ArticlePDF Available

Intranasal Vaccination with 1918 Influenza Virus-Like Particles Protects Mice and Ferrets from Lethal 1918 and H5N1 Influenza Virus Challenge

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

Influenza vaccines capable of inducing cross-reactive or heterotypic immunity could be an important first line of prevention against a novel subtype virus. Influenza virus-like particles (VLPs) displaying functional viral proteins are effective vaccines against replication-competent homologous virus, but their ability to induce heterotypic immunity has not been adequately tested. To measure VLP vaccine efficacy against a known influenza pandemic virus, recombinant VLPs were generated from structural proteins of the 1918 H1N1 virus. Mucosal and traditional parenteral administrations of H1N1 VLPs were compared for the ability to protect against the reconstructed 1918 virus and a highly pathogenic avian H5N1 virus isolated from a fatal human case. Mice that received two intranasal immunizations of H1N1 VLPs were largely protected against a lethal challenge with both the 1918 virus and the H5N1 virus. In contrast, mice that received two intramuscular immunizations of 1918 VLPs were only protected against a homologous virus challenge. Mucosal vaccination of mice with 1918 VLPs induced higher levels of cross-reactive immunoglobulin G (IgG) and IgA antibodies than did parenteral vaccination. Similarly, ferrets mucosally vaccinated with 1918 VLPs completely survived a lethal challenge with the H5N1 virus, while only a 50% survival rate was observed in parenterally vaccinated animals. These results suggest a strategy of VLP vaccination against a pandemic virus and one that stimulates heterotypic immunity against an influenza virus strain with threatening pandemic potential.
Content may be subject to copyright.
JOURNAL OF VIROLOGY, June 2009, p. 5726–5734 Vol. 83, No. 11
0022-538X/09/$08.000 doi:10.1128/JVI.00207-09
Copyright © 2009, American Society for Microbiology. All Rights Reserved.
Intranasal Vaccination with 1918 Influenza Virus-Like Particles
Protects Mice and Ferrets from Lethal 1918 and H5N1
Influenza Virus Challenge
Lucy A. Perrone,
1
Attiya Ahmad,
2
Vic Veguilla,
1
Xiuhua Lu,
1
Gale Smith,
2
Jacqueline M. Katz,
1
Peter Pushko,
2
and Terrence M. Tumpey
1
*
Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases,
Collaborating Centers for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia,
1
and
Novavax, Inc., Rockville, Maryland
2
Received 28 January 2009/Accepted 17 March 2009
Influenza vaccines capable of inducing cross-reactive or heterotypic immunity could be an important first
line of prevention against a novel subtype virus. Influenza virus-like particles (VLPs) displaying functional
viral proteins are effective vaccines against replication-competent homologous virus, but their ability to induce
heterotypic immunity has not been adequately tested. To measure VLP vaccine efficacy against a known
influenza pandemic virus, recombinant VLPs were generated from structural proteins of the 1918 H1N1 virus.
Mucosal and traditional parenteral administrations of H1N1 VLPs were compared for the ability to protect
against the reconstructed 1918 virus and a highly pathogenic avian H5N1 virus isolated from a fatal human
case. Mice that received two intranasal immunizations of H1N1 VLPs were largely protected against a lethal
challenge with both the 1918 virus and the H5N1 virus. In contrast, mice that received two intramuscular
immunizations of 1918 VLPs were only protected against a homologous virus challenge. Mucosal vaccination
of mice with 1918 VLPs induced higher levels of cross-reactive immunoglobulin G (IgG) and IgA antibodies
than did parenteral vaccination. Similarly, ferrets mucosally vaccinated with 1918 VLPs completely survived
a lethal challenge with the H5N1 virus, while only a 50% survival rate was observed in parenterally vaccinated
animals. These results suggest a strategy of VLP vaccination against a pandemic virus and one that stimulates
heterotypic immunity against an influenza virus strain with threatening pandemic potential.
Influenza A viruses represent a substantial public health
burden, with a yearly average of more than 220,000 hospitaliza-
tions and approximately 36,000 deaths in the United States alone
(http://www.cdc.gov/flu/keyfacts.htm). In addition to seasonal out-
breaks caused by antigenic variants of circulating influenza A and
B viruses, another pandemic influenza virus strain may emerge at
any time. The threat of a pandemic is greater than it has been in
decades. Confirmed cases of human infection with several sub-
types of avian influenza viruses have been reported since 1997 (2,
3, 38; http://www.who.int/csr/disease/avian_influenza/country/en
/index.html). Of the avian subtypes that have recently been intro-
duced into humans, the highly pathogenic avian influenza virus
H5N1 subtype is the most immediate public health problem.
More than 400 human H5N1 virus infections have occurred,
and approximately 60% have been fatal (http://www.who.int/csr
/disease/avian_influenza/country/en/index.html). Should these vi-
ruses acquire the ability to spread efficiently among humans lack-
ing immunity to the H5 hemagglutinin (HA), a pandemic could
occur. If the case fatality rate of the virus remained high, an H5
pandemic could recapitulate the devastating consequences of the
“Spanish” influenza pandemic of 1918, which resulted in an esti-
mated 50 million deaths and a 10-year reduction in the average
life expectancy in the United States (13). Although recent studies
suggest that most 1918 (H1N1) pandemic deaths were attributed
to secondary bacterial pneumonia (5), the inherent ability of the
virus to replicate efficiently and cause severe acute infection of the
respiratory tract was a critical underlying cause of this historic
public health disaster (24, 26, 28, 50, 72, 74, 78, 79). Thus, the 1918
influenza virus represents an ideal candidate for the study of
protective immunity to a pandemic influenza virus strain.
Traditional influenza vaccines provide optimal protection
against viruses that are antigenically closely matched with
those contained in the vaccine but have been less effective
against antigenic variants within a subtype and historically pro-
vide only minimal protection against viruses of novel HA sub-
types (1). Thus, there has been interest in developing a vaccine
or vaccine strategy that can induce broader cross-reactive im-
munity against multiple subtypes of influenza viruses contain-
ing multiple combinations of surface proteins, also known as
heterosubtypic immunity. In addition to a decrease in overall
morbidity following infection, heterosubtypically immune ani-
mals show decreased viral titers and duration of viral shedding
within the respiratory tract (23, 27, 36, 54, 65, 70, 75).
Pandemic influenza vaccines must meet a number of criteria,
which include low production cost, ease of manufacture, and
rapid production and delivery. In recent years, several dif-
ferent approaches have been tested. A promising technology
uses recombinant noninfectious virus-like particles (VLPs)
that present structurally native, immunologically relevant viral
antigens. VLP vaccines have proven effective in preventing
diseases in humans, as exemplified by the recently approved
human papillomavirus VLP vaccine for the prevention of cer-
vical cancer (21; http://www.fda.gov/cber/vaccines.htm). Non-
* Corresponding author. Mailing address: 1600 Clifton Rd. NE, MS
G-16, Atlanta, GA 30333. Phone: (404) 639-5444. Fax: (404) 639-2350.
E-mail: tft9@cdc.gov.
Published ahead of print on 25 March 2009.
5726
infectious VLPs morphologically resemble their live-virus
counterparts and are recognized and processed readily by an-
tigen-presenting cells of the immune system (4, 30, 60, 69, 76,
81). Recombinant VLPs do not involve the use of infectious
influenza virus and thus require no exceptional biosafety con-
tainment to produce and can be manufactured quickly for an
emergency response. Influenza VLPs have been generated in
insect cells by using three influenza virus proteins, i.e., HA,
neuraminidase (NA), and matrix (M1). To date, these VLPs
have been produced from the H1N1, H3N2, H5N1, H5N3, and
H9N2 subtypes (6, 7, 19, 41, 51–53). Unlike conventional split
and inactivated vaccines based only on HA content, VLPs
contain known quantities of the NA and M1 proteins. There-
fore, VLPs may have a greater potential for eliciting cross-
reactive antibody and T-cell immunity against newly emerging
antigenic drift (61) and shift variants (71). Influenza VLPs
have been shown to induce high neutralizing antibody titers
against homologous and heterologous strains in mice and fer-
rets (6, 7, 41, 53, 54), and a phase I clinical trial with an
investigational H5N1 VLP vaccine is ongoing (6).
In this study, we evaluated a 1918 VLP vaccine for the ability
to protect mice against the 1918 pandemic virus in the hope
that such a preclinical evaluation may pave the way for a 1918
vaccine which could be offered to laboratory workers working
with the virus to mitigate biosafety concerns. In addition, the
ability of the 1918 (H1N1) VLP vaccine to elicit protective
heterotypic immunity against a lethal challenge with a contem-
porary highly pathogenic avian influenza H5N1 virus was as-
sessed in mice and ferrets. The ferret model of influenza dis-
ease has been used to evaluate H5N1 virus virulence previously
(17, 40, 82), as well as the safety and efficacy of other H5N1
vaccine candidates (6, 18, 20, 35, 41). Mice immunized intra-
nasally (i.n.) with the 1918 VLP vaccine were protected not
only from death caused by the homologous 1918 virus but also
from a heterotypic H5N1 virus, whereas mice immunized par-
enterally were only protected against a homotypic 1918 virus
challenge. A study with ferrets further demonstrates that mu-
cosal VLP vaccination was clearly superior to parenteral vac-
cination for the induction of heterotypic immunity against an
H5N1 virus. Although subtype-cross-reactive neutralizing an-
tibodies were not associated with mouse and ferret survival,
high circulating and respiratory mucosal immunoglobulin G
(IgG) and IgA antibody levels correlated with heterotypic im-
munity protection in mice.
MATERIALS AND METHODS
Viruses and cells. The replication-competent influenza viruses used in these
experiments included (i) the reconstructed 1918 H1N1 (abbreviated 1918) virus
(72) possessing the A/South Carolina/1/18 HA and (ii) the A/Vietnam/1203/2004
H5N1 (abbreviated VN/1203) virus previously shown to be highly virulent for
both mice and ferrets (42, 74). The 1918 virus was generated with the 12-plasmid
reverse genetics system in a mixture of Madin-Darby canine kidney (MDCK;
ATCC, Manassas, VA) and 293T cells (ATCC) as previously described (72). The
VN/1203 virus was grown in embryonating hen’s eggs. All virus stock titers were
determined by plaque assay on MDCK cells, and virus stocks were maintained in
Dulbecco’s modified Eagle’s medium culture (Gibco, Grand Island, NY) sup-
plemented with 10% fetal calf serum (HyClone, Logan, UT) and 1% penicillin/
streptomycin (Gibco). All virus challenge experiments were performed under the
guidance of the U.S. National Select Agent Program in negative-pressure
HEPA-filtered biosafety level 3(BSL-3) enhanced laboratories with the use
of a battery-powered Racal HEPA filter respirator and according to Biomedical
Microbiological and Biomedical Laboratory procedures (58).
VLP generation. 1918 VLPs were made with recombinant baculovirus express-
ing the genes for HA, NA, and M1. The genes for HA, NA, and M1 used for the
generation of the VLPs were synthesized by reverse transcription-PCR with viral
RNA extracted from reconstructed 1918 influenza virus (72). Following reverse
transcription-PCR, the genes for the NA, M1, and HA cDNAs were combined in
this order within the pFastBac1 transfer vector (Invitrogen) essentially as de-
scribed previously (52). This resulted in a plasmid, pNVAX1250, that encoded
the genes for HA, NA, and M1, each within its own expression cassette that
included a polyhedrin promoter and transcription termination sequences. The
DNA sequences of the genes and flanking regulatory sequences were confirmed.
The HA gene was identical to that of influenza A virus A/South Carolina/1/18
(H1N1) (GenBank AAD17229), whereas the NA and M1 genes were identical to
the influenza A/Brevig Mission/1/18 virus genes (accession no. AAF77036 and
AAN06597, respectively). The DNA fragment from pNVAX1250 containing the
NA, M1, and HA expression cassettes was then transferred into bacmids and
transfected into Spodoptera frugiperda (Sf9) insect cells (ATCC CRL-1711) to
generate recombinant baculovirus (52). Recombinant baculoviruses were used to
infect Sf9 cells. VLPs were harvested at 72 h posttransfection. Recombinant
VLPs were purified to approximately 90% by sucrose density gradient ultra-
centrifugation, followed by ion-exchange chromatography. The influenza viral
proteins in the VLP preparations were confirmed by sodium dodecyl sulfate-
polyacrylamide electrophoresis and Western blot analysis. Semiquantitative
densitometry of the Coomassie-stained gel containing the purified VLP prepa-
ration showed that the HA, NA, and M1 proteins were present at a ratio of
approximately 61:5:22. The remaining 12% of the Coomassie-stained material
contained impurities derived from Sf9 cells and baculovirus. A single-radial-
immunodiffusion assay was used to quantitate the HA content of the VLPs as
described previously (80). Reference reagents for the single-radial-immunodif-
fusion standard curve included A/New Caledonia/20/99 (H1N1) IVR-116 (79
g/ml of HA) antigen and anti-HA reference sheep antiserum S-6706 (obtained
from the Center for Biologics Evaluation and Research, Food and Drug Admin-
istration, Rockville, MD). Control VLPs derived from human immunodeficiency
virus (HIV)-type Con-S gp145 (77) were prepared and quantitated similarly.
Mouse vaccination and virus challenge. All animal research was conducted
according to the guidance of the CDC Institutional Animal Care and Use
Committee in an Association for Assessment and Accreditation of Laboratory
Animal Care International-accredited facility. Eight- to 10-week-old female
BALB/c mice (Jackson Laboratories, Bar Harbor, ME) were anesthetized by
intraperitoneal injection of 0.2 ml of 2,2,2-tribromoethanol in tert-amyl alcohol
(Avertin; Sigma-Aldrich, Milwaukee, WI) (73) and vaccinated either i.n. or
intramuscularly (i.m.) (right hind leg) with 5 g (based on HA content) of 1918
VLPs in 50 l of phosphate-buffered saline (PBS). Fourteen days post primary
vaccination, mice were boosted with a second dose of 5 g of 1918 VLPs by
either the i.m. or the i.n. route. Mice were bled for collection of serum prior to
and following the first and second vaccinations. Lung and nasal washes were
collected as previously described in 1 ml of sterile PBS for antibody titration (75).
Two weeks post boost vaccination, mice were challenged i.n. with 50 50% lethal
doses (LD
50
) of either the 1918 or the VN/1203 virus in a volume of 50 l. Mice
were weighed daily and observed for illness. On day 5 post virus challenge (p.c.),
four mice per group were exsanguinated and euthanatized and their lungs were
removed for virus titration. Lungs were homogenized in 1 ml of sterile PBS, and
clarified homogenate virus titers were determined by a standard plaque assay for
virus infectivity. Briefly, 10-fold serial dilutions of lung homogenate were placed
onto confluent monolayers of MDCK cells in the presence of tosylsulfonyl
phenylalanyl chloromethyl ketone (TPCK) trypsin (1 g/ml). Following1hof
adsorption, cells were washed with PBS and a 2x L-15 medium (Cambrex/Lonza
Inc., Walkersville, MD) agar overlay was placed into each well. The statistical
significance of virus titer data was determined by using the analysis-of-variance
(ANOVA) test. Mouse survival data and mean time to death (MTD) were
subjected to Kaplan-Meier survival analysis with SPSS software.
Ferret vaccination and virus challenge. Eighteen male Fitch ferrets, 10 months
of age (Triple F Farms, Sayre, PA), that were serologically negative by hemag-
glutination inhibition (HI) assay for currently circulating influenza viruses were
used in these studies. 1918 VLP vaccinations were conducted under BSL-2
conditions, and H5N1 virus challenges were performed under BSL-3condi-
tions within a Duo-Flo Bioclean unit (Lab Products, Seaford, DE) with a max-
imum capacity of 18 ferrets. Serum samples, nasal washes, and temperature and
weight measurements were taken throughout the duration of the study. Tem-
peratures were monitored following the virus challenge by a subcutaneously
implanted transponder (BioMedic Data Systems, Seaford, DE). Prior to the
study, baseline serum was collected via the anterior vena cava from all 18 ferrets.
Six ferrets were vaccinated with 15 g/500 l 1918 VLPs by the i.n. route (250 l
in each nostril), and six ferrets were vaccinated by the i.m. route. The remaining
VOL. 83, 2009 HETEROTYPIC PROTECTION BY MUCOSAL VLP VACCINATION 5727
six ferrets received PBS in place of vaccine by the i.n. and i.m. routes. Fourteen
days following the initial VLP or PBS inoculation, serum was collected from all
of the ferrets and they were then boosted with 15 g/500 l by the same route
used previously. Fourteen days following the VLP boost, all 18 ferrets were
challenged i.n. with 50 50% ferret infectious doses in 1 ml of A/VN/1203/04
(H5N1) virus. Fifty percent ferret infectious doses were determined by inocu-
lating groups of ferrets each at 10
4
,10
3
,10
2
, and 10
1
50% egg infective doses and
calculating the outcome of infection by the method of Reed and Muench (57).
Ferrets were weighed daily and monitored for signs of illness. Nasal cavities of
infected ferrets were washed with 1 ml of PBS every other day, beginning on day
2 p.i., and measured for virus replication by 10-fold serial titration in hen’s eggs.
Serology. Sera collected from mice and ferrets prior to a virus challenge were
treated with receptor-destroying enzyme (RDE; Denka Seiken, Tokyo, Japan)
and tested for reactivity to the -propiolactone-inactivated VN/1203 and 1918
viruses by the HI assay with 1.0% horse or 0.5% turkey red blood cells, respec-
tively, starting with a serum dilution of 1:10. Briefly, virus stocks were treated
with 0.1% -propiolactone (Sigma-Aldrich) for 24 h at 4°C. Stocks were safety
tested by two serial passages in embryonating eggs before use in the HI assays.
Geometric mean titers (GMTs) and their standard deviations were calculated for
each treatment group. For the microneutralization assays, 100 50% tissue culture
infective doses of infectious VN/1203 or 1918 virus was incubated with twofold
serial dilutions of sera, starting at a 1:10 dilution, and incubated overnight on
MDCK cells (59). Titers represent the reciprocal dilution of serum giving
50% neutralization activity. An indirect enzyme-linked immunosorbent assay
(ELISA) was performed on RDE-treated mouse sera to determine the IgA and
IgG responses following vaccination (75). Briefly, IgA and IgG titers were mea-
sured with purified, formalin-inactivated VN/1203 and recombinant 1918HA/
NA:A/Texas/36/91 (49) viruses. Immulon microtiter plates (Thermo Scientific
Inc., Waltham, MA) were coated with 100 hemagglutination units of each virus,
and mouse antibody was detected with horseradish peroxidase-conjugated anti-
body (KPL Laboratories, Gaithersburg, MD). The ELISA endpoint titers were
expressed as the highest dilutions that yielded an optical densities greater than
the mean optical density plus 3 standard deviations of similarly diluted negative
control sera. Titers are expressed in the log
2
format, and the statistical signifi-
cance of differences between treatment groups was measured by the ANOVA
test.
RESULTS
Mucosal, but not parenteral, VLP vaccination induces het-
erotypic immunity in mice. 1918 (H1N1) VLPs generated in a
baculovirus expression system (Fig. 1A) from Sf9 cells were
purified from culture medium and negatively stained for elec-
tron microscopic examination. As shown in Fig. 1B, 1918 VLPs
had a diameter of approximately 80 to 120 nm with pro-
nounced surface protein spikes and a morphology characteris-
tic of replication-competent 1918 virions (Fig. 1C). We com-
pared the mucosal (i.n.) and parenteral (i.m.) vaccination
routes for the ability to induce protection against a homolo-
gous 1918 virus challenge, as well as an H5N1 heterotypic virus
challenge. BALB/c mice were vaccinated twice with 5 gof
1918 VLPs or control HIV VLPs and challenged i.n. 14 days
post VLP boost with 50 LD
50
of either VN/1203 (H5N1) or
1918 (H1N1) virus (Fig. 2). The extent of vaccine protection
was measured by (i) morbidity (weight loss), (ii) survival over
a 14-day p.c. period, and (iii) virus titers in the lower respira-
tory tract (lung) at 5 days p.c. Mice vaccinated with 1918 VLPs
by either the i.n or the i.m. vaccination route uniformly sur-
vived the lethal challenge with the homologous 1918 virus,
while control animals succumbed to infection (MTD 8.8
days; *, P0.05, Kaplan-Meier survival analysis) (Fig. 2A, left
side). In contrast to the control animals, mice in both vaccina-
tion groups showed no signs of sickness and gained weight
every day p.c. although mucosally vaccinated mice gained, on
average, 5% more body mass over time than parenterally vac-
cinated mice (Fig. 2B, left side). The 1918 virus replicated
efficiently in the lungs of control mice, with titers of approxi-
mately 5.7 log
10
PFU/ml (Fig. 2C, left column). However, 1918
VLP-vaccinated/1918 virus-challenged mice did not have de-
tectable lung virus titers on day 5 p.c.
In contrast to the homologous subtype challenge data, all mice
that received the 1918 VLPs by the i.m route succumbed to a
VN/1203 (H5N1) heterotypic virus challenge (MTD 6.4 days),
as did the HIV VLP control mice (MTD 5.5 days) (Fig. 2A,
right column). Moreover, parenterally vaccinated mice exhibited
dramatic weight loss from 2 days p.c. until death, similar to those
animals vaccinated with the HIV VLPs (Fig. 2B, right column). In
contrast, five of six mice that received the same dose of 1918 VLP
vaccine administered i.n. were protected against an H5N1 het-
erotypic virus challenge (Fig. 2A, right column). The surviving
mice exhibited morbidity that reached a mean maximum weight
loss of 17% on day 5 p.c. before weight gain was observed from
day 6 p.c. onward (Fig. 2B, right column). The mean lung virus
titers of mice administered 1918 VLP vaccine by the i.n. route
were approximately 300-fold lower than those of control mice
receiving PBS or HIV VLP vaccine (Fig. 2C, right column). Mice
vaccinated i.m. and challenged with the H5N1 virus exhibited
titers of nearly 10
6
PFU/ml, only twofold lower than those of
control mice. Collectively, these results demonstrate that mucosal
H1N1 VLP immunization provides greater heterotypic immunity
against the H5N1 virus than does parenteral VLP vaccination.
Mucosal vaccination with 1918 VLPs results in higher IgG
and IgA antibody titers in mice than does parenteral vaccina-
tion. Characterization of postvaccination antibody responses was
performed in attempts to identify cross-reactive antibodies that
may be responsible for conferring protection against a lethal
H5N1 heterotypic virus challenge. First, the virus neutralization
(v.n.) antibody titers against the 1918 (H1N1) and VN/1203
(H5N1) viruses were measured in pooled sera (n8or9per
group) collected from mice vaccinated twice with the 1918 VLPs
(Table 1). Mice vaccinated by either the i.n or the i.m. route
displayed v.n. antibody titers against the homologous 1918 virus
FIG. 1. 1918 VLP generation. VLPs were constructed in a baculo-
virus genetic background with the sequences of the genes for HA, NA,
and M1 of the 1918 pandemic virus and produced from Sf9 cells.
(A) Baculovirus construct for the expression of 1918 influenza VLPs.
Indicated are the polyhedrin promoter (PolH), polyadenylation signal
(pA), and influenza virus genes. 1918 VLPs had an average diameter
of 100 nm, as illustrated in this negatively stained transmission electron
micrograph. While not replication competent, the 1918 VLPs (B) mor-
phologically resemble the reconstructed 1918 virions that were col-
lected from supernatants of 1918 virus-infected MDCK cell cultures
(C). Mag, magnification.
5728 PERRONE ET AL. J. VIROL.
(1:800 to 1:1,600) but exhibited no detectable serum v.n. antibody
titer against the heterotypic H5N1 virus. As expected, mice vac-
cinated with the HIV VLPs had no detectable serum v.n. antibody
titer (10) to either influenza virus. The HI antibody response
among individual mice was also measured 2 weeks after the vac-
cine boost. Similarly, vaccination of mice with 1918 VLPs elicited
a detectable HI antibody response against the homologous 1918
virus and the mucosally vaccinated mice elicited a GMT (individ-
ual sera ranged from 320 to 1,280; GMT 607) that was 3.4-fold
greater than that of the parenterally vaccinated mice (range, 80 to
320; GMT 180). However, sera from either 1918 VLP vacci-
nation group did not exhibit any cross-reactivity to the H5N1 virus
in the HI assay (10). Mice vaccinated with HIV VLPs or given
PBS did not exhibit any HI antibody titer (10) against either the
1918 or the VN/1203 virus.
We next performed ELISAs to investigate the presence of
H5N1-cross-reactive IgG and IgA antibodies in serum samples
and respiratory washes from mice administered two doses of
the 1918 H1N1 VLP vaccine (Fig. 3). Antiviral IgG and IgA
antibodies in the sera and mucosal washes of i.n. and i.m.
vaccinated mice were detected by using ELISA plates coated
with purified whole H1N1 or heterologous H5N1 virus. Over-
all, 1918 VLP immunization by either route induced IgG an-
tibody to the homologous 1918 virus in serum and respiratory
FIG. 2. 1918 VLP vaccine efficacy in mice following a lethal H1N1 or H5N1 virus challenge. Mice were vaccinated with 5 g/50 l 1918 VLPs or
control HIV VLPs and challenged with 50 LD
50
of either 1918 (H1N1) (left column) or VN/1203 (H5N1) (right column) virus. Mice were monitored
daily for 14 days p.c. Survival rates (A) following the challenge were calculated based percent survival within each experimental group (n6 mice per
experimental group; *,P0.05, Kaplan-Meier survival analysis). Mice were weighed daily (B) to measure morbidity (loss of 25% of the original body
weight necessitates euthanasia). Average weights in each treatment group were measured for the duration of the study, and percent original body weight
was calculated based on the average starting weight for each group on day 0. On day 5 p.c., four mice per experimental group were euthanized and their
lungs were removed for virus titration (C). Lungs were homogenized in 1 ml PBS and clarified before titer determination by standard plaque assay on
MDCK cells in duplicate (limit of detection, 10 PFU). Bars represent the average titer for each experimental group; error bars indicate the standard
deviation between mice in each respective group (n4 mice per experimental group; *,P0.05, ANOVA).
TABLE 1. Neutralizing and HI antibody titers from mouse sera
Vaccination group
(route)
c
Neutralizing
antibody titer
a
HI antibody titer
b
1918 H5N1 1918 H5N1
1918 VLP (i.n.) 1,600 10 607 10
1918 VLP (i.m.) 800 10 180 10
HIV VLP (i.n.) 10 10 10 10
PBS (i.n. or i.m.) 10 10 10 10
a
Mouse serum samples from each treatment group were pooled and tested for
neutralizing antibody activity against either the 1918 or the VN/1203 virus in a
microneutralization assay with MDCK cells. The titers represents the reciprocal
serum dilution with virus-neutralizing activity.
b
HI assays used either 0.5% turkey (BPL-1918 virus) or 1% horse (BPL-VN/
1203 virus) red blood cells. The titers shown are GMTs.
c
Mice were treated twice with 1918 VLPs, HIV VLPs, or PBS.
5729
mucosa washes and these samples also exhibited cross-reactive
IgG titers against the H5N1 virus (Fig. 3A). As shown in Fig.
3A (top), i.n. vaccinated mice exhibited significantly more
H5N1-cross-reactive serum IgG antibody than did mice vacci-
nated by the parenteral route (ˆ, P0.05). Although homol-
ogous virus serum IgG titers were similar in both 1918 VLP
vaccination groups (GMT [log
2
]20.3 [i.m.] versus 21.5
[i.n.]), i.n. vaccination resulted in lung mucosal IgG antibody
titers against the homologous H1N1 (*, P0.05) and heter-
ologous H5N1 (ˆ, P0.05) viruses that were significantly
higher than those measured in i.m. vaccinated mice (Fig. 3A,
middle). This trend was also observed in nasal wash samples, in
which the differences between the IgG titers of the two vacci-
nation groups were even more pronounced (* and ˆ, P0.05)
(Fig. 3A, bottom). IgA antibodies to whole H1N1 and H5N1
virus were also examined. As shown in Fig. 3B, mucosal ad-
ministration of 1918 VLPs induced significantly higher homol-
ogous virus IgA titers in serum (top), and particularly lung
(middle) and nasal mucosal (bottom) washes, than did paren-
teral VLP vaccination (*, P0.05). Likewise, circulating levels
of cross-reactive IgA antibody to the H5N1 virus were signif-
icantly higher (ˆ, P0.05) in mice vaccinated by the i.n. route
(GMT [log
2
]10.3) than in those vaccinated by the i.m. route
(GMT [log
2
]2.9) (Fig. 3B, top). The differences in IgA titer
between these two vaccination groups was most pronounced in
lung wash samples (Fig. 3B, middle), and these trends were
also observed in nasal wash samples (Fig. 3B, bottom) (ˆ, P
0.05). Together, these results show that vaccination with 1918
H1N1 VLPs induced a population of HI- and v.n.-negative,
cross-reactive IgG and IgA antibodies that were greater following
mucosal vaccination than following parenteral vaccination.
Ferrets mucosally vaccinated with 1918 VLPs are protected
from a lethal H5N1 virus challenge. Because of its natural
susceptibility to influenza A viruses, the ferret is currently
accepted as an excellent mammalian host for influenza vaccine
efficacy studies. We next compared mucosal versus traditional
parenteral administration of the 1918 VLP vaccine for the
ability to induce heterotypic immunity in this species. Ferrets
were vaccinated twice with 15 g of the 1918 VLPs either i.n.
or i.m. and subsequently challenged 14 days post VLP boost
with 50 LD
50
of VN/1203 (H5N1) virus. Strikingly, all of the six
ferrets vaccinated with the 1918 VLPs i.n. survived a lethal
challenge with the H5N1 virus (Table 2), whereas 50% of the
ferrets vaccinated by the i.m. route and 100% of the control
animals succumbed to the lethal H5N1 virus infection. More-
over, mucosally vaccinated ferrets also exhibited less weight
loss, as a group, over time than did ferrets vaccinated by the i.m
route or with PBS alone (Fig. 4A). Ferrets vaccinated by the
i.m route did exhibit a longer survival time (MTD 8.3 days)
than did PBS-inoculated control animals (MTD 6.3 days).
While there were no significant temperature change differ-
ences observed between ferrets in the i.n. and i.m. vaccination
groups, overall the vaccinated animals exhibited less fever than
did the PBS-inoculated control animals (Fig. 4B). Ferrets vac-
cinated i.n. also exhibited reduced virus titers in their nasal
mucosa following infection than did the i.m. vaccinated ferrets,
and the differences between these two groups were statistically
significant on days 2, 4, 6, and 8 p.c. (Fig. 4C). All vaccinated
FIG. 3. Antibody responses in 1918 VLP-vaccinated mice. BALB/c mice were vaccinated with 1918 VLPs or HIV VLPs or given PBS. Serum,
lung wash, and nasal wash sample IgG (A) and IgA (B) antibody titers were determined following two vaccinations with 1918 VLPs. Sera was
treated with RDE, and all of the samples were individually tested in fourfold serial dilutions against the formalin-inactivated 1918 and VN/1203
viruses in an antibody capture ELISA. Plots show the geometric mean antibody titers (log
2
) of all of the mice in each treatment group, and error
bars indicate the standard deviation observed among the mice in each group. Significant differences between the antibody titers elicited by the
different vaccination routes are indicated (*[1918] and ˆ [VN/1203], P0.05, ANOVA).
5730 PERRONE ET AL. J. VIROL.
ferrets exhibited lower virus titers than did the PBS-inoculated
animals, indicating that while the i.m. vaccinated ferrets were
not completely protected against death, VLP vaccination still
induced an immune response capable of suppressing some
virus replication in the upper respiratory tract. The HI anti-
body responses to the H1N1 (1918) and H5N1 (VN/1203)
viruses were measured in individual serum samples collected
from ferrets (n6) vaccinated twice with 1918 VLPs. Ferrets
vaccinated by the i.n route had slightly higher circulating HI
antibody titers to the homologous virus (GMT 254) than did
i.m. vaccinated ferrets (GMT 160) (Table 2). HI antibody
against the H5N1 virus was not detected in either group of
VLP-vaccinated ferrets. Collectively, these results from the
ferret challenge model confirm the experimental outcomes
observed in the mouse studies, showing that mucosal influenza
immunization with 1918 VLPs can provide greater heterotypic
immunity than parenteral influenza vaccination.
DISCUSSION
The emergence of new influenza virus subtypes to which the
human population has little immunity is of great public health
concern, and vaccination remains the principal and most econom-
ically prudent public health intervention strategy against both
seasonal and pandemic influenza. The ability of the 1918 (H1N1
subtype) virus to spread rapidly and cause high rates of human
illness makes it an ideal candidate for studying vaccine efficacy
against a pandemic strain. Furthermore, scientists working with
the reconstructed 1918 virus would benefit from the development
of and access to an effective vaccine against this recognized killer.
The high mortality rate associated with human infections by
H5N1 viruses (15, 25, 34; http://www.who.int/csr/disease/avian
_influenza/country/en/index.html) highlights the need for the de-
velopment of improved vaccine technologies. To date, clinical
evaluations of many H5N1 vaccine candidates indicate the need
for improved adjuvants or alternative approaches that could en-
hance vaccine immunogenicity (31–33, 46, 47). Moreover, vaccine
strategies capable of inducing more cross-reactive immunity may
overcome limitations in vaccine efficacy imposed by antigenic
variability of influenza A viruses (11). VLPs have been generated
previously from a number of subtypes and have been shown to
promote a strong immune response to a challenge with a homol-
ogous virus (14, 41, 52, 53); however, their capacity to induce
heterotypic immunity has not been adequately addressed.
In the present study, we tested the ability of nonadjuvanted
1918 VLPs to provide protection against the reconstructed 1918
pandemic virus, as well as elicit cross-protection against a lethal
H5N1 virus challenge. Two routes of vaccination (mucosal and
parenteral) were compared to assess the potential effect of the
route of VLP administration on vaccine efficacy, and two mam-
malian models of highly pathogenic influenza disease were used
(39, 42, 72, 74). All of the mice vaccinated with the 1918 VLPs
and lethally challenged with the 1918 virus survived and were well
protected, regardless of the vaccination route, supporting results
from previous homologous virus challenge studies with baculo-
virus-expressed influenza VLPs (4, 41, 53, 55). Importantly, these
studies showed that mucosal VLP vaccination was superior to
parenteral vaccination for the induction of heterotypic immunity.
The cross-protective effect of mucosal vaccination was associated
with a reduction in weight loss and reduced H5N1 virus replica-
tion in the respiratory mucosa.
Antibodies are prime candidates for contributors to het-
erotypic immunity because they are generated to conserved
antigens, and further support comes from studies in which
FIG. 4. Ferret morbidity following 1918 VLP vaccination and an
H5N1 virus challenge. Eighteen ferrets were divided into three exper-
imental groups. Six ferrets were vaccinated by the i.n. or i.m. route with
15 g/500 ml 1918 VLPs. Six control ferrets received the same volume
of PBS i.n. plus i.m. Twenty-eight days following the administration of
two doses of 1918 VLPs or PBS, ferrets were challenged i.n. with 50
LD
50
of the A/Vietnam/1203/04 (H5N1) virus and monitored daily for
signs of illness. The ferrets were weighed (A) and their temperatures
were measured individually (B) for 14 days p.c. Control ferrets all
succumbed to infection by day 6 p.c. (†). The nasal cavities of infected
ferrets were washed with 1 ml PBS every other day beginning on day
2 p.c., and virus titers were determined (C). Differences in virus titers
between vaccination groups were analyzed by ANOVA (*,P0.005).
TABLE 2. Ferret mortality following 1918 VLP vaccination and
lethal H5N1 challenge
Group No. of
ferrets
No. that
survived virus
challenge
(% mortality)
MTD (days
postchallenge)
Serum HI
antibody GMT
prior to virus
challenge
1918 H5N1
VLP i.n. 6 6 (0) NA
a
254 20
VLP i.m. 6 3 (50) 8.3 180 20
PBS 6 0 (100) 6.3 20 20
a
NA, not applicable.
VOL. 83, 2009 HETEROTYPIC PROTECTION BY MUCOSAL VLP VACCINATION 5731
B-cell-deficient mice failed to survive a heterotypic chal-
lenge (45, 75). In the present study, characterization of the
postvaccination antibody responses identified differences
between the two routes of vaccination. ELISAs revealed the
presence of H5N1-cross-reactive IgG and IgA antibodies
among H1N1 VLP-vaccinated animals, suggesting the pres-
ence of common cross-reactive epitopes in the HA, NA, or
M1 protein. Mice vaccinated by the mucosal route generally
had higher levels of antiviral IgG and IgA antibodies than did
parenterally vaccinated mice. The differences in cross-reactive
antibody titers between the two vaccination groups were most
pronounced in the lung and upper respiratory tract. Polymeric
IgA antibody may contribute to heterotypic immunity by its
ability to pass through the lung epithelium and, during this
transcytosis, interfere with the production of viral proteins in
an infected cell (43, 44). However, heterotypic immunity can
be effective in IgA
/
mice, suggesting that this class of anti-
body is unlikely playing a role (11). Cross-reactive IgG anti-
body was found in the lungs along with IgA and has been
considered a correlate of heterotypic protection (75). In mice,
mucosal but not parenteral vaccination induced subtype-cross-
reactive lung and serum IgG anti-HA antibodies, suggesting
the presence of a common cross-reactive epitope in the HA of
the H3 and H5 subtypes (75).
Although mucosally vaccinated animals were largely pro-
tected against a lethal H5N1 virus challenge, neutralizing an-
tibody against the H5N1 virus was not detected in any group of
VLP-vaccinated animals. It is conceivable that the cross-reac-
tive anti-HA antibodies produced in VLP-vaccinated hosts act
by additional mechanisms in vivo to neutralize progeny virus
and/or enhance the clearance of virus-infected cells (11, 16, 37,
75). While the identity between the HA1 amino acid sequences
of the VN/1203 H5N1 and 1918 viruses is only 58%, antibodies
to the conserved HA2 stem region are generated and may
affect overall virus binding and immune recognition (13, 48,
64). More recent support for our findings presented here
showed that neutralizing antibody epitopes exist in the stem
region of the HA molecule and that while antibody bound to
the HA2 region does not classically “neutralize” the receptor
binding site, it prevents the molecule from conformationally
changing upon endocytosis and exposure to a low pH, thereby
preventing viral fusion with the cell membrane (9, 48, 63, 64,
67). It is reasonable to speculate that mucosal vaccination with
VLPs generates a population of v.n.-negative, cross-reactive
IgG and IgA antibodies to regions of the virus HA molecule
that promote virus clearance. A cross-reactive epitope(s) on
the NA may also be generating nonneutralizing antibodies.
Anti-NA antibodies can partially reduce the viral yield and
plaque size of influenza viruses from infected cells. The 1918
and VN/1203 viruses exhibited 85% identity in their NA amino
acid sequences, and N1-specific antibodies have been shown to
provide partial cross-protection against H5N1 virus infection
(61). Studies are under way to delineate the cross-reactive
epitope(s) on the HA and NA glycoproteins.
Mucosal vaccination with these VLPs may also be inducing
a stronger protective immune response because of the inherent
stimulation of innate immune sentinels such as Toll-like recep-
tors on the mucosal surface (22), as these VLPs contain viral
surface proteins in their native conformation and it is pre-
sumed that these particles are capable of binding to cells ex-
pressing the viral receptor -2-3 or -2-6 sialic acid, as do in-
fectious virus particles (66, 74). Mucosal vaccination may be
better than parenteral administration at delivering viral anti-
gens to the pertinent anatomical compartment, specifically to
dendritic cells in the respiratory tissue which play a critical role
in communicating with T and B cells to develop immunological
memory against influenza viruses (62). Supporting this hypoth-
esis is our observation that mice vaccinated mucosally express
twofold greater amounts of interleukin-2 (IL-2) (a cytokine
released by activated CD4
T cells to allow for clonal expan-
sion of antigen-specific T cells and also a potent activator of B
cells) in the lungs 5 days following an H5N1 virus challenge
than do mice vaccinated parenterally (data not shown), which
may contribute to the overall survival associated with this vac-
cination group. Also elevated in mucosally VLP-vaccinated/
H5N1-challenged surviving mice were the cytokines IL-9 and
IL-17 (data not shown), both of which are recognized to facil-
itate B-cell proliferation. The broadly cross-neutralizing anti-
bodies reported recently were of the IgG1 subclass (10, 67),
B-cell products that help mediate the Th2 CD4
T-cell and
cytokine response, including IL-9 and L-10 production. IL-10
was recently shown to be important in regulating pulmonary
inflammation in influenza virus-infected mice, and inhibition
of this cytokine results in severe lung inflammation (68). We
found that levels of IL-10 in lungs from VLP-vaccinated mice
were three times higher in those infected with the H5N1 virus
than in 1918 virus-challenged mice (data not shown), possibly
contributing to their survival. While the protective role of
CD8
cytotoxic T lymphocytes and memory B cells following
1918 VLP vaccination warrants further investigation, previous
studies on the induction of heterotypic immunity has revealed
CD8
T cells to be generally accessory to mouse survival
following a lethal virus challenge, while B cells are critical (8,
12, 45, 56, 75).
Our studies raise important questions regarding the appli-
cation of this vaccination technology, in both seasonal and
epidemic outbreak situations. Specifically, could mucosal ad-
ministration of influenza VLP vaccine bearing seasonal or pan-
demic influenza virus proteins reduce the widespread morbid-
ity and lethality due to a newly emerging subtype prior to the
production of a strain-specific vaccine? Other research has
indicated that this may be possible (23, 29). A vaccine that
could induce or boost heterotypic immunity through the stim-
ulation of a cross-reactive antibody could be an important
preventative measure against a novel subtype, allowing time
for the development of a pandemic strain-specific vaccine.
ACKNOWLEDGMENTS
L.A.P. was supported by a fellowship from the American Society for
Microbiology and the CDC Coordinating Center for Infectious Dis-
eases.
We thank the Vietnamese Ministry of Health for use of the A/Viet-
nam/1203/04 isolate and Jessica Belser for the ferret LD
50
determina-
tion of that virus stock. We thank Debra Wadford, Neal Van Hoeven,
Joshua DeVos, and Ebonee Butler for providing reagents and assisting
with the serological assays. We also thank Ye Liu and Tom Kort for
expert assistance in VLP purification and Feng Lui for his assistance in
statistical analysis of mouse mortality data.
The findings and conclusions in this report are ours and do not
necessarily represent the views of the funding agency.
We have no competing interests to report.
5732 PERRONE ET AL. J. VIROL.
REFERENCES
1. Ada, G. L., and P. D. Jones. 1986. The immune response to influenza
infection. Curr. Top. Microbiol. Immunol. 128:1–54.
2. Belser, J. A., O. Blixt, L. M. Chen, C. Pappas, T. R. Maines, N. Van Hoeven,
R. Donis, J. Busch, R. McBride, J. C. Paulson, J. M. Katz, and T. M.
Tumpey. 2008. Contemporary North American influenza H7 viruses possess
human receptor specificity: implications for virus transmissibility. Proc. Natl.
Acad. Sci. USA 105:7558–7563.
3. Belser, J. A., X. Lu, T. R. Maines, C. Smith, Y. Li, R. O. Donis, J. M. Katz,
and T. M. Tumpey. 2007. Pathogenesis of avian influenza (H7) virus infec-
tion in mice and ferrets: enhanced virulence of Eurasian H7N7 viruses
isolated from humans. J. Virol. 81:11139–11147.
4. Bessa, J., N. Schmitz, H. J. Hinton, K. Schwarz, A. Jegerlehner, and M. F.
Bachmann. 2008. Efficient induction of mucosal and systemic immune re-
sponses by virus-like particles administered intranasally: implications for
vaccine design. Eur. J. Immunol. 38:114–126.
5. Beveridge, W. I. 1991. The chronicle of influenza epidemics. Hist. Philos. Life
Sci. 13:223–234.
6. Bright, R. A., D. M. Carter, C. J. Crevar, F. R. Toapanta, J. D. Steckbeck,
K. S. Cole, N. M. Kumar, P. Pushko, G. Smith, T. M. Tumpey, and T. M.
Ross. 2008. Cross-clade protective immune responses to influenza viruses
with H5N1 HA and NA elicited by an influenza virus-like particle. PLoS
ONE 3:e1501.
7. Bright, R. A., D. M. Carter, S. Daniluk, F. R. Toapanta, A. Ahmad, V.
Gavrilov, M. Massare, P. Pushko, N. Mytle, T. Rowe, G. Smith, and T. M.
Ross. 2007. Influenza virus-like particles elicit broader immune responses
than whole virion inactivated influenza virus or recombinant hemagglutinin.
Vaccine 25:3871–3878.
8. Droebner, K., E. Haasbach, C. Fuchs, A. O. Weinzierl, S. Stevanovic, M.
Buttner, and O. Planz. 2008. Antibodies and CD4
T-cells mediate cross-
protection against H5N1 influenza virus infection in mice after vaccination
with a low pathogenic H5N2 strain. Vaccine 26:6965–6974.
9. Ekiert, D. C., G. Bhabha, M. A. Elsliger, R. H. Friesen, M. Jongeneelen, M.
Throsby, J. Goudsmit, and I. A. Wilson. 26 February 2009. Antibody recog-
nition of a highly conserved influenza virus epitope. Science doi:10.1126/
science.1171491.
10. Epstein, S. L. 2003. Control of influenza virus infection by immunity to
conserved viral features. Expert Rev. Anti. Infect. Ther. 1:627–638.
11. Epstein, S. L., A. Stack, J. A. Misplon, C. Y. Lo, H. Mostowski, J. Bennink,
and K. Subbarao. 2000. Vaccination with DNA encoding internal proteins of
influenza virus does not require CD8
cytotoxic T lymphocytes: either CD4
or CD8
T cells can promote survival and recovery after challenge. Int.
Immunol. 12:91–101.
12. Fleury, D., B. Barrere, T. Bizebard, R. S. Daniels, J. J. Skehel, and M.
Knossow. 1999. A complex of influenza hemagglutinin with a neutralizing
antibody that binds outside the virus receptor binding site. Nat. Struct. Biol.
6:530–534.
13. Frost, W. H. 1920. Statistics of influenza morbidity. Public Health Rep.
35:584.
14. Galarza, J. M., T. Latham, and A. Cupo. 2005. Virus-like particle (VLP)
vaccine conferred complete protection against a lethal influenza virus chal-
lenge. Viral Immunol. 18:244–251.
15. Gambotto, A., S. M. Barratt-Boyes, M. D. de Jong, G. Neumann, and Y.
Kawaoka. 2008. Human infection with highly pathogenic H5N1 influenza
virus. Lancet 371:1464–1475.
16. Gerhard, W., K. Mozdzanowska, M. Furchner, G. Washko, and K. Maiese.
1997. Role of the B-cell response in recovery of mice from primary influenza
virus infection. Immunol. Rev. 159:95–103.
17. Govorkova, E. A., J. E. Rehg, S. Krauss, H. L. Yen, Y. Guan, M. Peiris, T. D.
Nguyen, T. H. Hanh, P. Puthavathana, H. T. Long, C. Buranathai, W. Lim,
R. G. Webster, and E. Hoffmann. 2005. Lethality to ferrets of H5N1 influ-
enza viruses isolated from humans and poultry in 2004. J. Virol. 79:2191–
2198.
18. Govorkova, E. A., R. J. Webby, J. Humberd, J. P. Seiler, and R. G. Webster.
2006. Immunization with reverse-genetics-produced H5N1 influenza vaccine
protects ferrets against homologous and heterologous challenge. J. Infect.
Dis. 194:159–167.
19. Haynes, J. R., L. Dokken, J. A. Wiley, A. G. Cawthon, J. Bigger, A. G.
Harmsen, and C. Richardson. 2009. Influenza-pseudotyped Gag virus-like
particle vaccines provide broad protection against highly pathogenic avian
influenza challenge. Vaccine 27:530–541.
20. Hoffmann, E., A. S. Lipatov, R. J. Webby, E. A. Govorkova, and R. G.
Webster. 2005. Role of specific hemagglutinin amino acids in the immuno-
genicity and protection of H5N1 influenza virus vaccines. Proc. Natl. Acad.
Sci. USA 102:12915–12920.
21. Hung, C. F., B. Ma, A. Monie, S. W. Tsen, and T. C. Wu. 2008. Therapeutic
human papillomavirus vaccines: current clinical trials and future directions.
Expert Opin. Biol. Ther. 8:421–439.
22. Ichinohe, T., A. Kawaguchi, S. Tamura, H. Takahashi, H. Sawa, A. Ninomiya,
M. Imai, S. Itamura, T. Odagiri, M. Tashiro, J. Chiba, T. Sata, T. Kurata, and
H. Hasegawa. 2007. Intranasal immunization with H5N1 vaccine plus Poly
I:Poly C
12
U, a Toll-like receptor agonist, protects mice against homologous and
heterologous virus challenge. Microbes Infect. 9:1333–1340.
23. Ichinohe, T., S. Tamura, A. Kawaguchi, A. Ninomiya, M. Imai, S. Itamura,
T. Odagiri, M. Tashiro, H. Takahashi, H. Sawa, W. M. Mitchell, D. R.
Strayer, W. A. Carter, J. Chiba, T. Kurata, T. Sata, and H. Hasegawa. 2007.
Cross-protection against H5N1 influenza virus infection is afforded by intra-
nasal inoculation with seasonal trivalent inactivated influenza vaccine. J. In-
fect. Dis. 196:1313–1320.
24. Kash, J. C., T. M. Tumpey, S. C. Proll, V. Carter, O. Perwitasari, M. J.
Thomas, C. F. Basler, P. Palese, J. K. Taubenberger, A. Garcia-Sastre, D. E.
Swayne, and M. G. Katze. 2006. Genomic analysis of increased host immune
and cell death responses induced by 1918 influenza virus. Nature 443:578–
581.
25. Kelly, T. R., M. G. Hawkins, C. E. Sandrock, and W. M. Boyce. 2008. A
review of highly pathogenic avian influenza in birds, with an emphasis on
Asian H5N1 and recommendations for prevention and control. J. Avian
Med. Surg. 22:1–16.
26. Kobasa, D., S. M. Jones, K. Shinya, J. C. Kash, J. Copps, H. Ebihara, Y.
Hatta, J. H. Kim, P. Halfmann, M. Hatta, F. Feldmann, J. B. Alimonti, L.
Fernando, Y. Li, M. G. Katze, H. Feldmann, and Y. Kawaoka. 2007. Aber-
rant innate immune response in lethal infection of macaques with the 1918
influenza virus. Nature 445:319–323.
27. Kreijtz, J. H., R. Bodewes, G. van Amerongen, T. Kuiken, R. A. Fouchier,
A. D. Osterhaus, and G. F. Rimmelzwaan. 2007. Primary influenza A virus
infection induces cross-protective immunity against a lethal infection with a
heterosubtypic virus strain in mice. Vaccine 25:612–620.
28. LeCount, E. R. 1919. The pathologic anatomy of influenzal bronchopneu-
monia. JAMA 650–652.
29. Lee, L. Y., L. A. Ha do, C. Simmons, M. D. de Jong, N. V. Chau, R.
Schumacher, Y. C. Peng, A. J. McMichael, J. J. Farrar, G. L. Smith, A. R.
Townsend, B. A. Askonas, S. Rowland-Jones, and T. Dong. 2008. Memory T
cells established by seasonal human influenza A infection cross-react with
avian influenza A (H5N1) in healthy individuals. J. Clin. Investig. 118:3478–
3490.
30. Lenz, P., P. M. Day, Y. Y. Pang, S. A. Frye, P. N. Jensen, D. R. Lowy, and J. T.
Schiller. 2001. Papillomavirus-like particles induce acute activation of den-
dritic cells. J. Immunol. 166:5346–5355.
31. Leroux-Roels, I., R. Bernhard, P. Gerard, M. Drame, E. Hanon, and G.
Leroux-Roels. 2008. Broad clade 2 cross-reactive immunity induced by an
adjuvanted clade 1 rH5N1 pandemic influenza vaccine. PLoS ONE 3:e1665.
32. Leroux-Roels, I., A. Borkowski, T. Vanwolleghem, M. Drame, F. Clement, E.
Hons, J. M. Devaster, and G. Leroux-Roels. 2007. Antigen sparing and
cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic in-
fluenza vaccine: a randomised controlled trial. Lancet 370:580–589.
33. Levie, K., I. Leroux-Roels, K. Hoppenbrouwers, A. D. Kervyn, C. Vander-
meulen, S. Forgus, G. Leroux-Roels, S. Pichon, and I. Kusters. 2008. An
adjuvanted, low-dose, pandemic influenza A (H5N1) vaccine candidate is
safe, immunogenic, and induces cross-reactive immune responses in healthy
adults. J. Infect. Dis. 198:642–649.
34. Li, F. C., B. C. Choi, T. Sly, and A. W. Pak. 2008. Finding the real case-
fatality rate of H5N1 avian influenza. J. Epidemiol. Community Health
62:555–559.
35. Li, S., C. Liu, A. Klimov, K. Subbarao, M. L. Perdue, D. Mo, Y. Ji, L. Woods,
S. Hietala, and M. Bryant. 1999. Recombinant influenza A virus vaccines for
the pathogenic human A/Hong Kong/97 (H5N1) viruses. J. Infect. Dis. 179:
1132–1138.
36. Liang, S., K. Mozdzanowska, G. Palladino, and W. Gerhard. 1994. Hetero-
subtypic immunity to influenza type A virus in mice. Effector mechanisms
and their longevity. J. Immunol. 152:1653–1661.
37. Lu, X., L. E. Edwards, J. A. Desheva, D. C. Nguyen, A. Rekstin, I. Stephen-
son, K. Szretter, N. J. Cox, L. G. Rudenko, A. Klimov, and J. M. Katz. 2006.
Cross-protective immunity in mice induced by live-attenuated or inactivated
vaccines against highly pathogenic influenza A (H5N1) viruses. Vaccine
24:6588–6593.
38. Lu, X., M. Renshaw, T. M. Tumpey, G. D. Kelly, J. Hu-Primmer, and J. M.
Katz. 2001. Immunity to influenza A H9N2 viruses induced by infection and
vaccination. J. Virol. 75:4896–4901.
39. Lu, X., T. M. Tumpey, T. Morken, S. R. Zaki, N. J. Cox, and J. M. Katz. 1999.
A mouse model for the evaluation of pathogenesis and immunity to influ-
enza A (H5N1) viruses isolated from humans. J. Virol. 73:5903–5911.
40. Maher, J. A., and J. DeStefano. 2004. The ferret: an animal model to study
influenza virus. Lab. Anim. 33:50–53.
41. Mahmood, K., R. A. Bright, N. Mytle, D. M. Carter, C. J. Crevar, J. E.
Achenbach, P. M. Heaton, T. M. Tumpey, and T. M. Ross. 2008. H5N1 VLP
vaccine induced protection in ferrets against lethal challenge with highly
pathogenic H5N1 influenza viruses. Vaccine 26:5393–5399.
42. Maines, T. R., X. H. Lu, S. M. Erb, L. Edwards, J. Guarner, P. W. Greer,
D. C. Nguyen, K. J. Szretter, L. M. Chen, P. Thawatsupha, M. Chittagan-
pitch, S. Waicharoen, D. T. Nguyen, T. Nguyen, H. H. Nguyen, J. H. Kim,
L. T. Hoang, C. Kang, L. S. Phuong, W. Lim, S. Zaki, R. O. Donis, N. J. Cox,
J. M. Katz, and T. M. Tumpey. 2005. Avian influenza (H5N1) viruses iso-
VOL. 83, 2009 HETEROTYPIC PROTECTION BY MUCOSAL VLP VACCINATION 5733
lated from humans in Asia in 2004 exhibit increased virulence in mammals.
J. Virol. 79:11788–11800.
43. Mazanec, M. B., C. L. Coudret, and D. R. Fletcher. 1995. Intracellular
neutralization of influenza virus by immunoglobulin A anti-hemagglutinin
monoclonal antibodies. J. Virol. 69:1339–1343.
44. Mazanec, M. B., C. S. Kaetzel, M. E. Lamm, D. Fletcher, and J. G. Nedrud.
1992. Intracellular neutralization of virus by immunoglobulin A antibodies.
Proc. Natl. Acad. Sci. USA 89:6901–6905.
45. Nguyen, H. H., F. W. van Ginkel, H. L. Vu, J. R. McGhee, and J. Mestecky.
2001. Heterosubtypic immunity to influenza A virus infection requires B cells
but not CD8
cytotoxic T lymphocytes. J. Infect. Dis. 183:368–376.
46. Nicholson, K. G., A. E. Colegate, A. Podda, I. Stephenson, J. Wood, E. Ypma,
and M. C. Zambon. 2001. Safety and antigenicity of non-adjuvanted and
MF59-adjuvanted influenza A/Duck/Singapore/97 (H5N3) vaccine: a ran-
domised trial of two potential vaccines against H5N1 influenza. Lancet
357:1937–1943.
47. Nolan, T. M., P. C. Richmond, M. V. Skeljo, G. Pearce, G. Hartel, N. T.
Formica, K. Hoschler, J. Bennet, D. Ryan, K. Papanaoum, R. L. Basser, and
M. C. Zambon. 2008. Phase I and II randomised trials of the safety and
immunogenicity of a prototype adjuvanted inactivated split-virus influenza A
(H5N1) vaccine in healthy adults. Vaccine 26:4160–4167.
48. Okuno, Y., Y. Isegawa, F. Sasao, and S. Ueda. 1993. A common neutralizing
epitope conserved between the hemagglutinins of influenza A virus H1 and
H2 strains. J. Virol. 67:2552–2558.
49. Pappas, C., P. V. Aguilar, C. F. Basler, A. Solorzano, H. Zeng, L. A. Perrone,
P. Palese, A. Garcia-Sastre, J. M. Katz, and T. M. Tumpey. 2008. Single gene
reassortants identify a critical role for PB1, HA, and NA in the high virulence
of the 1918 pandemic influenza virus. Proc. Natl. Acad. Sci. USA 105:3064–
3069.
50. Perrone, L. A., J. K. Plowden, A. Garcia-Sastre, J. M. Katz, and T. M.
Tumpey. 2008. H5N1 and 1918 pandemic influenza virus infection results in
early and excessive infiltration of macrophages and neutrophils in the lungs
of mice. PLoS Pathog. 4:e1000115.
51. Prel, A., G. Le Gall-Recule, M. Cherbonnel, B. Grasland, M. Amelot, and V.
Jestin. 2007. Assessment of the protection afforded by triple baculovirus
recombinant coexpressing H5, N3, M1 proteins against a homologous H5N3
low-pathogenicity avian influenza virus challenge in Muscovy ducks. Avian
Dis. 51:484–489.
52. Pushko, P., T. M. Tumpey, F. Bu, J. Knell, R. Robinson, and G. Smith. 2005.
Influenza virus-like particles comprised of the HA, NA, and M1 proteins of
H9N2 influenza virus induce protective immune responses in BALB/c mice.
Vaccine 23:5751–5759.
53. Pushko, P., T. M. Tumpey, N. Van Hoeven, J. A. Belser, R. Robinson, M.
Nathan, G. Smith, D. C. Wright, and R. A. Bright. 2007. Evaluation of
influenza virus-like particles and Novasome adjuvant as candidate vaccine
for avian influenza. Vaccine 25:4283–4290.
54. Quan, F. S., R. W. Compans, H. H. Nguyen, and S. M. Kang. 2008. Induction
of heterosubtypic immunity to influenza virus by intranasal immunization.
J. Virol. 82:1350–1359.
55. Quan, F. S., D. Steinhauer, C. Huang, T. M. Ross, R. W. Compans, and S. M.
Kang. 2008. A bivalent influenza VLP vaccine confers complete inhibition of
virus replication in lungs. Vaccine 26:3352–3361.
56. Raulet, D. H. 1994. MHC class I-deficient mice. Adv. Immunol. 55:381–421.
57. Reed, L. J., and H. Muench. 1938. A simple method of estimating fifty
percent endpoints. Am. J. Hyg. 27:493–497.
58. Richmond, J. Y., and R. W. McKinney (ed.). 2008. Biosafety in microbio-
logical and biomedical laboratories, 5th ed., p. 28–58. U.S. Department of
Health and Human Services, Centers for Disease Control and Prevention,
Atlanta, GA.
59. Rowe, T., R. A. Abernathy, J. Hu-Primmer, W. W. Thompson, X. Lu, W. Lim,
K. Fukuda, N. J. Cox, and J. M. Katz. 1999. Detection of antibody to avian
influenza A (H5N1) virus in human serum by using a combination of sero-
logic assays. J. Clin. Microbiol. 37:937–943.
60. Rudolf, M. P., S. C. Fausch, D. M. Da Silva, and W. M. Kast. 2001. Human
dendritic cells are activated by chimeric human papillomavirus type-16 virus-
like particles and induce epitope-specific human T cell responses in vitro.
J. Immunol. 166:5917–5924.
61. Sandbulte, M. R., G. S. Jimenez, A. C. Boon, L. R. Smith, J. J. Treanor, and
R. J. Webby. 2007. Cross-reactive neuraminidase antibodies afford partial
protection against H5N1 in mice and are present in unexposed humans.
PLoS Med. 4:e59.
62. See, H., and P. Wark. 2008. Innate immune response to viral infection of the
lungs. Paediatr. Respir. Rev. 9:243–250.
63. Smirnov, Y. A., A. S. Lipatov, A. K. Gitelman, E. C. Claas, and A. D.
Osterhaus. 2000. Prevention and treatment of bronchopneumonia in mice
caused by mouse-adapted variant of avian H5N2 influenza A virus using
monoclonal antibody against conserved epitope in the HA stem region.
Arch. Virol. 145:1733–1741.
64. Smirnov, Y. A., A. S. Lipatov, A. K. Gitelman, Y. Okuno, R. Van Beek, A. D.
Osterhaus, and E. C. Claas. 1999. An epitope shared by the hemagglutinins
of H1, H2, H5, and H6 subtypes of influenza A virus. Acta Virol. 43:237–244.
65. Song, J. H., H. H. Nguyen, N. Cuburu, T. Horimoto, S. Y. Ko, S. H. Park, C.
Czerkinsky, and M. N. Kweon. 2008. Sublingual vaccination with influenza
virus protects mice against lethal viral infection. Proc. Natl. Acad. Sci. USA
105:1644–1649.
66. Stevens, J., O. Blixt, T. M. Tumpey, J. K. Taubenberger, J. C. Paulson, and
I. A. Wilson. 2006. Structure and receptor specificity of the hemagglutinin
from an H5N1 influenza virus. Science 312:404–410.
67. Sui, J., W. C. Hwang, S. Perez, G. Wei, D. Aird, L. M. Chen, E. Santelli, B.
Stec, G. Cadwell, M. Ali, H. Wan, A. Murakami, A. Yammanuru, T. Han,
N. J. Cox, L. A. Bankston, R. O. Donis, R. C. Liddington, and W. A. Marasco.
2009. Structural and functional bases for broad-spectrum neutralization of
avian and human influenza A viruses. Nat. Struct. Mol. Biol. 16:265–273.
68. Sun, J., R. Madan, C. L. Karp, and T. J. Braciale. 2009. Effector T cells
control lung inflammation during acute influenza virus infection by produc-
ing IL-10. Nat. Med. 15:277–284.
69. Swenson, D. L., K. L. Warfield, D. L. Negley, A. Schmaljohn, M. J. Aman,
and S. Bavari. 2005. Virus-like particles exhibit potential as a pan-filovirus
vaccine for both Ebola and Marburg viral infections. Vaccine 23:3033–3042.
70. Takada, A., S. Matsushita, A. Ninomiya, Y. Kawaoka, and H. Kida. 2003.
Intranasal immunization with formalin-inactivated virus vaccine induces a
broad spectrum of heterosubtypic immunity against influenza A virus infec-
tion in mice. Vaccine 21:3212–3218.
71. Tompkins, S. M., Z. S. Zhao, C. Y. Lo, J. A. Misplon, T. Liu, Z. Ye, R. J.
Hogan, Z. Wu, K. A. Benton, T. M. Tumpey, and S. L. Epstein. 2007. Matrix
protein 2 vaccination and protection against influenza viruses, including
subtype H5N1. Emerg. Infect. Dis. 13:426–435.
72. Tumpey, T. M., C. F. Basler, P. V. Aguilar, H. Zeng, A. Solorzano, D. E.
Swayne, N. J. Cox, J. M. Katz, J. K. Taubenberger, P. Palese, and A.
Garcia-Sastre. 2005. Characterization of the reconstructed 1918 Spanish
influenza pandemic virus. Science 310:77–80.
73. Tumpey, T. M., A. Garcia-Sastre, J. K. Taubenberger, P. Palese, D. E.
Swayne, M. J. Pantin-Jackwood, S. Schultz-Cherry, A. Solorzano, N. Van
Rooijen, J. M. Katz, and C. F. Basler. 2005. Pathogenicity of influenza
viruses with genes from the 1918 pandemic virus: functional roles of alveolar
macrophages and neutrophils in limiting virus replication and mortality in
mice. J. Virol. 79:14933–14944.
74. Tumpey, T. M., T. R. Maines, N. Van Hoeven, L. Glaser, A. Solorzano, C.
Pappas, N. J. Cox, D. E. Swayne, P. Palese, J. M. Katz, and A. Garcia-Sastre.
2007. A two-amino acid change in the hemagglutinin of the 1918 influenza
virus abolishes transmission. Science 315:655–659.
75. Tumpey, T. M., M. Renshaw, J. D. Clements, and J. M. Katz. 2001. Mucosal
delivery of inactivated influenza vaccine induces B-cell-dependent hetero-
subtypic cross-protection against lethal influenza A H5N1 virus infection.
J. Virol. 75:5141–5150.
76. Ulrich, R., D. Koletzki, S. Lachmann, A. Lundkvist, A. Zankl, A. Kazaks, A.
Kurth, H. R. Gelderblom, G. Borisova, H. Meisel, and D. H. Kruger. 1999.
New chimaeric hepatitis B virus core particles carrying hantavirus (serotype
Puumala) epitopes: immunogenicity and protection against virus challenge.
J. Biotechnol. 73:141–153.
77. Wang, B. Z., W. Liu, S. M. Kang, M. Alam, C. Huang, L. Ye, Y. Sun, Y. Li,
D. L. Kothe, P. Pushko, T. Dokland, B. F. Haynes, G. Smith, B. H. Hahn, and
R. W. Compans. 2007. Incorporation of high levels of chimeric human
immunodeficiency virus envelope glycoproteins into virus-like particles.
J. Virol. 81:10869–10878.
78. Winternitz, M. C., I. M. Wason, and F. P. McNamara. 1920. The pathology
of influenza. Yale University Press, New Haven, CT.
79. Wolbach, S. B. 1919. Comments on the pathology and bacteriology of fatal
influenza cases, as observed at Camp Devins, Mass. Bull. Johns Hopkins
Hosp. 338:104–109.
80. Wood, J. M., J. Mumford, G. C. Schild, R. G. Webster, and K. G. Nicholson.
1986. Single-radial-immunodiffusion potency tests of inactivated influenza
vaccines for use in man and animals. Dev. Biol. Stand. 64:169–177.
81. Yao, Q., Z. Bu, A. Vzorov, C. Yang, and R. W. Compans. 2003. Virus-like
particle and DNA-based candidate AIDS vaccines. Vaccine 21:638–643.
82. Zitzow, L. A., T. Rowe, T. Morken, W. J. Shieh, S. Zaki, and J. M. Katz. 2002.
Pathogenesis of avian influenza A (H5N1) viruses in ferrets. J. Virol. 76:
4420–4429.
5734 PERRONE ET AL. J. VIROL.
... In a study to measure VLP vaccine efficacy against a known influenza pandemic virus, recombinant VLPs were generated from structural proteins of the 1918 H1N1 virus (Perrone et al 2009). Mucosal versus traditional parenteral administration of H1N1 VLPs were compared for the ability to protect against the reconstructed 1918 virus and a highly pathogenic avian H5N1 virus isolated from a fatal human case. ...
Technical Report
Full-text available
This report reviews the current state-of-art of antiviral approaches including vaccines, pharmaceuticals and innovative technologies for delivery of therapeutics. The introduction starts with a practical classification of viral diseases according to their commercial importance. Various antiviral approaches are described including pharmaceuticals and molecular biological therapies such as gene therapy and RNA interference (RNAi) as well as vaccines for virus infections. Expert opinion is given about the current problems and needs in antiviral therapy. SWOT (strengths, weaknesses, opportunities and threats) analysis of antiviral approaches is presented against the background of concept of an ideal antiviral agent. A novel feature of this report is the use of nanotechnology in virology and its potential for antiviral therapeutics. Interaction of nanoparticles with viruses are described. NanoViricides are polymeric micelles, which act as nanomedicines to destroy viruses. Various methods for local as well as systemic delivery of antiviral agents and vaccines are described. Nanobiotechnology plays an important role in improving delivery of antivirals. Advantages and limitations of delivery of gene-based, antisense and RNAi antiviral therapeutics are discussed. Anti-influenza measures applicable to human as well as avian forms are described including the recent epidemic of swine flu. Resistance can develop against neuraminidase inhibitors although it is less than that with adamantanes. Considering these problems, there is need for a more effective agent. Investigations into alternative anti-influenza target will probably expand in the coming years. These include the development of mechanisms to inhibit fusion between the virus envelope and the cell membrane. After a discussion of current therapies of AIDS/HIV and their limitations, new strategies in development of antiviral agents are described. Drug resistance and toxicities are emerging as major treatment challenges. Based on a review of technologies and drugs in development, it can be stated that there are good prospects are of finding a cure for HIV/AIDS in the next decade. Hepatitis viruses are described with focus on hepatitis C virus (HCV) and hepatitis B virus (HBV). Despite the presence of numerous drug candidates in the anti-HCV pipeline, and the commitment of major R&D resources by many pharmaceutical companies, it might still take several years for any new anti-HCV drugs to reach the market. Although many companies are focusing their efforts on developing viral inhibitors, cellular targets in the host are beginning to emerge as attractive possibilities because they might enable the development of broad-spectrum antiviral drugs with less chance for developing viral resistance. Various commercially important viruses include herpes simplex (HSV) and human papilloma virus (HPV). There a number of treatments but HSV is not destroyed completely and remains dormant and activates from time to time to cause various clinical manifestations. There is discussion about the role of HPV in cervical cancer and vaccines available now seem to be adequate in preventing HSV-induced cervical cancer. There is no effective vaccine for respiratory syncytial virus (RSV) although monoclonal antibody (MAb) treatment is useful for prophylaxis and reducing the clinical manifestations. There is a need for an agent to eliminate this virus. Various viruses that either occur in epidemics or in tropics and some naturally emerging infectious diseases are described, e.g. viral hemorrhagic fevers such as dengue and West Nile virus infection. These are a constant threat and impossible to anticipate. Some of these lack antiviral agents or vaccines for prevention. Although these include some of the most serious viral disorders, the development of antiviral agents for these is not commercially attractive. Current research and approaches to these virus infections, particularly the current pandemic of COVID-19, are discussed. There are over 110 drugs and vaccine candidates in development of which 3 have been approved by the FDA and other health authorities around the world. Vaccination is being carried out in several countries Markets for antivirals are considered according to viruses and diseases caused by them and also according to management approaches: antiviral drugs, vaccines, MAbs and innovative approaches that include immunological and use of other technologies such as gene therapy, antisense, RNAi and nanobiotechnology. Antiviral markets are estimated starting with 2020 with projections up to the year 2030. Profiles of 197 companies that are involved in developing various technologies and products are profiled and with 182 collaborations. These include major pharmaceutical companies (12), Biopharmaceutical companies with antiviral products (87), Antiviral drug companies (26) as well as viral vaccine companies (71). The report is supplemented with 58 tables, 17 figures and 550 references from the literature.
... The small size of the particles (less than 100 nm) enables their permeation across the mucosa barrier increasing the interaction with the NALT cells [36]. IN immunization with VLPs containing the structural proteins of the H1N1 (1918) virus, found to protect mice against replication-competent homologous virus, as well as against the lethal challenge, inducing high levels of cross-reactive IgG and IgA antibodies [84]. This aspect was verified in a preclinical study where nasal administration of this system aimed to ensure immunization against HIV-1, after the incorporation of glycoproteinexpressing DNA sequences into the VLPs [85]. ...
Article
The importance of vaccination has been proven particularly significant the last three years, as it is revealed to be the most efficient weapon for the prevention of several infections including SARS-COV-2. Parenteral vaccination is the most applicable method of immunization, for the prevention of systematic and respiratory infections, or central nervous system disorders, involving T and B cells to a whole-body immune response. However, the mucosal vaccines, such as nasal vaccines, can additionally activate the immune cells localized on the mucosal tissue of the upper and lower respiratory tract. This dual stimulation of the immune system, along with their needle-free administration favors the development of novel nasal vaccines to produce long-lasting immunity. In recent years, the nanoparticulate systems have been extensively involved in the formulation of nasal vaccines as polymeric, polysaccharide and lipid ones, as well as in the form of proteosomes, lipopeptides and virosomes. Advanced delivery nanosystems have been designed and evaluated as carriers or adjuvants for nasal vaccination. To this end, several nanoparticulate vaccines are undergone clinical trials as promising candidates for nasal immunization, while nasal vaccines against influenza type A and B and hepatitis B have been approved by health authorities. This comprehensive literature review aims to summarize the critical aspects of these formulations and highlight their potential for the future establishment of nasal vaccination. Both preclinical (in vitro and in vivo) and clinical studies are incorporated, summarized, and critically discussed, as well as the limitations of nasal immunization.
... The well-studied poliovirus case with the known difference between inactivated polio vaccine and oral polio vaccine exemplifies the importance of such mimicry for providing sterilizing immunity (77,78). Many more studies also demonstrate that the mucosal route of vaccination provides such a beneficial protection against respiratory and digestive-tract virus and bacterial infections, including influenza and rotavirus and even SARS-CoV-2 and its emerging variants, e.g (5,27,67,71,(79)(80)(81)(82)(83)(84)(85)(86). However, formulating an immunogenic, broad, and safe "subunit" or "inactivated" mucosal viral vaccine, capable of eliciting longterm efficient and balanced mucosal-plus-systemic protections, remains challenging (87). ...
Article
Full-text available
Intramuscularly administered vaccines stimulate robust serum neutralizing antibodies, yet they are often less competent in eliciting sustainable “sterilizing immunity” at the mucosal level. Our study uncovers a strong temporary neutralizing mucosal component of immunity, emanating from intramuscular administration of an mRNA vaccine. We show that saliva of BNT162b2 vaccinees contains temporary IgA targeting the receptor-binding domain (RBD) of severe acute respiratory syndrome coronavirus-2 spike protein and demonstrate that these IgAs mediate neutralization. RBD-targeting IgAs were found to associate with the secretory component, indicating their bona fide transcytotic origin and their polymeric multivalent nature. The mechanistic understanding of the high neutralizing activity provided by mucosal IgA, acting at the first line of defense, will advance vaccination design and surveillance principles and may point to novel treatment approaches and new routes of vaccine administration and boosting.
Article
Aims: To develop an effective universal vaccine against antigenically different influenza viruses. Materials & methods: We generated influenza virus-like particles (VLPs) expressing the H1 and H3 antigens with or without M2e5x. VLP-induced immune responses and crossprotection against H1N1, H3N2 or H5N1 viruses were assessed to evaluate their protective efficacy. Results: H1H3M2e5x immunization elicited higher crossreactive IgG antibodies than H1H3 VLPs. Upon challenge, both VLPs enhanced lung IgG, IgA and germinal center B-cell responses compared with control. While these VLPs conferred protection, H1H3M2e5x showed greater lung viral load reduction than H1H3 VLPs with minimal body weight loss. Conclusion: Utilizing VLPs containing dual-hemagglutinin, along with M2e5x, can be a vaccination strategy for inducing crossprotection against influenza A viruses.
Article
Full-text available
Inducing humoral and cytotoxic mucosal immunity at the sites of pathogen entry has the potential to prevent the infection from getting established. This is different from systemic vaccination, which protects against the development of systemic symptoms. The field of mucosal vaccination has seen fewer technological advances compared to nucleic acid and subunit vaccine advances for injectable vaccine platforms. The advent of the next-generation adenoviral vectors has given a boost to mucosal vaccine research. Basic research into the mechanisms regulating innate and adaptive mucosal immunity and the discovery of effective and safe mucosal vaccine adjuvants will continue to improve mucosal vaccine design. The results from clinical trials of inhaled COVID-19 vaccines demonstrate their ability to induce the proliferation of cytotoxic T cells and the production of secreted IgA and IgG antibodies locally, unlike intramuscular vaccinations. However, these mucosal vaccines induce systemic immune responses at par with systemic vaccinations. This review summarizes the function of the respiratory mucosa-associated lymphoid tissue and the advantages that the adenoviral vectors provide as inhaled vaccine platforms.
Article
Vaccines that trigger mucosal immune responses at the entry portals of pathogens are highly desired. Here, we showed that antigen-decorated nanoparticle generated through CRISPR engineering of T4 bacteriophage can serve as a universal platform for the rapid development of mucosal vaccines. Insertion of Flu viral M2e into phage T4 genome through fusion to Soc (Small Outer Capsid protein) generated a recombinant phage, and the Soc-M2e proteins self-assembled onto phage capsids to form 3M2e-T4 nanoparticles during propagation of T4 in E. coli. Intranasal administration of 3M2e-T4 nanoparticles maintains antigen persistence in the lungs, resulting in increased uptake and presentation by antigen-presenting cells. M2e-specific secretory IgA, effector (TEM), central (TCM), and tissue-resident memory CD4+ T cells (TRM) were efficiently induced in the local mucosal sites, which mediated protections against divergent influenza viruses. Our studies demonstrated the mechanisms of immune protection following 3M2e-T4 nanoparticles vaccination and provide a versatile T4 platform that can be customized to rapidly develop mucosal vaccines against future emerging epidemics.
Article
Bacterial respiratory infections either acute or chronic are major threats for human health. Direct mucosal administration, through the airways, of therapeutic antibodies (Ab) offers a tremendous opportunity to benefit to patients with respiratory infections. The mode of action of anti-infective antibodies relies on pathogen neutralization and Fc-mediated recruitment of immune effectors to facilitate their elimination. Using a mouse model of acute pneumonia induced by P. aeruginosa, we depicted the immunomodulatory mode of action of a neutralizing anti-bacterial Ab. Beyond the rapid and efficient containment of the primary infection, the Ab delivered through the airways harnessed genuine innate and adaptive immune responses to provide long-term protection, preventing from a secondary bacterial infection. In vitro antigen-presenting cells stimulation assay, as well as in vivo bacterial challenges and serum transfer experiments indicate an essential contribution of immunes complexes with the Ab and pathogen in the induction of the sustained and protective antibacterial humoral response. Interestingly, the long-lasting response protected partially against secondary infections with heterologous P. aeruginosa strains. Overall, our findings suggest that Ab, delivered mucosally, promotes bacteria neutralization and provides protection against secondary infection. This opens novel perspectives for the development of anti-infective Ab delivered to the lung mucosa, to treat respiratory infections.
Article
Full-text available
In this study, recombinant virus-like particles (VLPs) were evaluated as a candidate vaccine against emerging influenza viruses with pandemic potential. The VLPs are composed of the hemagglutinin (HA), neuraminidase (NA), and matrix 1 (M1) proteins of the H5N1 A/Indonesia/05/2005 (clade 2.1; [Indo/05]) virus, which were expressed using baculovirus in Spodoptera frugiperda (Sf9) cells. Ferrets received either 2 injections of the VLP vaccine at escalating doses (based on HA content), recombinant HA, or were mock vaccinated. Vaccinated ferrets were then challenged with either H5N1 Indo/05 or H5N1 A/Viet Nam 1203/2004 (VN/04) wild-type viruses. All ferrets that received the VLP vaccine survived regardless of the VLP dose or challenge strain, whereas seven of eight mock vaccinated ferrets died. The VLP vaccine induced HAI antibodies against the homologous H5N1 clade 2.1 strain, as well as heterologous strains from H5N1 clades 1, 2.2, and 2.3. The magnitude of the HAI titers correlated with VLP dose. Neutralizing antibody responses against the Indo/05 and VN/04 strains showed a similar pattern. Affinity of the anti-HA antibodies raised by the H5N1 Indo/05 VLPs had a higher association rate to the homologous clade 2.1 HA than to the clade 1 (VN/04) HA; however, once bound, antibodies had similar slow disassociation rates. These results provide support for continued development of the H5N1 VLPs as a candidate vaccine against pandemic influenza. Exploration of immunologic correlates of protection for H5N1 vaccines beyond HAI and neutralizing antibody responses is warranted.
Article
During 1997 in Hong Kong, 18 human cases of respiratory illness, including 6 fatalities, were caused by highly pathogenic avian influenza A (H5N1) viruses. Since H5 viruses had previously been isolated only from avian species, the outbreak raised questions about the ability of these viruses to cause severe disease and death in humans. To better understand the pathogenesis and immunity to these viruses, we have used the BALB/c mouse model. Four H5N1 viruses replicated equally well in the lungs of mice without prior adaptation but differed in lethality for mice. H5N1 viruses that were highly lethal for mice were detected in multiple organs, including the brain. This is the first demonstration of an influenza A virus that replicates systemically in a mammalian species and is neurotropic without prior adaptation. The mouse model was also used to evaluate a strategy of vaccination against the highly pathogenic avian H5N1 viruses, using an inactivated vaccine prepared from nonpathogenic A/Duck/Singapore-Q/F119-3/97 (H5N3) virus that was antigenically related to the human H5N1 viruses. Mice administered vaccine intramuscularly, with or without alum, were completely protected from lethal challenge with H5N1 virus. Protection from infection was also observed in 70% of animals administered vaccine alone and 100% of mice administered vaccine with alum. The protective effect of vaccination correlated with the level of virus-specific serum antibody. These results suggests a strategy of vaccine preparedness for rapid intervention in future influenza pandemics that uses antigenically related nonpathogenic viruses as vaccine candidates.
Article
Immunity that cross-reacts between influenza type A viruses of distinct subtypes is called hetero(sub)typic (Het-I). We have studied Het-I by challenging PR8-immune mice with the heterosubtypic virus X31. Het-I did not prevent infection by X31 but, at its height, strongly aided in recovery. The nature of the effector mechanisms involved was investigated by simultaneous challenge with X31 and an immunologically unrelated influenza type B virus and by depleting individual lymphocyte subsets in PR8-immune mice before challenge. The study showed the following: 1) The effector mechanisms were intimately associated with immune recognition events. 2) In the nose, depletion of CD8+ or CD4+ T cells led to partial reduction of Het-I, and simultaneous depletion of both T cell subsets abrogated Het-I almost completely. This T cell-mediated immunity was short lived and had disappeared 4 to 5 mo after induction. 3) In trachea and lung, depletion of CD8+ T cells led to a partial reduction of Het-I, whereas depletion of CD4+ T cells was without significant effect. The CD8-mediated component appeared short lived, whereas the residual immunity (in CD4/8-depleted mice) was long lived and persisted past 7 mos after induction. 4) Depletion of NK cells did not significantly reduce the strength of Het-I in either nose or lung. In conclusion, the study shows that Het-I in this system is mediated by a complex combination of immune mechanisms that differ, in part, between upper and lower respiratory tract.
Article
It has been known that influenza A virus infection induces a cross-protective immunity against infection by viruses with different subtypes of viral envelope proteins, hemagglutinin (HA) and neuraminidase (NA). This heterosubtypic immunity is generally mediated by cytotoxic T lymphocytes (CTL) reactive to specific epitopes in the viral internal proteins, such as nucleoprotein and matrix protein. By contrast, immunization with inactivated virus antigens has been thought to be unable to generate heterosubtypic immunity, since inactivated antigens do not usually induce CTL responses. However, we show that intranasal immunization with formalin-inactivated intact virus, but not ether-split vaccines, induced a broad spectrum of heterosubtypic protective immunity in mice. The protection may be mediated by the mucosal immune response, most likely secretory IgA antibodies to the viral proteins. This approach may overcome limitations in the efficacy of inactivated influenza vaccines and confer potent immunity to humans against viruses with new pandemic potential.
Article
... Survival rates of the mice are also shown in Table 1. Mice immunized with the Hok67 (H5N4) vaccine were all protected from lethal infection without any disease signs. Most of the mice immunized intranasally with influenza A virus vaccines , regardless of the HA and NA ...
Article
Traditionally, immunoglobulin A (IgA) was thought to neutralize virus by forming complexes with viral attachment proteins, blocking attachment of virions to host epithelial cells. Recently we have proposed an intracellular action for dimeric IgA,,which is actively transported through epithelial cells by the polymeric immunoglobulin receptor (pIgR), in that it may be able to bind to newly synthesized viral proteins, within the cell, preventing viral assembly. To this effect, we have previously demonstrated that IgA monoclonal antibodies against Sendai virus, a parainfluenza virus, colocalize with the viral hemagglutinin-neuraminidase protein within infected epithelial cells and reduce intracellular viral titers. Here we determine whether IgA can interact with influenza virus hemagglutinin (HA) protein within epithelial cells. Polarized monolayers of Madin-Darby canine kidney epithelial cells expressing the pIgR were infected on their apical surfaces with influenza virus A/Puerto Rico/8-Mount Sinai. Polymeric IgA anti-HA, but not IgG anti-HA, delivered to the basolateral surface colocalized with HA protein within the cell by immunofluorescence. Compared with those of controls, viral titers were reduced in the supernatants and cell lysates from monolayers treated with anti-HA IgA but not,vith anti-HA IgG. Furthermore, the addition of anti-IgA antibodies to supernatants did not interfere with the neutralizing activity of IgA placed in the basal chamber, indicating that IgA was acting within the cell and not in the extracellular medium to interrupt viral replication. Thus, these studies provide additional support for the concept that IgA can inhibit replication of microbial pathogens intracellularly.
Article
In this report, based on approximately 200 necropsies, no attempt will be made to distinguish between changes due to the unknown virus and those from mixed or secondary infection; only the most outstanding features are discussed.Perhaps the most lasting impression from long association with lobar pneumonia in postmortem examination is that when it alone is responsible for death, with very few exceptions,1 a considerable part of the total pulmonary parenchyma is consolidated, undistensible and heavier than normal; even when limited to one upper lobe, that lobe is as a rule huge, and the lung weight as a consequence frequently doubled.Therefore, the first feature of the lungs in influenza to attract attention was the relatively small amount of lung tissue solid with grossly demonstrable pneumonia. Even when measured, the total of such regions is so small that it is difficult to ascribe death to the pneumonia with an