.
was comparable between group 3 and group 4, group 3 had [10] A. Ainai, S. Tamura, T. Suzuki, E. van Riet, R. Ito, T. Odagiri, M. Tashiro,
significantly lower levels virus-specific IgA in nasal wash (Fig. 3). In T. Kurata, H. Hasegawa, Intranasal vaccination with an inactivated whole
general, the magnitude of antibody response differs among influenza virus vaccine induces strong antibody responses in serum and nasal
mucus of healthy adults, Hum. Vaccines Immunother. 9 (2013) 1962e1970.
parenteral (e.g., subcutaneous, intramuscular, intradermal)
[11] T. Suzuki, A. Kawaguchi, A. Ainai, S. Tamura, R. Ito, P. Multihartina,
administration routes [23,24]. Therefore, modifying the parenteral V. Setiawaty, K.N. Pangesti, T. Odagiri, M. Tashiro, H. Hasegawa, Relationship
inoculation regimen for boosting after intranasal priming might of the quaternary structure of human secretory IgA to neutralization of
influenza virus, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) 7809e7814.
enhance the production of virus-specific IgA in the upper respira-
[12] A. Ainai, T. Suzuki, S.I. Tamura, H. Hasegawa, Intranasal administration of
tory tract. whole inactivated influenza virus vaccine as a promising influenza vaccine
In summary, we show here that using different routes for candidate, Viral Immunol. 30 (2017) 451e462.
vaccination priming and boosting, especially intranasal priming [13] H. Asanuma, F. Koide, Y. Suzuki, T. Nagamine, C. Aizawa, T. Kurata, S. Tamura,
Cross-protection against influenza virus infection in mice vaccinated by
and subcutaneous boosting, may be an optimal regimen for pro- combined nasal/subcutaneous administration, Vaccine 13 (1995) 3e5.
tecting against viral infection in both the upper and lower respi- [14] H. Asanuma, C. Aizawa, T. Kurata, S. Tamura, IgA antibody-forming cell re-
ratory tracts. Because many pathogenic viruses, including influenza sponses in the nasal-associated lymphoid tissue of mice vaccinated by
intranasal, intravenous and/or subcutaneous administration, Vaccine 16
viruses and SARS-CoV-2, enter through the nasal surface and (1998) 1257e1262.
induce severe pulmonary damage in the lower respiratory tract, [15] F. Fiorino, E. Pettini, G. Pozzi, D. Medaglini, A. Ciabattini, Prime-boost strate-
preventing infection at both sites likely is an effective strategy for gies in mucosal immunization affect local IgA production and the type of th
response, Front. Immunol. 4 (2013) 128.
combating such pathogens [25]. Therefore, the concept that we [16] A. Ciabattini, G. Prota, D. Christensen, P. Andersen, G. Pozzi, D. Medaglini,
demonstrate here will further the development of novel vaccines Characterization of the antigen-specific CD4(þ) T cell response induced by
against influenza viruses and other respiratory viruses. prime-boost strategies with CAF01 and CpG adjuvants administered by the
intranasal and subcutaneous routes, Front. Immunol. 6 (2015) 430.
[17] H.S. Hwang, S. Puth, W. Tan, V. Verma, K. Jeong, S.E. Lee, J.H. Rhee, More robust
gut immune responses induced by combining intranasal and sublingual
routes for prime-boost immunization, Hum. Vaccines Immunother. 14 (2018)
Declaration of competing interest 2194e2202.
[18] C.B. Roces, M.T. Hussain, S.T. Schmidt, D. Christensen, Y. Perrie, Investigating
Y.Y. is employed by the Research Foundation for Microbial Dis- prime-pull vaccination through a combination of parenteral vaccination and
intranasal boosting, Vaccines (2019) 8. Basel.
eases (Osaka University, Japan). The other authors have no conflicts [19] S. Shirai, M. Shibuya, A. Kawai, S. Tamiya, L. Munakata, D. Omata, R. Suzuki,
of interests to declare. T. Aoshi, Y. Yoshioka, Lipid nanoparticles potentiate CpG-
oligodeoxynucleotide-based vaccine for influenza virus, Front. Immunol. 10
(2019) 3018.
[20] M. Shibuya, T. Aoshi, E. Kuroda, Y. Yoshioka, Murine cross-reactive non-
neutralizing polyclonal IgG1 antibodies induced by influenza vaccine inhibit
Acknowledgments the cross-protective effect of IgG2 against heterologous virus in mice, J. Virol.
(2020) 94.
This study was supported by grants from the Japan Society for [21] K. Sato, Y. Takahashi, Y. Adachi, H. Asanuma, M. Ato, M. Tashiro, S. Itamura,
Efficient protection of mice from influenza A/H1N1pdm09 virus challenge
the Promotion of Science (JSPS KAKENHI grant no. JP20H03404 to Y. infection via high avidity serum antibodies induced by booster immunizations
Yoshioka), Nagai Memorial Research Scholarship from the Phar- with inactivated whole virus vaccine, Heliyon 5 (2019), e01113.
maceutical Society of Japan (to S. Tamiya), and BIKEN Taniguchi [22] V.C. Huber, R.M. McKeon, M.N. Brackin, L.A. Miller, R. Keating, S.A. Brown,
N. Makarova, D.R. Perez, G.H. Macdonald, J.A. McCullers, Distinct contributions
Scholarship (to A. Kawai). We thank Dr. Yasuyuki Gomi (The
of vaccine-induced immunoglobulin G1 (IgG1) and IgG2a antibodies to pro-
Research Foundation for Microbial Diseases of Osaka University) for tective immunity against influenza, Clin. Vaccine Immunol. 13 (2006)
providing the split vaccine and inactivated whole-virion vaccine 981e990.
[23] L. Zhang, W. Wang, S. Wang, Effect of vaccine administration modality on
and Dr. Hideki Asanuma (National Institute of Infectious Diseases)
immunogenicity and efficacy, Expert Rev. Vaccines 14 (2015) 1509e1523.
for providing influenza viruses.
171
M. Shibuya, S. Tamiya, A. Kawai et al. Biochemical and Biophysical Research Communications 554 (2021) 166e172

[24] S.T.T. Schetters, L.J.W. Kruijssen, M.H.W. Crommentuijn, H. Kalay, J.M.M. den H.H. Hoffmann, T.Y. Oliveira, D.A. Oren, V. Ramos, L. Nogueira, E. Michailidis,
Haan, Y. van Kooyk, Immunological dynamics after subcutaneous immuni- D.F. Robbiani, A. Gazumyan, C.M. Rice, T. Hatziioannou, P.D. Bieniasz,
zation with a squalene-based oil-in-water adjuvant, Faseb. J. 34 (2020) M. Caskey, M.C. Nussenzweig, Enhanced SARS-CoV-2 neutralization by
12406e12418. dimeric IgA, Sci. Transl. Med. 13 (2021).
[25] Z. Wang, J.C.C. Lorenzi, F. Muecksch, S. Finkin, C. Viant, C. Gaebler, M. Cipolla,

172

You might also like