ArticlePDF Available

H5N1 Vaccine-Specific B Cell Responses in Ferrets Primed with Live Attenuated Seasonal Influenza Vaccines

PLOS
PLOS ONE
Authors:

Abstract and Figures

Live attenuated influenza H5N1 vaccines have been produced and evaluated in mice and ferrets that were never exposed to influenza A virus infection (Suguitan et al., Plos Medicine, e360:1541, 2006). However, the preexisting influenza heterosubtypic immunity on live attenuated H5N1 vaccine induced immune response has not been evaluated. Primary and recall B cell responses to live attenuated H5N1 vaccine viruses were examined using a sensitive antigen-specific B cell ELISpot assay to investigate the effect of preexisting heterosubtypic influenza immunity on the development of H5N1-specific B cell immune responses in ferrets. Live attenuated H5N1 A/Hong Kong/213/03 and A/Vietnam/1203/04 vaccine viruses induced measurable H5-specific IgM and IgG secreting B cells after intranasal vaccination. However, H5-specific IgG secreting cells were detected significantly earlier and at a greater frequency after H5N1 inoculation in ferrets previously primed with trivalent live attenuated influenza (H1N1, H3N2 and B) vaccine. Priming studies further revealed that the more rapid B cell responses to H5 resulted from cross-reactive B cell immunity to the hemagglutinin H1 protein. Moreover, vaccination with the H1N1 vaccine virus was able to induce protective responses capable of limiting replication of the H5N1 vaccine virus to a level comparable with prior vaccination with the H5N1 vaccine virus without affecting H5N1 vaccine virus induced antibody response. The findings indicate that previous vaccination with seasonal influenza vaccine may accelerate onset of immunity by an H5N1 ca vaccine and the heterosubtypic immunity may be beneficial for pandemic preparedness.
Content may be subject to copyright.
H5N1 Vaccine-Specific B Cell Responses in Ferrets Primed
with Live Attenuated Seasonal Influenza Vaccines
Xing Cheng
1.
, Michael Eisenbraun
1.
,QiXu
1
, Helen Zhou
1
, Deepali Kulkarni
1
, Kanta Subbarao
2
, George
Kemble
1
, Hong Jin
1
*
1MedImmune, Mountain View, California, United States of America, 2Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases (NIAID),
National Insitutes of Health, Bethesda, Maryland, United States of America
Abstract
Background:
Live attenuated influenza H5N1 vaccines have been produced and evaluated in mice and ferrets that were
never exposed to influenza A virus infection (Suguitan et al., Plos Medicine, e360:1541, 2006). However, the preexisting
influenza heterosubtypic immunity on live attenuated H5N1 vaccine induced immune response has not been evaluated.
Methodology and Principal Findings:
Primary and recall B cell responses to live attenuated H5N1 vaccine viruses were
examined using a sensitive antigen-specific B cell ELISpot assay to investigate the effect of preexisting heterosubtypic
influenza immunity on the development of H5N1-specific B cell immune responses in ferrets. Live attenuated H5N1 A/Hong
Kong/213/03 and A/Vietnam/1203/04 vaccine viruses induced measurable H5-specific IgM and IgG secreting B cells after
intranasal vaccination. However, H5-specific IgG secreting cells were detected significantly earlier and at a greater frequency
after H5N1 inoculation in ferrets previously primed with trivalent live attenuated influenza (H1N1, H3N2 and B) vaccine.
Priming studies further revealed that the more rapid B cell responses to H5 resulted from cross-reactive B cell immunity to
the hemagglutinin H1 protein. Moreover, vaccination with the H1N1 vaccine virus was able to induce protective responses
capable of limiting replication of the H5N1 vaccine virus to a level comparable with prior vaccination with the H5N1 vaccine
virus without affecting H5N1 vaccine virus induced antibody response.
Conclusion:
The findings indicate that previous vaccination with seasonal influenza vaccine may accelerate onset of
immunity by an H5N1 ca vaccine and the heterosubtypic immunity may be beneficial for pandemic preparedness.
Citation: Cheng X, Eisenbraun M, Xu Q, Zhou H, Kulkarni D, et al. (2009) H5N1 Vaccine-Specific B Cell Responses in Ferrets Primed with Live Attenuated Seasonal
Influenza Vaccines. PLoS ONE 4(2): e4436. doi:10.1371/journal.pone.0004436
Editor: Linqi Zhang, Comprehensive AIDS Reseach Center, China
Received September 11, 2008; Accepted December 17, 2008; Published February 11, 2009
This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration which stipulates that, once placed in the public
domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose.
Funding: The funding was provided by MedImmune. This research was supported in part by the Intramural Research Program of NIAID, NIH. The funders had no
role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: jinh@medimmune.com
.These authors contributed equally to this work.
Introduction
Influenza pandemics can occur when new influenza subtypes
capable of both infecting and spreading easily among humans
emerge with a new hemagglutinin (HA) subtype (antigenic shift) to
which there is little or no population immunity. During the last
century, three novel influenza A hemagglutinin subtypes (H1, H2
and H3) have appeared; an H1N1 strain caused the catastrophic
‘‘Spanish flu’’ pandemic in 1918 [1] followed by milder pandemics
in 1957 and 1968 caused by H2N2 and H3N2 strains, respectively.
Importantly, the origin of the pandemic H2N2 and H3N2 viruses
has since been attributed to genetic reassortment events where
circulating human influenza viruses acquired novel HA subtypes
from avian influenza viruses [2,3]. Alarmingly, in the past decade, a
number of avian influenza viruses containing HA subtypes not
typically found in humans have crossed species barrier and infected
humans, raising concerns about a future pandemic. Highly
pathogenic avian H5N1 influenza viruses have infected only a
small number of individuals but are associated with a high mortality
rate and are perceived as a potential major global health threat.
Several strategies have been used to develop vaccines against
H5N1 viruses including inactivated whole virus vaccines, split or
subunit vaccines, live attenuated influenza vaccine (LAIV),
vectored vaccines, and DNA vaccines; many of these candidates
have shown promise in preclinical studies [4]. Seasonal LAIV has
demonstrated several attributes that would be important for an
effective pandemic vaccine including efficacy, an ability to protect
against antigenically drifted strains, an ability to elicit a rapid
immune response in an immunologically naı
¨ve population, and a
highly efficient production system for the vaccine [5,6,7,8]. Several
prototypic pandemic LAIV (pLAIV) 6:2 reassortant viruses
containing the H5N1 HA and NA gene segments have been
produced on the backbone of six internal gene segments from the
cold-adapted (ca) A/Ann Arbor/6/60 vaccine strain [9], the
master donor virus (MDV-A) used to produce influenza A vaccine
strains for the seasonal FluMistHinfluenza vaccines (MedImmune).
These candidate H5N1 vaccine strains, A/HK/491/97 (HK97
ca), A/HK/213/03 (HK03 ca), and A/VN/1203/04 (VN04 ca),
were found to provide complete protection against lethal challenge
with homologous and heterologous wild-type (wt) H5N1 viruses in
PLoS ONE | www.plosone.org 1 February 2009 | Volume 4 | Issue 2 | e4436
mice and offered complete protection against pulmonary replica-
tion of wt H5N1 virus in ferrets [7].
It has been observed that individuals who have recovered from
influenza infections develop broad subtype-specific immunity that
can protect them from subsequent infection by closely related drift
variants of the same subtype [10,11,12]. Although not nearly as
common, Schulman and Kilbourne [13] reported heterosubtypic
immunity in mice, where protection was induced by an influenza
virus belonging to a different subtype. Recently, there have been
several reports describing heterosubtypic immunity against H5N1
infection induced by influenza virus infection or vaccines in mice
[8,14,15,16]. The mechanistic basis of this type of immunity
remains undetermined, however, one study demonstrated a role of
the N1 component of the vaccine [17] and other studies suggest
that structural similarity of the H5 and H1 HA may mediate this
type of protection [18,19]. Kreijtz et al. [20] reported cross-
recognition of avian H5N1 influenza virus by human cytotoxic T-
lymphocyte populations directed against human influenza viruses
and suggested that the preexisting cross-reactive T-cell immunity
in humans may dampen the impact of a next pandemic if it is
caused by an H5N1 virus. The ferret is considered to be a suitable
mammalian host for seasonal influenza vaccine research [21,22]
and for efficacy studies of HPAI H5N1 vaccines [7,23,24].
Although ferrets immunized with a H1N1 ca strain were not
protected from replication of a wild-type H5N1 virus [7], however,
because LAIV has been shown to provide protection from strains
that are antigenically different from the vaccine antigen, we
investigated whether priming with a heterologous seasonal LAIV
vaccine containing different subtypes could influence the immune
response to H5N1 viruses in the ferret model. Such studies will
also help us to understand whether live attenuated H5N1 vaccine
could induce effective immune response in individuals that have
immunity to seasonal influenza viruses.
HAI and microneutralization assays are frequently used to
measure humoral antibody responses, however, these assays may
not be sensitive enough to detect early and local antibody
responses. To assess the presence and magnitude of heterosubtypic
immunity following immunization with LAIV, a sensitive B cell
ELISpot assay was developed that could detect early induction of
immunity at a time when the HAI assay was less sensitive. Using
this assay, we show that local B cell responses induced by the
H5N1 VN04 ca and HK03 ca vaccine viruses can be detected at a
virus-specific and HA-specific level. Previous infection with an
H1N1 virus induced a faster and higher level B cell response to
H5N1 vaccination and could prevent shedding of the H5N1
vaccine virus. The data implies that priming with a non-H5
vaccine may enable a more rapid memory response to an H5
vaccine, however, whether this would be beneficial to the
effectiveness of an H5 vaccine remains to be determined.
Materials and Methods
Viruses
Influenza virus vaccine strains H1N1 A/New Caledonia/20/99
ca (NC99 ca), H3N2 A/Wyoming/03/03 ca (WY03 ca), H3N2 A/
California/7/04 ca (CA04 ca), H5N1 A/Hong Kong/213/03 ca
(HK03 ca), H5N1 A/Vietnam/1203/04 ca (VN04 ca), H2N2 A/
AA/6/60 ca, (AA60 ca or MDV-A) [22] and H1N2 reassortant ca
virus containing the H1 HA from A/New Caledonia/20/99 and
the N2 NA from A/Wyoming/03/03 were generated by reverse
genetics. All viruses were expanded at 33uC for 3 days in the
allantoic cavity of 10-day-old embryonated SPAFAS hen’s eggs
(Norwich, CT). Allantoic fluids collected from infected eggs were
examined by hemagglutination assay using 0.5% turkey (tRBC) or
horse (hRBC) erythrocytes to determine HA titer. Infectious virus
titer was determined by plaque assay (plaque forming unit, PFU)
or 50% tissue culture infectious dose (TCID
50
) using Madin-Darby
canine kidney (MDCK) cells. Viruses were inactivated by
treatment with b-propriolactone (BPL) for use as antigen in
ELISpot and ELISA assays. Trivalent LAIV (FluMistH) was
manufactured by MedImmune and contained 10
7
TCID
50
each of
6:2 reassortant vaccine strains, A/New Caledonia/20/99 ca,A/
California/7/04 ca, and B/Jilin/20/03 ca.
Animal studies
Ferrets between 7 and 10 weeks of age from Triple F Farms
(Sayre, PA) were screened prior to use in experiments for
preexisting antibodies to H1N1, H3N2 and H5N1 influenza
viruses by hemagglutination inhibition (HAI) assay. Sero-negative
ferrets were inoculated intranasally on day 0 with a predetermined
does of a monovalent vaccine virus (10
5
to 10
7
PFU or TCID
50
virus), trivalent LAIV, or medium (mock control). To examine the
B cell response and virus replication after primary infection,
animals were sacrificed 5 or 10 days post-inoculation to collect
paratracheal lymph nodes (TLN) and whole blood for B cell
ELIspot assays; serum was collected to measure serum antibody
level and nasal turbinates were harvested to examine virus
replication in the upper respiratory tract. To examine the effect
of previous influenza virus infection on the induction of H5N1-
specific immune responses, ferrets were given a second intranasal
inoculation of 10
7
or 10
8
PFU of homologous or heterologous
vaccine virus 4–6 weeks after the initial vaccination. Animals in
these latter groups were sacrificed 3–10 days later to collect TLN,
blood, serum, and nasal turbinate samples or were used to collect
serum samples for up to three weeks after the second vaccination.
All animal study protocols were approved by MedImmune’s
Institutional Animal Care and Use Committee and performed in
an AAALAC certified facility.
Measurement of virus titers in animal tissues
Nasal turbinate tissues were homogenized in MEM medium
and centrifuged at 400 g for 10 min. Serial 10-fold dilutions of
supernatants collected from each preparation were inoculated into
three 10- to 11-day old embryonated SPAFAS hen’s eggs. After
incubation at 33uC for 72 hr, allantoic fluid from each egg was
collected for HA assay using 0.5% tRBC. Virus titers in the tissues
are reported as a 50% egg infectious dose (EID
50
) per gram of
tissue processed.
B cell ELISpot assay
AcroWell
TM
96-well PVDF filter plates (Pall Life Sciences, Ann
Arbor, MI) were coated with 50 mL/well PBS containing either
2,000 HA unit/mL of BPL-treated vaccine virus or 10 mg/mL
recombinant HA protein derived from H5N1 A/VN/1203/2004
(rH5), H1N1 A/New Caledonia/20/99 (rH1), or H3N2 A/
Wyoming/03/03 (rH3) that were purified from recombinant
baculovirus infected insect cells (Protein Sciences, Meriden, CT).
After overnight incubation at 4uC, plates were washed 3 times with
PBS and blocked with RPMI-1640 medium containing 10% FBS
for 2 hr at 37uC prior to the addition of cell samples.
Whole blood samples from ferrets were collected in EDTA
tubes and processed using LympholyteH-Mammal (Cedarlane,
Ontario, Canada) to isolate peripheral blood mononuclear cells
(PBMC). PBMC were washed once with RPMI-1640/10% FBS
by centrifugation (300 g for 10 min), counted, and resuspended in
complete medium (RPMI-1640, 10% FBS, 2 mM L-glutamine,
0.5 nM ß-mercaptoethanol and penicillin/streptomycin). TLN
were harvested from each ferret and placed in cold PBS/5% FBS
H5N1 Vaccine
PLoS ONE | www.plosone.org 2 February 2009 | Volume 4 | Issue 2 | e4436
and the cells were released from TLN into the media by gently
rubbing partially minced tissue against a sterile mesh screen with a
glass pestle. The resultant cell suspension was collected, passed
through a cell strainer to remove large debris, and pelleted by
centrifugation (300 g for 10 min). Cell pellets were washed once,
counted, and resuspended in the RPMI-1640 complete medium.
PBMC and TLN cell suspensions were added to triplicate wells
(100 mL/well) at a concentration of 3610
6
/mL for PBMC or 10
5
to 10
6
/mL for TLN samples and incubated at 37uC, 5% CO2 for
5 hr. The plates were washed 5 times with PBS containing 0.05%
Tween-20 (PBS-T) to remove the cells from the plate. To measure
isotype-specific B cell responses, goat anti-ferret IgM (Rockland,
Gilbertville, PA) or goat anti-ferret IgG (Bethyl Laboratories,
Montgomery, TX) diluted 1:1000 in PBS-T/1% BSA and
incubated overnight at 4uC. After 5 washes with PBS-T, HRP-
conjugated rabbit anti-goat Ig (Dako, Carpinteria, CA) diluted
1:2000 in PBS-T/BSA was added to all wells and incubated at
37uC for 1 hr. Plates were washed 3 times with PBS-T and 3 times
with PBS before development with AEC substrate (Vector Labs,
Burlingame, CA) for 10 min at room temperature (RT). Wells
were rinsed extensively with water and allowed to dry completely
before spots in each well were counted using an ImmunoSpot plate
reader (Cellular Technologies, Ltd., Cleveland, OH).
HAI and microneutralization assays
Prior to serologic analysis, ferret sera were treated with
receptor-destroying enzyme (RDE) (Denka Seiken, Tokyo, Japan)
that was reconstituted with 10 mL of 0.9% NaCl per vial. 0.1 mL
serum was mixed with 0.15 mL RDE and incubated at 37uC for
18 hr and adjusted to a final 1:4 dilution by adding 0.15 mL of
0.9% sodium citrate followed by incubation at 56uC for 45 min.
Strain-specific serum HAI titers were determined using 0.5%
tRBC or hRBC and the HAI titers are presented as the reciprocal
value of the highest serum dilution that did not inhibit
hemagglutination. Serum neutralizing antibody titers were
determined by microneutralization assay using MDCK cells.
RDE-treated ferret serum was 2-fold serially diluted, incubated
with 100 TCID
50
virus at 33uC for 1 hr and transferred onto
MDCK cell monolayers in 96-well culture plates (Costar, Corning,
NY). After 6 days’ incubation at 33uC, the cell monolayers were
fixed with 10% formaldehyde, incubated with chicken MDV-A
polyclonal antibody followed by incubation with an HRP-
conjugated rabbit anti-chicken IgG (Thermo, Rockford, IL), and
developed with TMB substrate (Sigma, St. Louis, MO). The
reaction was stopped with an equal volume of 0.1 N HCl and the
absorbance at 450 nm was determined using a SpectraMax plate
reader (Molecular Devices, Sunnyvale, CA). Neutralizing antibody
titers were calculated as the highest serum dilution with a value less
than that calculated by the formula of (average OD of virus-
infected wells - average OD of cell control wells)/2+average OD of
cell control wells.
ELISA analysis of HA-specific antibody responses
96-well EIA plates (Costar, Corning, NY) were coated with
0.025 mg/well of rH1, rH3 or rH5 in PBS overnight at 4uC. Plates
were washed 3 times with PBS-T and blocked with SuperBlock
Blocking Buffer (Pierce, Rockford, IL) for 1 hr at 37uC. RDE-
treated ferret sera were 2-fold serially diluted with PBS-T,
transferred to 96-well plates (50 mL/well), and incubated for
1hrat37uC. Plates were washed with PBS-T and incubated for
30 min at 37uC with 100 mL/well HRP-conjugated goat anti-
ferret IgG (Bethyl Laboratories, Montgomery, TX) diluted
1:10,000 in PBS-T/1% BSA. After washing with PBS-T, plates
were developed with TMB substrate and read as described above
in the microneutralization assay. Antibody titers are expressed as
the highest dilution with an optical density (OD) reading greater
than 2 times the mean OD+standard deviation of similarly diluted
negative control samples.
Results
Detection of H5N1-specific B cell responses after
immunization with pLAIV using a sensitive B cell ELIspot
assay
Pandemic live attenuated influenza vaccines (pLAIV) developed
for H5N1 viruses confer protection against wild-type virus
challenge in mice and ferrets [7], however, the level of the serum
HAI antibody responses induced by the VN04 vaccine was low.
We sought to implement a more sensitive ELISpot assay to
measure whole virus- and HA-specific antibody secreting cells
(ASC) in draining paratracheal lymph node (TLN) and PBMC
from vaccinated ferrets. The ELISpot assay has been shown to be
a sensitive tool for detecting cellular immunity following influenza
vaccination in humans [25,26,27]. To test the sensitivity of this
ELISpot assay, ferrets were inoculated intranasally with either the
H5N1 VN04 ca or H5N1 HK03 ca vaccine virus and were
sacrificed 5 or 10 days later to isolate TLN cells and PBMC. The
ELISpot data obtained from TLN samples (Fig. 1) showed that
H5N1 virus-specific ASC could be detected after vaccination with
either of the H5N1 ca vaccine viruses and similar data were
obtained from PBMC (data not shown). However, the magnitude
and kinetics of IgM and IgG ASC induced by the VN04 ca vaccine
appeared to be lower and slower than the HK03 vaccine virus on
both days 5 and 10 post-vaccination (Fig 1). The virus-specific IgM
ASC response was higher on day 5 than day 10 for HK03 ca virus
immunized animals, but higher on day 10 than day 5 for VN04 ca
virus immunized animals, indicating that the initial IgM response
induced by the VN04 ca virus was slower and weaker than that
induced by the HK03 ca virus. The IgG secreting ASC response
was much higher on day 10 than day 5 for both HK03 ca and
VN04 ca vaccine immunized animals. Again, HK03 ca vaccine
induced more IgG ASC than VN04 ca vaccine (p,0.05). H5 HA-
specific IgM and IgG ASC were also detected on day 10 for both
viruses. However, the level of HA-specific ASC was only about
15% of the total virus-specific ASC, indicating a majority of the
ASC induced by H5N1 ca virus vaccine were against antigens
other than HA. The level of the H5 HA-specific ASC on day 5 was
lower than on day 10 (data not shown). Thus, consistent with the
HAI data, the ELISpot data also showed that the HK03 ca vaccine
induced a better immune response than the VN04 ca vaccine.
LAIV-vaccinated animals show a more rapid immune
responses to the H5N1 ca vaccine than unvaccinated
animals
Most people have some immunity to influenza H1N1, H3N2,
and B viruses due to previous natural infections or immunization
with influenza vaccines. To mimic this sero-positive status and to
determine whether preexisting heterosubtypic immunity affects
responses to subsequent vaccination with H5N1ca vaccine viruses,
ferrets were primed intranasally with live attenuated trivalent
seasonal influenza vaccines (LAIV) or medium control prior to
vaccination with the H5N1 HK03 or VN04 ca viruses. Five days
after vaccination with H5N1 ca vaccine, TLN and PBMC samples
were collected for analysis by B cell ELISpot assay to measure
virus- and HA-specific IgG ASC. Responses detected using whole
virus reagents (Fig. 2A) revealed that the number of H2N2 (AA60,
MDV-A) and H5N1 (HK03) virus-specific IgG ASC were
H5N1 Vaccine
PLoS ONE | www.plosone.org 3 February 2009 | Volume 4 | Issue 2 | e4436
significantly higher in ferrets primed with LAIV (Fig. 2A). This
was not completely unexpected because both the H1N1 and
H3N2 viruses in LAIV share the same internal viral proteins such
as the M1 and NP that likely stimulate rapid B cell responses.
However, LAIV priming also significantly enhanced H5 HA-
specific IgG ASC responses, which were not detected in the
unprimed ferrets (Fig. 2A). This finding suggested that the
enhanced H5 HA-specific responses could occur as a result of
expansion of memory B cells that were elicited against the H1N1
and/or H3N2 influenza A virus components in the LAIV vaccine.
H1N1 vaccine primes faster B cell responses to the H5N1
vaccine than the H3N2 vaccinated animals
To determine whether the higher numbers of H5-specific B cell
responses observed after trivalent LAIV priming were due to only
one or both of the influenza A virus components, groups of ferrets
were primed with monovalent H1N1 NC99 ca, H3N2 CA04 ca,
H5N1 HK03 ca vaccine or medium six weeks prior to a second
inoculation with the H5N1 HK03 ca vaccine. Five days after
vaccination with the H5N1 HK03 ca vaccine (Fig. 2B), the level of
virus-specific IgG ASC was very low in the ferrets that initially
received medium, similar to that observed in the first study. In
contrast, ferrets that were primed with the H1N1 NC99, H3N2
CA04 or H5N1 HK03 ca vaccine viruses, had significantly higher
numbers of H5N1 HK03 ca virus-specific IgG ASC after
vaccination with the H5N1 HK03 ca vaccine. The ferrets that
were previously vaccinated with the H1N1, H3N2 and H5N1 ca
vaccine viruses also had B cell response to the H1N1 and H3N2
vaccine virus (Fig. 2B). The number of the H5 HA-specific IgG
ASC was the highest (approximately 10% of total virus-specific
ASC) in the group that received 2 doses of HK03 ca virus (Fig. 2C).
Interestingly, a significant number of H5 HA-specific IgG ASC
(approximately 180 ASC per 10
6
cells) was also observed in the
group that previously received the H1N1 NC99 ca vaccine virus. A
much lower number of ASC was found in the group that received
the H3N2 CA04 ca vaccine virus. As expected, a significant
number of H1 and H3 HA-specific IgG ASC (approximately 250
and 140 per 10
6
cells, respectively) were detected in the groups
that were primed with the H1N1 NC99 and H3N2 CA04 ca
vaccine viruses, respectively.
H1N1 ca vaccine-induced faster B cell responses to the
H5N1 vaccine virus is due to the H1 HA
The neuraminidases of the H5N1 and H1N1 viruses are of the
same (N1) subtype. Several studies have reported a role of the N1
NA-mediated immunity against H5N1 infection in the murine
model and in serology analysis of human serum samples
[14,17,28]. Because it was shown earlier that previous exposure
to H3N2 virus did not affect H5-specific ASC responses and the
H1 HA and H5 HA share some structural similarity [18] and
common epitopes [19], the effect of the H1N1-mediated priming
on the H5N1 ca vaccines was investigated. To confirm that the
enhanced B cell response to the H5N1 virus was due to a primary
immune response to the H1 HA not the N1 NA, an H1N2 virus
containing the HA from the H1N1 NC99 ca virus, the NA from
the H3N2 WY03 ca virus and the internal protein gene segments
from MDV-A was generated. Priming with this H1N2 virus in
ferrets elicited a similar level of B cell response (122 ASC per
million cells) as those (120 ASC per million cells) primed with the
H5N1 HK03 vaccine virus (Fig. 3), confirming that the enhanced
H5N1 B cell response was due to the H1 HA and not the N1 NA.
The number of the H5 HA-specific IgG ASC in the H1N2 virus
primed ferrets was higher than those primed with the H1N1 ca
vaccine virus, which was possibly due to the better replication of
the H1N2 ca virus than the H1N1 ca virus in the upper respiratory
tract of ferrets (data not shown).
H5-, H1- and H3-specific serum antibody responses
following prime-boost vaccination in ferrets
As described earlier, previous exposure to a virus containing the
H1 HA resulted in an increased H5-specific B cell response. To
examine if the increased ASC response reflected a greater
antibody response, serum antibody titers were examined by
HAI, microneutralization and ELISA assays. Groups of ferrets
were immunized with medium, the H1N1 NC99 ca, H3N2 CA04
ca, H5N1 HK03 ca or VN04 ca virus, and 6 weeks later, a second
dose of the H5N1 HK03 ca or VN04 ca vaccine was administered
intranasally. Serum samples were collected 6 weeks after the 1
st
vaccination (post dose 1) and 1 and 3 weeks after the 2
nd
vaccination (Table 1). At 6 weeks after the first dose neutralizing
antibodies against homologous vaccine virus were detected in the
ferrets that received vaccine virus but not in the control group that
Figure 1. H5N1-specific B cells were detected in ferrets infected
with live attenuated H5N1
ca
vaccines. Ferrets were intranasally
administered the H5N1 VN04 ca or HK03 ca viruses on day 0. Five and
ten days post-inoculation, ferrets were sacrificed to collect paratracheal
lymph nodes (TLN) and a B cell ELISpot assay was performed using
lymphocytes isolated from TLN and BPL-inactivated H5N1 HK03 ca virus
or rH5 HA antigens. The number of IgM ASC (A) and IgG ASC (B) are
presented as per 10
6
lymphocytes.
doi:10.1371/journal.pone.0004436.g001
H5N1 Vaccine
PLoS ONE | www.plosone.org 4 February 2009 | Volume 4 | Issue 2 | e4436
Figure 2. Previous exposure with seasonal LAIV or H1N1
ca
virus induced a faster H5N1-specific immune response. (A) Ferrets were
intranasally inoculated with either medium or trivalent LAIV on day 0. Six weeks later, ferrets were intranasally inoculated with the H5N1 HK03 ca
vaccine and five days later, lymphocytes isolated from TLN were examined for ASC against H2N2 MDV-A, H5N1 HK03 ca viruses and rH5 HA antigens
by B cell ELISpot analysis. Ferrets were intranasally inoculated with medium or the monovalent H1N1 NC99, H3N2 CA04, H5N1 HK03 ca vaccine
H5N1 Vaccine
PLoS ONE | www.plosone.org 5 February 2009 | Volume 4 | Issue 2 | e4436
received medium only. The H3N2 CA04 ca vaccine induced the
highest mean neutralizing antibody (titer of 4064) and the H1N1
NC99 ca vaccine induced a mean neutralizing antibody titer of 50.
Neither H1N1 nor H3N2 ca viruses induced antibodies that cross-
neutralized the H5N1 HK03 ca virus. The H5N1 HK03 ca
vaccine induced the H5-specific neutralizing antibody at a mean
titer of 320 (Table 1). One week after administration of the H5N1
HK03 ca vaccine, the ferrets that received two doses of the HK03
ca vaccine had levels of H5-specific neutralizing antibody that were
significantly higher than in animals that received a single dose of
the H5N1 HK03 ca virus. In contrast to the B cell ELISpots
results, animals primed with a dose of H1N1 ca and boosted with a
second dose of H5N1 ca for one week had low titers of antibodies
against the H5 antigen (Table 1). Previous vaccination with the
H3N2 CA04 ca vaccine had little effect on the antibody response
to the H5N1 ca vaccine. When measured at three weeks after
vaccination with the H5N1 HK03 ca virus, the level of the H5-
specific antibody in the animals that received a dose of medium,
the H1N1 NC99 or H3N2 CA04 ca vaccines as the first dose was
much lower (3- to 5-fold) than the animals that received two doses
of the H5N1 HK03 ca vaccine virus.
The effect of H1N1 ca vaccination on the H5N1 VN04 ca
vaccine induced antibody response was also evaluated (Table 1).
As expected, H1N1-specific antibody did not cross-react with the
H5N1 VN04 ca virus. One week after the second vaccination with
the H5N1 VN04 ca virus, H5N1-specific neutralizing (titer of 50)
antibodies were detected in the ferrets that previously received the
H1N1 NC99 ca vaccine, and this titer was significantly higher than
the ferrets that received medium only (titer of 14, p,0.005) but
much lower than the animals that received two doses of the H5N1
VN04 ca vaccine (titer of 806). However, three weeks after
vaccination with the H5N1 VN04 ca vaccine there was no
difference in antibody levels between the animals that received the
first dose of medium or the H1N1 NC99 ca vaccine. Again,
animals that received two doses of the H5N1 HK03 ca vaccine had
neutralizing antibodies more than 5-fold higher than those that
received the H1N1 or H3N2 ca vaccine as the first dose. Thus, the
H1 HA induced enhanced production of H5N1-specific neutral-
izing antibody was temporary. Similar results were also obtained
by the HAI assay (data not shown).
To further evaluate the H1 induced priming effect on the H5N1
antibody response, an ELISA assay was performed to measure
levels of HA-specific serum IgG antibodies using rHA as the
antigen (Fig. 4). Six weeks after the 1
st
vaccination, a substantial
Figure 3. The priming effect of the H1N1
ca
vaccine is induced
by the H1 HA. Ferrets were intranasally inoculated with the H1N1
NC99 ca virus, a reassortant H1N2 ca, H3N2 CA04 ca or H5N1 HK03 ca
virus on day 0 and intranasally inoculated with the H5N1 HK03 ca virus
five weeks later. The animals were sacrificed 5 days later and B cell
ELISpot analysis was performed with lymphocytes isolated from TLN
using rH5 HA as antigen. IgG antibody secreting B cells are presented as
the number of ASC per 10
6
lymphocytes.
doi:10.1371/journal.pone.0004436.g003
viruses on day 0 and intranasally inoculated with the H5N1 HK03 ca vaccine six weeks later. The B cell ELISpot analysis was performed with
lymphocytes isolated from TLN using the indicated ca vaccine virus (B) or rHA (C) as antigens. The IgG antibody secreting B cells are presented as the
number of ASC per 10
6
lymphocytes.
doi:10.1371/journal.pone.0004436.g002
Table 1. Serum antibody response to influenza viruses after one and two doses of intranasal vaccine.
1
st
dose vaccine (Day 0) 2
nd
dose vaccine (Day 42) Neutralizing antibody GMT against the indicated vaccine virus antigens
6 wk post dose-1 1 wk post dose-2 3 wk post dose-2
1st Vac H5N1 H5N1 H5N1
Medium H5N1 HK03 ca ,10 ,10 40 761
H1N1 NC99 ca 50 ,10 50 403
H3N2 CA04 ca 4064 ,10 18 419
H5N1 HK03 ca 320 320 2560 2281
Medium H5N1 VN04 ca ,10 ,10 14 71
H1N1 NC99 ca 63 ,10 50 71
H5N1 VN04 ca 45 45 806 403
Groups of three ferrets were vaccinated intranasally with the indicated 1
st
dose of vaccine and 42 days later were inoculated with a 2
nd
dose of vaccine (H5N1 HK03 ca
or VN04 ca). Serum samples were collected 6 weeks after the 1
st
dose (pre-dose 2), 1 week and 3 weeks after the 2
nd
dose, respectively, and antibody titers (geometric
mean titers from 3 animals) against the first or second vaccine viruses were determined by microneutralization assay.
doi:10.1371/journal.pone.0004436.t001
H5N1 Vaccine
PLoS ONE | www.plosone.org 6 February 2009 | Volume 4 | Issue 2 | e4436
level of homologous HA-specific IgG was present (Fig. 4A). It was
noted that the H1 and H5 specific antibodies had a low level of
cross reactivity with the H5 and H1 antigens, respectively. The
level of the H5-specific ELISA antibodies was much higher in the
group that received the H1N1 NC99 ca virus as the first dose and
the H5N1 HK03 ca virus as the second dose than the ferrets that
previously received the H3N2 CA04 ca virus and those that did not
receive any virus (medium), although the titer was lower than the
ferrets that received 2 doses of the H5N1 HK03 ca vaccine
(Fig. 4B). The antibodies from the ferrets that received 2 doses of
the H5N1 HK03 ca vaccine also reacted with the rH1 HA. The
H5-specific IgG antibodies continued to increase until 3 weeks
following the 2
nd
dose and reached a level that was similar among
all the groups (data not shown). Similar data were also obtained for
Figure 4. HA-specific antibodies measured by an ELISA assay. Ferrets were intranasally inoculated with medium or the H1N1 NC99, H3N2
CA04, H5N1 HK03 ca vaccine viruses on day 0 and intranasally inoculated with the HK03 ca vaccine virus six weeks later. Serum samples were
collected 6 weeks after the 1st dose (A) and one week after the 2nd dose (B). ELISA was performed with RDE-treated serum using rH5, rH1 or rH3 HA
as antigens.
doi:10.1371/journal.pone.0004436.g004
H5N1 Vaccine
PLoS ONE | www.plosone.org 7 February 2009 | Volume 4 | Issue 2 | e4436
ferrets that received the H5N1 VN04 ca vaccine as a second dose
(data not shown). Thus, these data indicated that the H1- and H5-
specific binding antibodies cross-reacted with each other and
previous exposure to H1N1 vaccine appeared to result in a more
rapid H5-specific humoral immune response to the H5 HA
protein.
H1N1 vaccine-induced immunity prevented replication
of the H5N1 ca vaccine virus
To determine whether previous vaccination with the H1N1
NC99 ca vaccine would affect subsequent replication of the H5N1
ca vaccine virus in the respiratory tract, groups of six naı
¨ve ferrets
were primed with the H1N1 NC99 ca, H3N2 WY03 ca, H5N1
HK03 ca vaccine or medium only. Four weeks later, animals were
inoculated intranasally with the H5N1 HK03 ca vaccine and
sacrificed on day 3 post-inoculation to collect nasal turbinates to
quantify replication of the H5N1 HK03 ca vaccine virus in the
upper respiratory tract of ferrets. As shown in Table 2, the mean
titer of the H5N1 HK03 ca vaccine virus detected in the upper
respiratory tract of ferrets that had previously received medium
was 10
4.5
EID
50
/g of tissue. In contrast, none of the ferrets that
were primed with the H1N1 NC99 ca or the H5N1 HK03 ca
vaccine had detectable virus in the upper respiratory tract. The
H3N2 WY03 ca vaccine priming protected 2 of 6 ferrets and the
H5N1 HK03 virus replicated to a mean titer of 10
2.2
EID
50
/g of
tissue in this group of ferrets. These results indicated that
heterosubtypic immunity from H1N1 ca virus infection or
vaccination reduced H5N1 ca vaccine virus replication.
Discussion
In this study, we demonstrate that live attenuated H5N1 vaccine
induced immunity can be detected by the B cell ELISpot assay.
The protective level of ASC is not well established yet, however,
the B cell response to the H5N1 HK03 ca vaccine is greater than to
the VN04 ca vaccine and the magnitude of the ASC response
correlates with serum antibody level as determined by micro-
neutralization assay. The H5N1 HK03 and VN04 viruses differ by
9 amino acids in the HA molecule and the amino acid at position
223 is known to contribute to receptor binding specificity [29].
The HA of the HK03 virus that preferentially binds to sialic acid
receptors with a2,6-linked oligosaccharide linkages contains serine
at residue 223 and the HA of the VN04 virus that prefers an avian-
like receptor with a2,3-linked oligosaccharide linkages contains
asparagine at this residue [30,31]. In addition, the length of the
NA of the H5N1 HK03 virus differs from the VN04 virus; the
VN04 virus, like most of the H5N1 isolates, has a deletion of 20
amino acids in the NA stalk whereas the HK03 virus does not have
this deletion. The differences in the HA and NA sequences
between the H5N1 HK03 and VN04 viruses presumably
contribute to the observed difference in vaccine immunogenicity.
Despite the lower immune response induced by VN04 ca virus,
two doses of the H5N1 VN04 ca vaccine offered complete
protection against homologous and heterologous H5N1 wt virus
lethal challenge in mice and provided protection against
replication of the H5N1 wt virus in the respiratory tracts of mice
and ferrets [7].
In addition to the local draining lymph nodes (TLN), ASC were
also detected in the PBMC of the vaccinated ferrets at a level
slightly lower than those detected in TLN, therefore, only the data
obtained with TLN are presented. Virus-specific memory B cells
in the lungs can persist for a long time along with germinal center
B cells and plasma cells and appear to be a unique feature of the
mucosal memory response [16]. In response to re-encountered
antigens, memory B cells robustly secrete antibodies against the
pathogen and this memory response is much faster than that of
primary B cells due to quantitative and qualitative changes in
antigen-specific B cells and helper T cells. In this study, we found
that previous exposure to the H1N1 ca virus could accelerate the
memory B cell response to the H5N1 virus. The heterosubtypic
antibody response as detected by ELISA in the serum could
prevent replication of the H5N1 HK03 ca vaccine virus in the
respiratory tract, suggesting that protective immunity was
enhanced by a priming dose of H1N1 ca vaccine. A recent study
also showed that the ferrets immunized with the H1N1 virus-like
particles (VLP) had a low level of neutralizing antibody against the
H5N1 virus and cleared the H5N1 challenge virus rapidly and had
reduced morbidity [32]. There was a concern that heterosubtypic
immunity might reduce vaccine efficacy by reducing vaccine virus
replication in the upper respiratory tract. However, despite
restricted replication of the H5N1 ca virus in the upper respiratory
tract of the H1N1 exposed ferrets, the level of H5N1-specific
neutralizing antibodies in the animals that previously received the
H1N1 ca vaccine was similar to the seronegative animals. Our
previous study indicated that two doses of the H1N1 A/New
Caledonia/20/99 ca virus were unable to protect ferrets from
replication of a high dose (10
7
TCID
50
) of H5N1 HK97 wt virus in
the respiratory tracts of ferrets [7]. However, it remains to be
determined whether the H1N1 ca virus could offer a protective
benefit from a lower challenge dose of H5N1 wt virus. Despite the
faster onset of immunity to the H5N1 ca vaccine, the antibodies
produced in animals that received the H1N1 ca vaccine followed
by the H5N1 ca vaccine are at least 5-fold lower that the animals
that received two doses of the H5N1 ca vaccines. Thus, it is likely
that the immunity provided by previous immunization with an
H1N1 ca virus is limited and priming with seasonal LAIV cannot
replace the use of 2 doses of an H5-specific vaccine.
Table 2. Effect of the H1N1 and H3N2 ca vaccines on replication of the H5N1 HK03 ca virus in the upper respiratory tract of ferrets.
Vaccine
#of animals
per group
GMT of homologous
HAI antibody
#of Animals with H5N1
detected in NT
Mean Virus Titer in NT
(log
10
EID
50
/g6SE)
Medium 6 ,46 4.560.5
H1N1 NC99 ca 647 0 #1.5
H3N2 WY03 ca 6 203 4 2.260.7
H5N1 HK03 ca 640 0 #1.5
Groups of ferrets were vaccinated with the indicated virus and four weeks later inoculated with the H5N1 HK03 ca vaccine. Antibody titers against homologous vaccine
virus were determined by HAI assay and expressed as geometric mean titer. The H5N1 HK03 ca virus titer in nasal turbinates (NT) on day 3 post-inoculation is expressed
as log
10
EID
50
per gram of tissues calculated from the mean of 6 animals.
doi:10.1371/journal.pone.0004436.t002
H5N1 Vaccine
PLoS ONE | www.plosone.org 8 February 2009 | Volume 4 | Issue 2 | e4436
It has been observed frequently that individuals recovered from
influenza virus infection are protected against subsequent infection
by antigenic drift variant viruses within the same subtype
[10,11,12] and to a lesser extent from infection by a different
subtype due to heterosubtypic immunity [13]. Recently, several
reports have described heterosubtypic immunity from seasonal
influenza vaccines to H5N1 infection in mice and humans
[8,14,15,16]. Ichinohe et al [14] showed that intranasal inocula-
tion of an inactivated trivalent seasonal influenza vaccine provided
cross-protection against H5N1 infection in mice. Such studies are
difficult to conduct in humans. Ferrets develop symptoms upon
influenza infection that resemble those of humans including
sneezing, body temperature variation and weight loss and have
been shown to be an appropriate model for influenza virus
research. In this study, we demonstrated that the faster H5N1 B
cell response induced by the H1N1 ca vaccine in ferrets was
mediated by the H1 HA protein as demonstrated by a similar
effect caused by an H1N2 virus. Although the accelerated H5N1
response following previous exposure to the H1N1 ca virus was
barely detected by HAI and microneutralization assays, we found
a temporal rise of mincroneutralizing antibody in H5N1 VN04 ca
vaccinated ferrets that were previously exposed to the H1N1
NC04 ca virus (Table 1). We could demonstrate cross-reactivity of
H1N1 and H5N1 ca vaccine induced HA antibodies by ELISA
assay. These data confirmed that the H1 and H5 HA contain
some conserved epitopes that could elicit cross-reactive antibodies
[19,33] because of their structure similarity [18].
N1 NA-induced protection against experimental H5N1 virus
infection has been reported in mice and by the finding that human
sera are capable of inhibiting the NA enzymatic activity of the
H5N1 VN04 virus [17]. Our study was not designed to examine
the contribution of the N1 protein of the H1N1 virus to H5N1
immunity. We cannot exclude the possibility that the N1-induced
immunity might also contribute to the restricted replication of the
H5N1 ca virus in the upper respiratory tract of ferrets.
Our current study indicates that previous exposure to the H3N2
ca virus was less protective than the H1N1 ca virus in restricting
replication of the H5N1 ca vaccine virus in the upper respiratory
tract of ferrets. However, replication of the H5N1 vaccine virus in
ferrets previously primed with an H3N2 ca virus was also greatly
reduced compared to the control animals. This could be because
the H1N1, H3N2 and H5N1 ca vaccine viruses share 6 internal
protein gene segments. As shown by the ELISpot assay, the
number of ASC against the vaccine virus was much higher than
HA-specific ASC (compare Fig. 2B with Fig. 2C). In addition to
the protective immune response against the HA and NA surface
proteins, influenza viruses also induce immune responses against
conserved viral proteins such as NP and M1 that could result in
heterosubtypic protection [34] to restrict H5N1 virus replication.
An earlier report [35] showed that previous mucosal delivery of
trivalent influenza vaccine offered protection against H5N1 wt
virus lethal infection in the mouse model. We also showed
previously that H2N2 AA ca vaccinated mice were partially
protected from the lethal challenge of the H5N1 wt viruses [7].
This type of heterosubtypic response could be mediated by
secondary CTL responses involving CD8
+
or CD4
+
T cells as
reported previously [15,36].
In summary, our study supports the notion that previous
vaccination with seasonal influenza vaccine may accelerate onset
of immunity by an H5N1 ca vaccine. Since the influenza pandemic
vaccine may not be available until some time well into the first
wave or early in the second wave of a pandemic, an earlier
response may be of value in pandemic preparedness.
Acknowledgments
We are very grateful to Scott Jacobson, Kim Ngo, Brett Pickell, Ernesto
Madariaga, Stephanie Gee and Nick Nguyen for performing the ferret
studies, Anu Cherukuri for review of the manuscript, Jennifer Woo and
members of HJ’s group for technical support and discussions.
Author Contributions
Conceived and designed the experiments: ME GK HJ. Performed the
experiments: XC ME QX HZ DK. Analyzed the data: XC ME QX HJ.
Contributed reagents/materials/analysis tools: XC ME QX HZ DK HJ.
Wrote the paper: XC KS GK HJ.
References
1. Taubenberger JK (2006) The origin and virulence of the 1918 ‘‘Spanish’’
influenza virus. Proc Am Philos Soc 150: 86–112.
2. Scholtissek C, Rohde W, Von Hoyningen V, Rott R (1978) On the origin of the
human influenza virus subtypes H2N2 and H3N2. Virology 87: 13–20.
3. Schafer JR, Kawaoka Y, Bean WJ, Suss J, Senne D, et al. (1993) Origin of the
pandemic 1957 H2 influenza A virus and the persistence of its possible
progenitors in the avian reservoir. Virology 194: 781–788.
4. Subbarao K, Joseph T (2007) Scientific barriers to developing vaccines against
avian influenza viruses. Nat Rev Immunol 7: 267–278.
5. Belshe RB, Gruber WC, Mendelman PM, Mehta HB, Mahmood K, et al.
(2000) Correlates of immune protection induced by live, attenuated, cold-
adapted, trivalent, intranasal influenza virus vaccine. J Infect Dis 181:
1133–1137.
6. Brokstad KA, Cox RJ, Eriksson JC, Olofsson J, Jonsson R, et al. (2001) High
prevalence of influenza specific antibody secreting cells in nasal mucosa.
Scand J Immunol 54: 243–247.
7. Suguitan AL Jr, McAuliffe J, Mills KL, Jin H, Duke G, et al. (2006) Live,
attenuated influenza A H5N1 candidate vaccines provide broad cross-protection
in mice and ferrets. PLoS Med 3: e360.
8. Lu X, Edwards LE, Desheva JA, Nguyen DC, Rekstin A, et al. (2006) Cross-
protective immunity in mice induced by live-attenuated or inactivated vaccines
against highly pathogenic influenza A (H5N1) viruses. Vaccine 24: 6588–6593.
9. Maassab HF (1967) Adaptation and growth characteristics of influenza virus at
25 degrees C. Nature 213: 612–614.
10. Liew FY, Russell SM, Appleyard G, Brand CM, Beale J (1984) Cross-protection
in mice infected with influenza A virus by the respiratory route is correlated with
local IgA antibody rather than serum antibody or cytotoxic T cell reactivity.
Eur J Immunol 14: 350–356.
11. McMichael AJ, Gotch F, Cullen P, Askonas B, Webster RG (1981) The human
cytotoxic T cell response to influenza A vaccination. Clin Exp Immunol 43:
276–284.
12. Couch RB, Kasel JA (1983) Immunity to influenza in man. Annu Rev Microbiol
37: 529–549.
13. Schulman JL, Kilbourne ED (1965) Induction of Partial Specific Heterotypic
Immunity in Mice by a Single Infection with Influenza a Virus. J Bacteriol 89:
170–174.
14. Ichinohe T, Tamura S, Kawaguchi A, Ninomiya A, Imai M, et al. (2007) Cross-
protection against H5N1 influenza virus infection is afforded by intranasal
inoculation with seasonal trivalent inactivated influenza vaccine. J Infect Dis
196: 1313–1320.
15. Kreijtz JH, Bodewes R, van Amerongen G, Kuiken T, Fouchier RA, et al.
(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.
16. Takahashi Y (2007) Memory B cells in systemic and mucosal immune response:
implications for successful vaccination. Biosci Biotechnol Biochem 71: 2358–2366.
17. Sandbulte MR, Jimenez GS, Boon AC, Smith LR, Treanor JJ, et al. (2007)
Cross-reactive neuraminidase antibodies afford partial protection against H5N1
in mice and are present in unexposed humans. PLoS Med 4: e59.
18. Ha Y, Stevens DJ, Skehel JJ, Wiley DC (2001) X-ray structures of H5 avian and
H9 swine influenza virus hemagglutinins bound to avian and human receptor
analogs. Proc Natl Acad Sci U S A 98: 11181–11186.
19. Smirnov YA, Lipatov AS, Gitelman AK, Okuno Y, Van Beek R, et al. (1999) An
epitope shared by the hemagglutinins of H1, H2, H5, and H6 subtypes of
influenza A virus. Acta Virol 43: 237–244.
20. Kreijtz JH, de Mutsert G, van Baalen CA, Fouchier RA, Osterhaus AD, et al.
(2008) Cross-recognition of avian H5N1 influenza virus by human cytotoxic T-
lymphocyte populations directed to human influenza A virus. J Virol 82:
5161–5166.
21. Hoffmann E, Mahmood K, Chen Z, Yang C, Spaete J, et al. (2005) Multiple
gene segments control the temperature sensitivity and attenuation phenotypes of
ca B/Ann Arbor/1/66. J Virol 79: 11014–11021.
H5N1 Vaccine
PLoS ONE | www.plosone.org 9 February 2009 | Volume 4 | Issue 2 | e4436
22. Jin H, Lu B, Zhou H, Ma C, Zhao J, et al. (2003) Multiple amino acid residues
confer temperature sensitivity to human influenza virus vaccine strains (FluMist)
derived from cold-adapted A/Ann Arbor/6/60. Virology 306: 18–24.
23. Govorkova EA, Webby RJ, Humberd J, Seiler JP, Webster RG (2006)
Immunization with reverse-genetics-produced H5N1 influenza vaccine protects
ferrets against homologous and heterologous challenge. J Infect Dis 194:
159–167.
24. Hampson AW (2006) Ferrets and the challenges of H5N1 vaccine formulation.
J Infect Dis 194: 143–145.
25. Lindemann M, Witzke O, Lutkes P, Fiedler M, Kreuzfelder E, et al. (2006)
ELISpot assay as a sensitive tool to detect cellular immunity following influenza
vaccination in kidney transplant recipients. Clin Immunol 120: 342–348.
26. Cox RJ, Haaheim LR, Ericsson JC, Madhun AS, Brokstad KA (2006) The
humoral and cellular responses induced locally and systemically after parenteral
influenza vaccination in man. Vaccine 24: 6577–6580.
27. Sasaki S, Jaimes MC, Holmes TH, Dekker CL, Mahmood K, et al. (2007)
Comparison of the influenza virus-specific effector and memory B-cell responses
to immunization of children and adults with live attenuated or inactivated
influenza virus vaccines. J Virol 81: 215–228.
28. Gioia C, Castilletti C, Tempestilli M, Piacentini P, Bordi L, et al. (2008) Cross-
subtype immunity against avian influenza in persons recently vaccinated for
influenza. Emerg Infect Dis 14: 121–128.
29. Hoffmann E, Lipatov AS, Webby RJ, Govorkova EA, Webster RG (2005) Role
of specific hemagglutinin amino acids in the immunogenicity and protection of
H5N1 influenza virus vaccines. Proc Natl Acad Sci U S A 102: 12915–12920.
30. van Riel D, Munster VJ, de Wit E, Rimmelzwaan GF, Fouchier RA, et al. (2006)
H5N1 Virus Attachment to Lower Respiratory Tract. Science 312: 399.
31. Yamada S, Suzuki Y, Suzuki T, Le MQ, Nidom CA, et al. (2006)
Haemagglutinin mutations responsible for the binding of H5N1 influenza A
viruses to human-type receptors. Nature 444: 378–382.
32. Haynes JR, Dokken L, Wiley JA, Cawthon AG, Bigger J, et al. (2008) Influenza-
pseudotyped Gag virus-like particle vaccines provide broad protection against
highly pathogenic avian influenza challenge. Vaccine, In Press.
33. Ahmed R, Oldstone MB, Palese P (2007) Protective immunity and susceptibility
to infectious diseases: lessons from the 1918 influenza pandemic. Nat Immunol
8: 1188–1193.
34. Scherle PA, Gerhard W (1986) Functional analysis of influenza-speci fic helper T
cell clones in vivo. T cells specific for internal viral proteins provide cognate help
for B cell responses to hemagglutinin. J Exp Med 164: 1114–1128.
35. Tumpey TM, Renshaw M, Clements JD, Katz JM (2001) Mucosal delivery of
inactivated influenza vaccine induces B-cell-dependent heterosubtypic cross-
protection against lethal influ enza A H5N1 virus infection. J Virol 75:
5141–5150.
36. Tamura S, Tanimoto T, Kurata T (2005) Mechanisms of broad cross-protection
provided by influenza virus infection and their application to vaccines.
Jpn J Infect Dis 58: 195–207.
H5N1 Vaccine
PLoS ONE | www.plosone.org 10 February 2009 | Volume 4 | Issue 2 | e4436
... The effects of A(H1N1) and A(H3N2) pre-immunity on the A(H5N1) subtype or the effect of A(H5N1) on A(H1N1) or A(H3N2) vaccination or subsequent viral challenge are not well known. Although studies have been performed in pre-immune mice [90,91], few studies have addressed pre-immunity using the viruses of the A(H5N1) subtype [92] (Table S4). Pre-pandemic vaccines for A(H5N1) influenza viruses elicited low seroconversion proportions in immunologically naïve ferrets. ...
... Without letting the immune system return to baseline, misrepresentation of cross-protection may be observed. Cheng et al. [92] found that the heterologous protective titers waned to levels not significantly different than the negative naïve control ferret group providing only temporary protection. ...
... Before this reagent development, research was limited to characterizing serum and nasal washes for neutralizing antibodies and measuring clinical signs after infection. Therefore, future studies can capture the cellular immune reactions of T-cell responses with IFN-γ ELISAs [120], peripheral blood leukocyte tracking [121], and ELISpots [88,122], as well as the humoral immunity [18,92,123,124]. ...
Article
Full-text available
The initial exposure to influenza virus usually occurs during childhood. This imprinting has long-lasting effects on the immune responses to subsequent infections and vaccinations. Animal models that are used to investigate influenza pathogenesis and vaccination do recapitulate the pre-immune history in the human population. The establishment of influenza pre-immune ferret models is necessary for understanding infection and transmission and for designing efficacious vaccines.
... The attenuated phenotype is attributed to five mutations in the PB1, PB2, and NP gene segments and does not replicate in the lungs of mice and ferrets (Jin et al., 2003). LAIV vaccines induce innate and adaptive immune responses at the site of administration and have been associated with increased systemic IgG antibodies and mucosal IgA antibodies (Beyer et al., 2002;Cheng et al., 2009). ...
... The dilution of turkey and guinea pig RBCs is fairly standard (generally 0.5%v/v RBCs in saline). However, several studies detected H5N1 HI titres using horse RBCs ranging from 0.5%-2% without additional explanation (Cheng et al., 2009, Stephenson et al, 2004, Suguitan et al., 2006. ...
... Long-term research has been conducted on influenza virus-specific antibody responses. Particularly, in recent years, with the advancements in the development of technology that make it possible to analyze the antibodies created by individual human B cells, we have made enormous strides in our knowledge of the antibody response to IV [265]. Although the exact processes are unknown, it is known that antibody responses elicited by natural illness are often more extensive and durable than antibody reactions induced by vaccination. ...
Article
Full-text available
The avian influenza A virus (AIV) is naturally prevalent in aquatic birds, infecting different avian species and transmitting from birds to humans. Both AIVs, the H5N1 and H7N9 viruses, have the potential to infect humans, causing an acute influenza disease syndrome in humans, and are a possible pandemic threat. AIV H5N1 is highly pathogenic, whereas AIV H7N9 has comparatively low pathogenicity. A clear insight into the disease pathogenesis is significant to understand the host’s immunological response, which in turn facilitates the design of the control and prevention strategies. In this review, we aim to provide comprehensive details on the pathogenesis and clinical features of the disease. Moreover, the innate and adaptive immunological responses to AIV and the recent studies conducted on the CD8+ T cell immunity against AIVs are detailed upon. Further, the current status and advancement in the development of AIV vaccines, along with the challenges, are also discussed. The information provided will be helpful in combating the transmission of AIV from birds to humans and, thus, preventing severe outbreaks leading to pandemics worldwide.
... Early studies demonstrated that preexisting antibodies to H1N1 or H3N2 influenza strains did not interrupt the infectivity, immunogenicity, and replication of H1N1 and H3N2 LAIVs in young infants and children, suggesting that preexisting immunity does not limit the use of LAIVs in humans [122]. Furthermore, ferrets primed by trivalent LAIVs (H1N1, H3N2, and B) elicited more robust cross-reactive antibody responses following H5N1 LAIV than naive animals, presenting the beneficial effects of vaccination-induced preexisting immunity on boosting cross-reactive antibody responses by LAIVs [123]. The efficiency of boosting of HA antibodies with LAIVs showed differences between children and adults, according to their history of exposure to influenza viruses. ...
Article
Full-text available
Influenza virus infection remains a major public health challenge, causing significant morbidity and mortality by annual epidemics and intermittent pandemics. Although current seasonal influenza vaccines provide efficient protection, antigenic changes of the viruses often significantly compromise the protection efficacy of vaccines, rendering most populations vulnerable to the viral infection. Considerable efforts have been made to develop a universal influenza vaccine (UIV) able to confer long-lasting and broad protection. Recent studies have characterized multiple immune correlates required for providing broad protection against influenza viruses, including neutralizing antibodies, non-neutralizing antibodies, antibody effector functions, T cell responses, and mucosal immunity. To induce broadly protective immune responses by vaccination, various strategies using live attenuated influenza vaccines (LAIVs) and novel vaccine platforms are under investigation. Despite superior cross-protection ability, very little attention has been paid to LAIVs for the development of UIV. This review focuses on immune responses induced by LAIVs, with special emphasis placed on the breadth and the potency of individual immune correlates. The promising prospect of LAIVs to serve as an attractive and reliable vaccine platforms for a UIV is also discussed. Several important issues that should be addressed with respect to the use of LAIVs as UIV are also reviewed.
... The contribution of the different arms of the immune system to LAIV-induced protection has been evaluated in mice and ferrets [22][23][24][25]. The body temperature of ferrets is about 39°C; therefore, ferrets are used to test the attenuation phenotype conferred by the temperature-sensitive mutations of the Ann Arbor cold-adapted virus, with replication limited to the upper respiratory tract. ...
... Cytotoxic CD8 + T cells that recognize conserved peptides can kill virus-infected cells and also produce antiviral cytokines, promoting viral clearance and recovery. live attenuated influenza vaccine (LAIV) on days 5 and 10 postinfection, and their magnitude correlated with serum neutralizing Ab responses (48). The kinetics of these responses were consistent with the human Ab-secreting cell response to seasonal LAIV (49). ...
Article
Avian influenza A viruses (IAVs) naturally infect different avian species, and aquatic birds are their natural reservoir. Sporadically, avian IAVs can be transmitted to humans, and some, such as H5N1 and H7N9 viruses, cause severe disease in humans. Antigenically novel avian influenza viruses that infect and cause disease in humans pose a potential pandemic threat if they are able to spread efficiently from person to person. The immune response of the host is crucial in determining disease pathogenesis and is the basis for the development of control strategies. In this review, we examine the innate and adaptive immune responses to avian influenza viruses and their role in disease and recovery. Furthermore, we discuss the progress in developing vaccines against avian IAVs and summarize obstacles in designing universal and pandemic influenza vaccines.
... The follow-up studies are needed to elucidate the role of mucosal immunity, such as IgA in protection. Additionally, we used ferrets because they are considered to be the most suitable animal model for influenza vaccine efficacy studies [15,[48][49][50]. Using this animal model, experimental influenza VLP vaccines have previously shown protective efficacy against potentially pandemic avian influenza H5N1 and H9N2 subtype viruses [17,51]. ...
... Ferrets are considered to be the most suitable animal model for influenza vaccine efficacy studies [13,[20][21][22]. Using this and other animal models, experimental influenza VLP vaccines have shown protective efficacy against potentially pandemic H5N1 and H9N2 viruses, as well as the 2009 pandemic H1N1 virus [7,13,15,23,24,16]. ...
Article
Full-text available
In March 2013, diagnosis of the first reported case of human infection with a novel avian-origin influenza A(H7N9) virus occurred in eastern China. Most human cases have resulted in severe respiratory illness and, in some instances, death. Currently there are no licensed vaccines against H7N9 virus, which continues to cause sporadic human infections. Recombinant virus-like particles (VLPs) have been previously shown to be safe and effective vaccines for influenza. In this study, we evaluated the immunogenicity and protective efficacy of a H7N9 VLP vaccine in the ferret challenge model. Purified recombinant H7N9 VLPs morphologically resembled influenza virions and elicited high-titer serum hemagglutination inhibition (HI) and neutralizing antibodies specific for A/Anhui/1/2013 (H7N9) virus. H7N9 VLP-immunized ferrets subsequently challenged with homologous virus displayed reductions in fever, weight loss, and virus shedding compared to these parameters in unimmunized control ferrets. H7N9 VLP was also effective in protecting against lung and tracheal infection. The addition of either ISCOMATRIX or Matrix-M1 adjuvant improved immunogenicity and protection of the VLP vaccine against H7N9 virus. These results provide support for the development of a safe and effective human VLP vaccine with potent adjuvants against avian influenza H7N9 virus with pandemic potential. Copyright © 2015. Published by Elsevier Ltd.
Chapter
Cellular immune responses, through both T and B cells, are critical to understanding the role and regulation of lymphocytes following viral infection, as well as defining responses to vaccination. T cells play a critical role in adaptive immunity, including pathogen elimination through the engagement of CD4 and CD8 receptors, which trigger signaling mechanisms. B cells contribute to generating antibodies following exposure to foreign pathogens through interactions with CD4+ lymphocytes. While these different cell types have distinctly different modes of action in terms of contributions to protection (cytotoxic versus antibody mediated), they account for the majority of adaptive immunity induced following infection or vaccination. While the ability to measure cell-mediated immunity (CMI) has steadily improved, there is much to learn with regard to their contribution to the protection of birds against diseases induced by avian influenza virus. The rapidly increasing knowledge of genomic avian sequences, along with the increasing availability of monoclonal antibodies detecting avian cell-associated antigen markers, has made techniques to measure CMI more specific and informative for researchers.
Article
The measurement of cell-mediated immunity (CMI) is critical to understanding the role and regulation of avian lymphocytes following avian influenza virus (AIV) infection. While these different cell types have distinctly different modes of action in terms of contributions to protection, they account for the majority of adaptive immunity induced following infection or vaccination. Although the ability to measure CMI has steadily improved over the last few years, few studies have examined its role in protection of birds against AIV. The increasing availability of monoclonal antibodies recognizing various avian cell-associated antigens has made this technique more specific and informative.
Article
Full-text available
We compared the effects of adoptively transferred Th cell clones specific for the influenza hemagglutinin (HA), matrix (M), or nucleoprotein (NP) on the antibody response of nude mice infected with A/PR/8/34 influenza virus. We show that the production of antibodies to the HA absolutely requires the presence of virus-specific Th cells. Further, transfer of a Th clone specific for the internal proteins, M or NP, was as effective as was transfer of an HA-specific clone in supporting an antibody response to the HA. With each of the clones, the kinetics of the response were accelerated by approximately 3 d compared with the antibody response of normal BALB/c mice. The HA- and M-specific clones supported an isotype switch from IgM to IgG and IgA similar to that which occurs during a normal antibody response. Finally, as shown by coinfection experiments, the response required a cognate T-B interaction whether the determinants recognized by the Th and B cell are located on the same viral protein or on different viral proteins within the same virus particle. The implications of these findings for understanding the T-B interactions that occur during an effective antiviral antibody response are discussed.
Article
Influenza-pseudotyped Gag virus-like particles (VLPs) were produced via the expression of influenza hemagglutinin (HA), neuraminidase (NA) and the murine leukemia virus Gag product in the baculovirus-insect cell expression system. Hemagglutination specific activities of sucrose gradient-purified VLPs were similar to those of egg-grown influenza viruses but particle morphologies were gamma retrovirus-like in the form of consistent 100nm spheres. Immunization of mice and ferrets demonstrated robust immunogenicity and protection from challenge with no measurable morbidity. Ferret data were striking in that immunization with H5N1 VLPs representing either A/Vietnam/1203/04 or A/Indonesia/5/05 resulted in solid protection against highly pathogenic A/Vietnam/1203/04 challenge with no detectable virus in the upper respiratory tract post-challenge in either group. H1N1 VLP immunization of ferrets resulted in partial protection against H5N1 challenge with markedly accelerated virus clearance from the upper respiratory tract relative to controls. The immunogenicity of influenza-pseudotyped VLPs was not dependent on the adjuvant properties of replication competent contaminating baculovirus. These data demonstrate robust vaccine protection of Gag-based, influenza-pseudotyped VLPs carrying a variety of influenza antigens and suggest applicability toward a number of additional respiratory viruses.
Article
The RNA of the human influenza virus Singapore (H2N2) strain has been labeled in vivo by phosphorus-32 and separated by polyacrylamide gel electrophoresis into eight segments, which were correlated to the corresponding gene functions and/or proteins. The base sequence homology between the individual genes (segments) of the H2N2 virus and those of different influenza A strains has been determined by molecular hybridization. Segments 1, 5, 7, and 8 of the Singapore strain exhibit a base sequence homology of almost 100% as compared to the FM1 strain (HlNl), while the homology between the other segments is significantly lower (24–76%). For the Singapore and Hong Kong (H3N2) strains all segments except that coding for the hemagglutinin (HA, 24%) exhibit a homology close to 100%. The 32P-labeled segment 4 (HA-gene) of the avian influenza A strain duck Ukraine (Hav7Neg2) shows a homology of 92% to Hong Kong, while the homology of at least two other segments is significantly lower. These results are taken as an indication that the H2N2 subtype is derived from the HlN1 subtype by a recombination event retaining four H1N1 segments, while the other four segments are gained from another yet unknown strain. The H3N2 subtype is presumably derived from a H2N2 subtype, retaining seven segments of the H2N2 subtype, while the gene coding for the HA is obtained from the duck Ukraine or another highly related strain.
Article
The observations summarized in this review indicate immunity to infection with type A influenza viruses is subtype specific since little or none is conveyed to subtypes possessing immunologically distinct HA and NA proteins. However, within a subtype, a prior antigenic experience with one variant may prevent or modify illness to another. The resulting degree of subtype immunity depends on the extent of relatedness between variants. Observations with H3N2 viruses indicate that homotypic resistance to subsequent infection and illness with the same virus is potent and of relatively long duration. The long lasting durability of such immunity was indicated by the epidemiologic pattern following the reappearance of H1N1 virus. Knowledge of the duration and specificity of immunity aids considerably in assessing mechanisms that account for host resistance to influenza. Recovery from influenza virus infection must involve a variety of humoral and cell-mediated immune mechanisms, and conclusions regarding the relative importance of each one are not possible at present. To prevent infection, involved immune mechanism(s) must account for: (a) subtype specificity, (b) reduced cross-reactivity of immunity for succeeding antigenic variants, (c) a long duration of immunity, and (d) immunity at the mucosal surface. Only antibody directed toward the HA molecule presently satisfies these properties and thus should be considered the major mediator of resistance to infection. Study of naturally occurring infection is needed for determining the duration and specificity of secretory IgA in nasal and lower respiratory secretions so as to establish its relative importance as a mediator of immunity. However, the described duration, specificity, and consistent relationship to immunity suggest that IgG antibody in respiratory secretions, derived entirely or partly from serum, is the most likely mediator of resistance to natural influenza.
Article
Mice previously infected with an aerosol of A/Rec 31 influenza virus were strongly protected against an aerosol challenge with A/Vic influenza as judged by lung virus titers recovered 2 days after the challenge infection. Such complete homotypic immunity was not achieved by priming with live Rec 31 virus injected i.v. or UV-inactivated Rec 31 virus administered s.c. together with Al(OH)3 and saponin. The reason for the superior protective effect of the natural infection was investigated. The protection induced by respiratory infection with Rec 31 virus was specific for influenza A viruses. It was not correlated with specific serum hemagglutination inhibition antibody titer or cross-reactive cytotoxic T (Tc) cell reactivity. Moreover, the transfer of splenic and lymphoid T cell populations with strong secondary Tc activity did not significantly reduce lung virus titers in recipient mice 3 days after infection. The protection however occurred in parallel with the presence of cross-reactive IgA antibody in the lung washings. It thus appears that local secretory IgA plays a causal role in the prevention of cross-infection by influenza A virus. Serum antibody and Tc cells, on the other hand, may be crucial for recovery from such infection. All mice primed with live Rec 31 virus, administered i.v. or by aerosol and expressing equally high levels of Tc reactivity, survived a lethal challenge with A/PR8 virus. The same challenge, however, killed half of the mice immunized s.c. with inactivated Rec 31 virus which induced only a low level of Tc reactivity.
Article
The human cytotoxic T lymphocyte (CTL) response to challenge with influenza A vaccine was studied. Six of eight volunteers given killed whole influenza virus A/USSR (H1N1) vaccine showed substantial increases on the level of CTL memory 1 month after immunization. The CTL measured at this time showed complete cross-reactivity in their specificity for influenza A/USSR (H1N1) and A/X31 (H3N2) infected cells and also showed HLA restriction. The level of CTL memory increased in only three out of nine donors given subunit vaccine and showed no change in those not given vaccine. If cytotoxic T cells are important in influenza prophylaxis, killed whole virus vaccine should offer better protection than subunit vaccine.
Article
H2N2 influenza A viruses caused the Asian pandemic of 1957 and then disappeared from the human population 10 years later. To assess the potential for similar outbreaks in the future, we determined the antigenicity of H2 hemagglutinins (HAs) from representative human and avian H2 viruses and then analyzed the nucleotide and amino acid sequences to determine their evolutionary characteristics in different hosts. The results of longitudinal virus surveillance studies were also examined to estimate the prevalence of avian H2 isolates among samples collected from wild ducks and domestic poultry. Reactivity patterns obtained with a large panel of monoclonal antibodies indicated antigenic drift in the HA of human H2 influenza viruses, beginning in 1962. Amino acid changes were clustered in two regions of HA1 that correspond to antigenic sites A and D of the H3 HA. By contrast, the antigenic profiles of the majority of avian H2 HAs were remarkably conserved through 1991, resembling the prototype Japan 57 (H2N2) strain. Amino acid changes were distributed throughout HA1, indicating that antibodies do not play a major role in the selection of avian H2 viruses. Phylogenetic analysis revealed two geographic site-specific lineages of avian H2 HAs: North American and Eurasian. Evidence is presented to support interregion transmission of gull H2 viruses. The human H2 HAs that circulated in 1957-1968 form a separate phylogenetic lineage, most closely related to the Eurasian avian H2 HAs. There was an increased prevalence of H2 influenza viruses among wild ducks in 1988 in North America, preceding the appearance of H2N2 viruses in domestic fowl. As the prevalence of avian H2N2 influenza viruses increased on turkey farms and in live bird markets in New York City and elsewhere, greater numbers of these viruses have come into direct contact with susceptible humans. We conclude that antigenically conserved counterparts of the human Asian pandemic strain of 1957 continue to circulate in the avian reservoir and are coming into closer proximity to susceptible human populations.