You are on page 1of 6

Vaccine 29 (2011) 6352–6357

Contents lists available at ScienceDirect

Vaccine
journal homepage: www.elsevier.com/locate/vaccine

A DNA vaccine encoding the E protein of West Nile Virus is protective and
can be boosted by recombinant domain DIII
Anne Schneeweiss a , Stefan Chabierski a , Mathias Salomo a , Nicolas Delaroque a , Samiya Al-Robaiy a ,
Thomas Grunwald b , Kurt Bürki c , Uwe G. Liebert d , Sebastian Ulbert a,∗
a
Vaccine Technologies Unit, Fraunhofer Institute for Cell Therapy and Immunology, Perlickstrasse 1, D-04103 Leipzig, Germany
b
Department of Molecular and Medical Virology, Ruhr-University, Universitätsstrasse 150, 44801 Bochum, Germany
c
Institute of Animal Science, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
d
Institute of Virology, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany

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

Article history: West Nile Virus (WNV) is an emerging pathogenic flavivirus with increasing distribution worldwide. Birds
Received 8 December 2010 are the natural host of the virus, but also mammals, including humans, can be infected. In some cases,
Received in revised form 18 April 2011 a WNV infection can be associated with severe neurological symptoms. All currently available WNV
Accepted 29 April 2011
vaccines are in the veterinary sector, and there is a need to develop safe and effective immunization
Available online 17 May 2011
technologies, which can also be used in humans. An alternative to current vaccination methods is DNA
immunization. Most current DNA vaccine candidates against flaviviruses simultaneously express the
Keywords:
viral envelope (E) and membrane (prM) proteins, which leads to the formation of virus-like particles.
DNA vaccine
West Nile Virus
Here we generated a DNA plasmid, which expresses only the E-protein ectodomain. Vaccination of mice
Heterologous prime boost stimulated anti-WNV T-cell responses and neutralizing antibodies that were higher than those obtained
after immunizing with a recombinant protein previously shown to be a protective WNV vaccine. A single
dose of the plasmid was sufficient to protect animals from a lethal challenge with the virus. Moreover,
immunogenicity could be boosted when DNA injection was followed by immunization with recombinant
domain DIII of the E-protein. This resulted in significantly enhanced neutralizing antibody titers and a
more prominent cellular immune response. The results suggest that the WNV E-protein is sufficient
as a protective antigen in DNA vaccines and that protection can be significantly improved by adding a
recombinant protein boost to the DNA prime.
© 2011 Elsevier Ltd. All rights reserved.

1. Introduction affecting European countries such as Hungary, Italy and


Romania [4–6].
West Nile Virus (WNV) belongs to the genus Flavivirus, which In mammals, only about 20% of the infected individuals develop
includes several medically important pathogens such as Japanese disease symptoms upon an infection with WNV, indicating effective
Encephalitis Virus (JEV), Dengue Virus (DENV), Yellow Fever Virus immune responses against the virus in most cases.
(YFV) or the Tick-Borne Encephalitis Virus (TBEV). WNV is a Studies performed in mouse models have demonstrated that the
zoonotic virus, which is transmitted by mosquitoes and mainly mammalian innate and adaptive immune systems are able to con-
infects birds. However, mosquitoes can also transmit WNV to trol a WNV infection. Inhibiting the class 1 interferon system, as
mammals such as humans or horses. Particularly older or immune- well as antibody- or T-cell-responses leads to increased suscepti-
suppressed individuals are at risk for severe neurological symptoms bility to WNV (recently reviewed in [7,8]).
caused by WNV, such as encephalitis, meningitis or flaccid paraly- Several veterinary WNV vaccines have been commercialized
sis, which might even be fatal [1]. to date, including inactivated virus or viral vector systems [9,10].
WNV is one of the most widespread flaviviruses, and In contrast, no vaccine is yet available to protect humans. WNV
over the last years outbreaks have frequently been observed affects older and immune-compromised people most severely;
in Africa, Asia, Australia and, since 1999, America [2,3]. In therefore live vaccines are not optimal due to potential side effects.
Europe, severe WNV cases have increased in recent years, Innovative techniques for inactive subunit vaccines are under
development, amongst them are recombinant proteins or DNA vac-
cines [11–13].
∗ Corresponding author. Tel.: +49 341 35536 2106; fax: +49 341 35536 82106. DNA vaccination has become a well-established technique over
E-mail address: Sebastian.ulbert@izi.fraunhofer.de (S. Ulbert). the last two decades and is an alternative to existing immunization

0264-410X/$ – see front matter © 2011 Elsevier Ltd. All rights reserved.
doi:10.1016/j.vaccine.2011.04.116
A. Schneeweiss et al. / Vaccine 29 (2011) 6352–6357 6353

approaches, especially due to the development of novel delivery GGAATGAG (TPA sequence is underlined) and 3 fullE:
techniques such as in vivo electroporation [14]. A DNA vaccine GCGGGCCCTAGTCAGCGTGCACGTTCACGG or 3 ectoE: GCCT-
against WNV for horses was licensed in the USA. This vaccine, sim- GCAGCTAAGACTTGTGCCAATGGTGATTG. The resulting PCR
ilar to most WNV DNA vaccines reported in the literature, is based fragments were inserted into the backbone vector via BamHI
on the co-expression of the viral envelope (E) and membrane (prM) and PstI restriction sites, resulting in pT-WNV-FL (full length E
proteins [15–17]. The E protein has functions in several aspects of with TPA) and pT-WNV-E (E ectodomain with TPA). pCBWN was
the viral life cycle, and prM stabilizes the conformation of E during kindly provided by Michael Diamond (Washington University,
virion assembly. Both proteins are targets for neutralizing anti- USA).
bodies (NA) [18,19]. The E protein consists of a large ectodomain DNA was purified by endotoxin-free GigaPrep Kit
and a small hydrophobic C-terminal transmembrane region. Within (Sigma–Aldrich, Hamburg, Germany) according to the man-
the ectodomain, domains DI, DII and DIII function in virion assem- ufacturer’s instructions. DNA concentration and purity were
bly, membrane fusion and receptor binding, respectively. NAs are determined by UV spectroscopy. HeLa cells were transiently trans-
directed against all three domains, but DIII elicits the strongest neu- fected using Lipofectamine 2000 (Invitrogen, Karlsruhe, Germany)
tralizing responses [19,20]. The co-expression of flaviviral prM/E according to the manufacturer’s instructions. Cell culture super-
leads to the formation of virus-like particles (VLPs). It is believed natants were collected and cells were lysed with 0.1% SDS Tris
that these structures stabilize the proteins and are critical for opti- 24 h post transfection. The cell lysates and supernatants were
mal presentation of the antigens to the immune system [15,21]. analyzed by SDS PAGE and western blotting using a polyclonal
This is supported by results showing that DNA vaccines exclusively rabbit antiserum raised against the peptide GEAHNDKRADPAFVC
encoding the E protein, or its domain DIII, have until now conferred from the WNV E protein.
only partial protection to mice [21–23]. On the other hand, several
studies have demonstrated that in recombinant form the E protein 2.3. Expression and purification of recombinant WNV proteins
(in the absence of prM) or DIII can be used successfully to protect
mammals against WNV [12,13,24]. Therefore, it remains to be clar- The sequence encoding the domain DIII of the WNV E pro-
ified for DNA vaccines whether the WNV E protein can be used as tein was PCR amplified from pT-WNV-E and cloned via BamHI and
a protective antigen without prM. XhoI restriction sites into the bacterial expression vector pMAL-
In this study, a DNA plasmid coding for the ectodomain of the c2X (New England Biolabs, Ipswich, USA), which was modified to
WNV E protein was generated. Mice vaccinated with a single dose include a C-terminal 6× His tag. The resulting coding sequences
of this DNA are protected against a lethal challenge with the virus. contained an N-terminal maltose binding protein (MBP) and a C-
The plasmid elicits neutralizing antibodies, which can be enhanced terminal His tag. The fusion-protein was expressed in the E. coli
by a recombinant protein boost. Rosetta DE3 strain (Merck, Darmstadt, Germany) by induction
with 1 mM isopropyl-␤-d-thiogalactopyranoside. Bacteria were
harvested and lysed in buffer I (20 mM Tris pH 7.4, 200 mM NaCl,
2. Materials and methods
1 mM EDTA) by repeated sonification and freezing in liquid nitro-
gen. Insoluble cell debris containing the WNV fusion-protein was
2.1. Cells, viruses and materials
collected by centrifugation, resuspended in lysis buffer II (20 mM
Tris pH 7.4, 200 mM NaCl, 1 mM EDTA, 8 M Urea) and sonicated.
HeLa cells were grown in DMEM supplemented with 10% heat-
After centrifugation the recombinant WNV fusion-protein was
inactivated fetal calf serum (FCS) and 1% antibiotics. Vero V76 (from
purified from the supernatant on HisTrap HP columns (GE Health-
vervet monkey) cells were grown in DMEM supplemented with 5%
care Bio-Sciences, Munich, Germany) by liquid chromatography.
FCS and 1% antibiotics. Media und supplements were from Invitro-
Thereafter, the MBP-fusion protein was purified on an amylose
gen (Karlsruhe, Germany).
resin column (New England Biolabs). Subsequently the MBP-tag
WNV strains NY 2000, which is WNV 2000-crow3356 (Bern-
was removed by cleaving the fusion-protein with Factor Xa (New
hard Nocht Institute, Hamburg, Germany), and IS-98-ST1 (Pasteur
England Biolabs) and the resulting His-tagged protein was isolated
Institute, Paris, France) were propagated on VeroV76 cells. Cell
by a second His-tag purification as above and dialyzed against PBS.
culture supernatant was harvested and virus was purified by ultra-
centrifugation. Virus pellets were diluted in phosphate-buffered
2.4. Mouse immunizations and WNV challenge studies
saline (PBS) and titrated on Vero V76 cells in 96 well culture plates.
Titers are expressed as tissue culture infective dose 50 (TCID50 ).
Vaccination studies were carried out in groups of five female
Balb/c mice (obtained from Sociéte Janvier, Le Genest-St-Isle,
2.2. Construction of a WNV DNA vaccine candidate France), 6–8 weeks old. Experiments were performed after
approval of local authorities and according to the guidelines
The pVAX1 vector (Invitrogen) was used as backbone. To of the Ethics Committees for Animal Studies of Germany and
increase gene expression levels, two SV40 enhancer elements Switzerland. For DNA vaccination mice were narcotised with
(72 bp each) [25] were integrated into a unique MluI restriction Ketamin/Xylazin (5 mg/kg Xylazin and100 mg/kg Ketamin; Bayer,
site upstream of the CMV promoter. The genomic sequence Leverkusen, Germany). 25 ␮l of DNA (at 1 mg/ml) in 0.9% sodium
for the WNV NY E -protein (accession number AF346318) was chloride (w/v) were injected intramuscularly (i.m.) into the quadri-
amplified by RT-PCR on genomic RNA from WNV and cloned via ceps muscle (29-gauge needle). Injections were immediately
BamHI and ApaI restriction sites into the backbone vector, yielding followed by electroporation with the electric pulse generator Clin-
pWNV-FL. In addition, the sequence coding for the C-terminal ivet (IGEA, Modena, Italy). A four-electrode array was placed at
amino acids G401-A501 was deleted, yielding only the ectodomain the injection site and two electric pulses with 450 V (50 ␮s) fol-
of the protein (pWNV-E). A secretory signal sequence from the lowed by eight low volt pulses (110 V, 20 ms, 20 ms interval) were
murine tissue plasminogen activator (TPA) was added by PCR applied. Alternatively, one group of mice received pT-WNV-E sub-
in-frame at the N-terminus of the full length E-Protein and E cutaneously (s.c.) without electroporation.
ectodomain, using the primers TPA-sense: CGGGATCCACCAT- For protein immunizations 20 ␮g of recombinant protein were
GAAGAGAGAGCTGCTGTGTGTACTGCTGCTTTGTGGACTGGCTTTCC- injected intraperitoneally (i.p.) together with 20 ␮g ODN 1826
CATTGCCTGACCAGGGAATACATGGGAGGTTCGCATTCAACTGCCTT (Eurofins, Ebersberg, Germany), a CpG containing oligonucleotide,
6354 A. Schneeweiss et al. / Vaccine 29 (2011) 6352–6357

as adjuvant. In a BSL3 animal facility, mice were challenged intra-


venously 6 weeks after the first immunization with a lethal dose
(2 × 107 TCID50 ) of the WNV strain IS-98-ST1 (E-protein sequence
100% identical in amino acids to T-WNV-E) and monitored for sur-
vival, body weight and clinical symptoms for 15 days.

2.5. Murine IFN- ELISpot

The ELISpot test for detection of murine IFN-␥ was carried out
according to the manufacturer’s instructions (Mabtech, Hamburg,
Germany). Two weeks after the last immunizations, splenocytes
were isolated from the mice and stimulated with 1 ␮g of recom-
binant ectodomain of WNV E (insect cell derived and kindly
provided by Michael Diamond). IFN-␥ activity was measured using
an ELISpot Reader (AID Diagnostika, Straßberg, Germany).

2.6. Virus neutralisation test and antibody analysis

Vero V76 cells were seeded into 96-well plates at a density of


7000 cells in 100 ␮l per well 1 day before start of the test. For
the virus neutralisation test, heat inactivated (30 min at 56 ◦ C) sera
from immunized mice were serially diluted in culture medium. Two
hundred TCID50 of WNV NY were added to each serum dilution, the
mixtures were incubated for 1 h at 37 ◦ C and added to the cells (in
triplicates). After 60 min culture fluids were replaced by fresh Vero
V76 medium (5% FCS) and cells were propagated for additional 5
days. Titers of neutralising antibodies were determined by monitor-
ing of the cytopathic effect (CPE). Neutralising activity was reported
until two out of three wells of infected cells showed no CPE [26].
To investigate IgG production upon vaccination, sera from
immunized mice were pooled, diluted 1:100 and analyzed in an
enzyme-linked immunosorbent assay (ELISA) using rDIII as antigen
(4 ng per well of a microtiter plate), following standard protocols.
Bound IgG were detected with a rabbit anti-mouse antibody conju-
gated with horseradish peroxidase diluted 1:1000 (Dako, Hamburg,
Germany). Mean values of the optical density at 450 nm (OD450) of
triplicate measurements were determined. Correct conformation of
rDIII was analyzed in an ELISA with the monoclonal antibody E16
(kindly provided by Michael Diamond) in a 1:1000 dilution. Fig. 1. (A) HeLa cells were transfected with plasmids coding for the proteins men-
tioned below and analyzed for expression of the WNV E-protein 24 h later. The
3. Results top panel shows cell lysates, and the bottom panel displays cell culture super-
natants. Lanes: 1, full length E-protein (pWNV-FL); 2, full length E-protein carrying a
N-terminal TPA signal sequence (pT-WNV-FL, specific band marked with an arrow-
3.1. Development of a DNA vaccine head); 3, Ecto-domain of the E-protein (pWNV-E, specific band marked with an
asterisk); 4, Ecto-domain of the E-protein carrying a N-terminal TPA signal sequence
Expression of the full length WNV E protein from plasmid (pT-WNV-E); 5, non-transfected cells. (B) SDS-gel electrophoresis with recombinant
domain DIII. The protein was purified from bacteria using nickel and amylose based
pWNV-FL was tested via in vitro transfection studies in HeLa cells.
affinity chromatography (lane 1); the fusion protein was released from the MBP tag
However, as can be seen in Fig. 1A (lane 1), no clear signal for the via proteolytic cleavage by factor X (lane 2); a second purification of the His contain-
E protein was detectable in a western blot, indicating poor pro- ing proteins separated the MBP (lane 3) from the WNV antigen (lane 4). The correct
duction of the protein. Therefore, the plasmid was modified by size of rDIII is indicated on the right. (C) ELISA using the monoclonal antibody E16 on
deleting the hydrophobic C-terminal domain of E, which might be recombinant DIII protein and recombinant MBP as a negative control. The proteins
were used in the amounts indicated, rDIII after the removal of the MBP tag.
responsible for the weak expression level. The resulting protein is
a truncated version of E which resembles the ectodomain (pWNV-
E). Now, the protein was detectable in the western blot, although
at very low level (lane 3). In addition, a secretory signal sequence,
derived from the murine tissue plasminogen activator (TPA), was However, as the TPA tagged version of the full-length E protein
added to the N-terminus. The resulting plasmid, termed pT-WNV- was absent in the supernatant, these results indicate that the TPA
E, showed a very pronounced increase in expression of the protein, signal sequence mediated the secretion of the E-ectodomain.
as compared to the wild type version (Fig. 1A, lane 4). Also the For control immunization experiments recombinant DIII (rDIII)
full length version of the E-protein was expressed more efficiently was used, as this domain is efficiently expressed in bacteria
when the TPA sequence was added to the N-terminus (pT-WNV-FL; (Fig. 1B), and it was shown previously to be a protective vaccine
lane 2). antigen [12,24]. To test for the correct conformation of the recom-
The TPA-modified version of the E-ectodomain was found in the binant domain DIII after expression, the protein was analyzed with
cell culture supernatant (Fig. 1A, lower panel), visible as a triple the monoclonal antibody E16, which recognizes a structural epi-
band. In contrast, the proteins without the TPA sequences were not tope along the lateral ridge of DIII [27]. There was a clear signal
detectable in the supernatant. It might be possible that the signals detectable in the ELISA (Fig. 1C), indicating the correct conforma-
were under the detection limit due to the low expression levels. tion of the protein.
A. Schneeweiss et al. / Vaccine 29 (2011) 6352–6357 6355

Table 1 with a WNV antigen (E-protein) or a control protein (MBP). Spleen


Vaccination schemes used in this study (groups of 5 mice).
cells derived from mice immunized once with pT-WNV-E showed
Group Nr. Vaccine Day 0 Day 14 Day 28 elevated IFN-␥ activity upon stimulation with the WNV antigen
1 Empty plasmid DNA (Fig. 2). I.m. injection with electroporation seems to be essential
2 pT-WNV-E DNA for this T-cell response, as s.c. injection did not lead to detectable
3 pT-WNV-E 2× DNA DNA IFN-␥. Booster injections with rDIII increased the cellular response
4 pT-WNV-E + 1× rDIII DNA Protein to pT-WNV-E, regardless whether the adjuvant was used or not.
5 pT-WNV-E + 2× rDIII DNA Protein Protein
Immunizing twice with pT-WNV-E led to the highest IFN-␥ produc-
6 pT-WNV-E + 2× rDIII no adj. DNA Protein Protein
7 pT-WNV-E s.c. DNA tion. Also the cellular response to pCBWN increased by subsequent
8 pWNV-E DNA immunizations with rDIII (Fig. 2). These results indicate that boost-
9 pCBWN DNA ing with recombinant protein can enhance the cellular immune
10 pCBWN + 2× rDIII DNA Protein Protein
response to these DNA immunizations. However, boosting with a
11 3× rDIII Protein Protein Protein
second application of pT-WNV-E is most powerful.

3.2. Immunizations 3.4. Antibody production

Table 1 shows the different immunization groups. Balb/c mice The humoral immune response following the immunizations
were immunized with the plasmids pT-WNV-E and pWNV-E was evaluated by analysing the mouse sera for the presence of
(groups 2–7 and 8, respectively). In addition, for comparison with WNV-specific IgG in an ELISA. Specific antibodies were detected
a construct encoding the prM/E expression cassette plus a viral sig- with all three immunization strategies (Table 2). Antibody lev-
nal sequence, the plasmid pCBWN [15] was used (groups 9 and 10). els of animals immunized once with pT-WNV-E were comparably
Combinations of DNA and rDIII were applied as heterologous DNA- low and increased after a second application. Boosting with rDIII
prime and protein-boost strategies (groups 4–6, 9, 10). In group 6, increased the IgG values, both for priming with pT-WNV-E and
the adjuvant was omitted in the protein boost injections. Three rDIII pCBWN (Table 2, groups 2, 3 and 10). The plasmid pWNV-E (group
immunizations (without DNA prime) served as a positive control 8) and the empty DNA backbone vector led to no detectable anti-
(group 11). WNV IgG (group 1), Similar to the T-cell response, electroporation
DNA was injected i.m. followed by in vivo electroporation. To of pT-WNV-E seems to be crucial for antibody production, as no sig-
compare the results with an alternative delivery route, group 7 nals were detected in animals that received the DNA s.c. without
received pT-WNV-E s.c. without electroporation (Table 1). Con- electroporation (group 7).
trol animals (group 1) received one dose of the empty backbone To investigate if the WNV-specific immune response elicited by
plasmid. Injections were performed with 2-weeks intervals, and the different immunization protocols exhibited WNV-neutralising
immune responses were analyzed six weeks after the first applica- activity, the sera were studied in a virus neutralisation assay. Titers
tion. of NA are shown in Table 2. Overall, the results were similar to
the total IgG responses. The highest neutralizing activities were
3.3. IFN- production detected after immunization with the heterologous DNA-prime
protein-boost, when either pT-WNV-E or pCBWN was used as DNA.
Induction of cellular immunity was determined with an ELISpot The addition of the ODN1826 adjuvant did not seem to influence
assay, which measured the IFN-␥ production upon stimulation the outcome of protein boost, as both total IgG and neutralizing

Fig. 2. WNV-specific IFN-␥ levels after immunization. Splenocytes from mice immunized with the antigens indicated were stimulated with cell culture medium (white
columns), recombinant WNV E ectodomain (grey columns) or recombinant MBP (black columns). An ELISpot assay was performed to determine IFN-␥ activity upon
stimulation. Error bars represent the standard deviation.
6356 A. Schneeweiss et al. / Vaccine 29 (2011) 6352–6357

Table 2
Anti-WNV IgG production and neutralizing titers upon immunization with the indicated plasmids and recombinant proteins (mean values of groups of 5 mice). Samples
were taken six weeks after the first immunizations.

Group Nr. Vaccine Anti WNV IgG (OD) (±STDV) WNV neutralizing titer

1 Empty plasmid 0.006 (±0.001) <16


2 pT-WNV-E 0.306 (±0.01) 16
3 pT-WNV-E 2× 0.768 (±0.03) 128
4 pT-WNV-E + 1× rDIII 0.874 (±0.12) 32
5 pT-WNV-E + 2× rDIII 1.799 (±0.03) 256
6 pT-WNV-E + 2× rDIII no adj. 1.804 (±0.06) 512
7 pT-WNV-E s.c. 0.003 (±0.005) <16
8 pWNV-E 0.007 (±0.005) <16
9 pCBWN 0.167 (±0.02) 32
10 pCBWN + 2× rDIII 1.729 (±0.02) 256
11 3× r DIII 1.646 (±0.03) <16

activity in group 6 were comparable to the groups receiving the 4. Discussion


adjuvant (Table 2, group 5). Mice which obtained the empty DNA
vector showed no neutralising activity. Vaccination of rDIII alone In this study the immunization with a DNA vaccine plasmid
did not lead to neutralizing antibodies above the detection limit of (pT-WNV-E) coding for the ectodomain of the WNV E protein is
1:16, although high IgG levels were measured in this group. reported. Fusion of the TPA leader sequence to the ectodomain
leads to a significant increase in expression level compared to
the wildtype version and to the generation of protective immune
responses. In contrast, the plasmid without the TPA sequence
3.5. In vivo protection studies
did not induce measurable anti-WNV IgG or neutralizing activity,
although a minor cellular immune response was observed.
To test the effectiveness of pT-WNV-E as a protective vaccine,
Splenocytes from mice vaccinated with pT-WNV-E produced
viral challenge experiments were performed. Mice were immu-
IFN-␥ upon stimulation with WNV antigen, and the level was
nized either with one dose of pT-WNV-E (Fig. 3, group 1), with one
strongly increased after a second injection of the plasmid. IFN-␥
dose of pT-WNV-E plus two doses of rDIII (group 2), or with three
values were also markedly increased after two booster injections
doses of rDIII (group 3). Negative control mice received one dose
with rDIII, but were still lower as compared to the DNA boost. This
of the empty backbone plasmid (group 4). Six weeks after the first
could be explained by the fact that the ectodomain of E contains
immunization, animals were injected intravenously with a lethal
more T cell epitopes than its domain DIII [28]. In addition, DNA
dose of WNV and their survival was monitored. Mice that received
vaccines are known potent inducers of cellular immune responses,
the negative control DNA all developed severe symptoms between
especially after delivery via electroporation [29].
5 and 7 days post infection (Fig. 3) and were euthanized according
Booster injections with rDIII increased antibody titers in serum
to the termination criteria set. In contrast, all animals immunized
to a higher extend than a second application of pT-WNV-E.
with pT-WNV-E, with rDIII or with the heterologous prime-boost
Together, this indicates that, after a DNA prime, the DNA boost
survived without displaying any measurable signs of disease. These
especially augments cellular immunity, whereas the rDIII boost
data demonstrate that a single vaccination with pT-WNV-E is suf-
strengthens antibody responses. In contrast, NA titers in animals
ficient to protect mice from a lethal WNV infection.
immunized with rDIII were borderline, although the total IgG level
against the antigen was high. This result differs from two previ-
ous studies, which showed that bacterially expressed rDIII leads
to efficient NA production [12,24]. The rDIII protein used here was
of the correct conformation, as it was bound by the monoclonal
antibody E16, which recognizes a structural epitope. Therefore,
technical differences in study design (e.g. injection route, mouse
strain, neutralization assays) might contribute to this discrepancy.
In addition, low-level NA titers after rDIII immunization have been
reported previously [23]. Nevertheless, consistent with the studies
described in [12,24], immunization with rDIII led to protection of
mice from WNV in the work described here.
When challenged with WNV, mice immunized with a single
dose of pT-WNV-E (with or without rDIII boost) did not show
signs of disease. Protection from a lethal WNV-challenge was also
achieved with DNA vaccines containing the prM/E expression cas-
sette, such as the plasmid pCBWN [15], which was included in
the present study. Expression of pCBWN leads to the formation
of VLPs, which forms the basis of most current DNA immunization
approaches against flaviviruses [15–17,30]. The pT-WNV-E plas-
mid does not contain the prM or viral signal sequences present
in the prM/E constructs. Moreover, it does not lead to formation
Fig. 3. In vivo protection study. Mice were vaccinated and subsequently challenged of VLPs (data not shown). However, humoral or cellular responses
with a lethal dose of WNV. Survival rates were determined for 15 days post infection were not significantly different when comparing immunizations
(p.i). Immunizations were performed with one dose of pT-WNV-E (group 1), one with pT-WNV-E and pCBWN. This indicates that co-expression of
dose of pT-WNV-E plus two doses of rDIII (group 2), three doses of rDIII (group 3)
or the empty DNA backbone plasmid (group 4). Control mice remained uninfected
prM and the formation of VLPs is not a prerequisite for induction
(group 5). of an efficient anti-WNV immune response upon DNA vaccina-
A. Schneeweiss et al. / Vaccine 29 (2011) 6352–6357 6357

tion. Therefore, a plasmid, which encodes only the ectodomain of [6] Sirbu A, Ceianu CS, Panculescu-Gatej RI, Vazquez A, Tenorio A, Rebreanu R, et al.
WNV-E, fused to the TPA sequence seems to be a valid alternative Outbreak of West Nile virus infection in humans, Romania, July to October 2010.
Euro Surveill 2011;16(January (2)).
to constructs expressing the proteins prM and E together with a [7] Diamond MS, Mehlhop E, Oliphant T, Samuel MA. The host immunologic
viral signal sequence. This is supported by recent studies on DIII response to West Nile encephalitis virus. Front Biosci 2009;14:3024–34.
based DNA vaccines which were optimized by the addition of DNA [8] Ulbert S, West Nile Virus—the complex biology of an emerging pathogen. Inter-
virology, in press.
encoded protein adjuvants [23,31]. In addition, it was also shown
[9] Ng T, Hathaway D, Jennings N, Champ D, Chiang YW, Chu HJ. Equine vaccine
that the ectodomain of E from JEV is more efficiently expressed for West Nile virus. Dev Biol (Basel) 2003;114:221–7.
after fusion to a TPA signal sequence in the context of vaccination [10] El Garch H, Minke JM, Rehder J, Richard S, Edlund Toulemonde C, Dinic S, et al.
[22]. A West Nile virus (WNV) recombinant canarypox virus vaccine elicits WNV-
specific neutralizing antibodies and cell-mediated immune responses in the
The results presented here indicate synergistic effects when a horse. Vet Immunol Immunopathol 2008;123:230–9.
single immunization with pT-WNV-E is followed by two booster [11] Martin JE, Pierson TC, Hubka S, Rucker S, Gordon IJ, Enama ME, et al. A West
immunizations with rDIII. Mice were equally well protected in the Nile virus DNA vaccine induces neutralizing antibody in healthy adults during
a phase 1 clinical trial. J Infect Dis 2007;196:1732–40.
challenge experiment, but the protein boost strongly enhanced the [12] Martina BE, Koraka P, van den Doel P, van Amerongen G, Rimmelzwaan GF,
immunogenicity of a single pT-WNV-E application. Osterhaus AD. Immunization with West Nile virus envelope domain III protects
The combination of DNA prime and protein boost is reportedly mice against lethal infection with homologous and heterologous virus. Vaccine
2008;26:153–7.
associated with an improvement of cellular and humoral immune [13] Lieberman MM, Nerurkar VR, Luo H, Cropp B, Carrion Jr R, de la Garza M, et al.
responses, although the exact reasons for this synergy still have Immunogenicity and protective efficacy of a recombinant subunit West Nile
to be determined (reviewed in [32]). An inactivated WNV vaccine virus vaccine in rhesus monkeys. Clin Vaccine Immunol 2009;16:1332–7.
[14] Kutzler MA, Weiner DB. DNA vaccines: ready for prime time? Nat Rev Genet
was used to optimize a DNA vaccine based on the pr/M expression
2008;9:776–88.
cassette [17]. However, the use of rDIII as booster component has [15] Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, et al. West Nile
the advantages that the protein is relatively easy to express and virus recombinant DNA vaccine protects mouse and horse from virus challenge
that the vaccine manufacturing does not involve culturing of WNV. and expresses in vitro a noninfectious recombinant antigen that can be used in
enzyme-linked immunosorbent assays. J Virol 2001;75:4040–7.
For vaccine strategies encoding single antigens, such as the plas- [16] Anwar A, Chandrasekaran A, Ng ML, Marques E, August JT. West Nile
mids described here or recombinant proteins [13], care must be premembrane-envelope genetic vaccine encoded as a chimera containing the
taken to monitor emerging viral strains with potential sequence transmembrane and cytoplasmic domains of a lysosome-associated membrane
protein: increased cellular concentration of the transgene product, targeting
variations in order to guarantee protection. On the other hand, to the MHC II compartment, and enhanced neutralizing antibody response.
especially DNA vaccines can rapidly be adapted to novel sequences Virology 2005;332:66–77.
by adapting the nucleotide sequences and by co-injection of several [17] Ishikawa T, Takasaki T, Kurane I, Nukuzuma S, Kondo T, Konishi E. Co-
immunization with West Nile DNA and inactivated vaccines provides
sequence variants. synergistic increases in their immunogenicities in mice. Microbes Infect
In summary, a DNA plasmid encoding a part of the E protein of 2007;9:1089–95.
WNV was generated, which protects animals from WNV. Neutral- [18] Vázquez S, Guzmán MG, Guillen G, Chinea G, Pérez AB, Pupo M, et al.
Immune response to synthetic peptides of dengue prM protein. Vaccine
izing antibody and cellular immune responses to a single injection 2002;20:1823–30.
of this plasmid were strongly increased by subsequent injections [19] Beasley DW, Barrett AD. Identification of neutralizing epitopes within
of recombinant DIII protein. This strategy was clearly superior to structural domain III of the West Nile virus envelope protein. J Virol
2002;76:13097–100.
vaccination with DNA or protein alone. As a vaccine to be used in
[20] Oliphant T, Nybakken GE, Austin SK, Xu Q, Bramson J, Loeb M, et al. Induc-
humans against WNV has to be both, effective and long-lasting, tion of epitope-specific neutralizing antibodies against West Nile virus. J Virol
further studies are warranted using the approach presented. 2007;81:11828–39.
[21] Aberle JH, Aberle SW, Allison SL, Stiasny K, Ecker M, Mandl CW, et al. A
DNA immunization model study with constructs expressing the tick-borne
encephalitis virus envelope protein E in different physical forms. J Immunol
Acknowledgements 1999;163:6756–61.
[22] Ashok MS, Rangarajan PN. Protective efficacy of a plasmid DNA encoding
We thank Diana Riethmüller for excellent technical assistance, Japanese encephalitis virus envelope protein fused to tissue plasminogen acti-
vator signal sequences: studies in a murine intracerebral virus challenge model.
Drs. Michael Szardenings and Gustavo Makert dos Santos for crit- Vaccine 2002;20:1563–70.
ically reading of the manuscript and Dr. Matthias Giese (Institute [23] Dunn MD, Rossi SL, Carter DM, Vogt MR, Mehlhop E, Diamond MS, et al.
for Molecular Vaccines, Heidelberg) for acquisition of funding from Enhancement of anti-DIII antibodies by the C3d derivative P28 results in lower
viral titers and augments protection in mice. Virol J 2010;7:95.
BLE. We also thank Dr. Philippe Despres (Pasteur Institute, Paris)
[24] Chu JH, Chiang CC, Ng ML. Immunization of flavivirus West Nile recombinant
for supplying the WNV strain IS-98-ST1 and Dr. Michael Diamond envelope domain III protein induced specific immune response and protection
(Washington University, St. Louis) for providing the E16 antibody, against West Nile virus infection. J Immunol 2007;178:2699–705.
pCBWN and recombinant ectoE. This work was supported by the [25] Li HS, Liu Y, Li DF, Zhang RR, Tang HL, Zhang YW, et al. Enhancement of DNA
vaccine-induced immune responses by a 72-bp element from SV40 enhancer.
German Federal Agency for Agriculture and Food (BLE) and by the Chin Med J (Engl) 2007;120:496–502.
European Commission under FP7, Project 261426 (WINGS-West [26] Reed LJ, Muench H. A simple method of estimating fifty per cent endpoints. Am
Nile Integrated Shield Project). J Hygiene 1938;27:493–7.
[27] Nybakken GE, Oliphant T, Johnson S, Burke S, Diamond MS, Fremont DH. Struc-
tural basis of West Nile virus neutralization by a therapeutic antibody. Nature
2005;437:764–9.
References [28] Parsons R, Lelic A, Hayes L, Carter A, Marshall L, Evelegh C, et al. The memory
T cell response to West Nile virus in symptomatic humans following natural
[1] Hayes EB, Sejvar JJ, Zaki SR, Lanciotti RS, Bode AV, Campbell GL. Virology, pathol- infection is not influenced by age and is dominated by a restricted set of CD8+
ogy, and clinical manifestations of West Nile virus disease. Emerg Infect Dis T cell epitopes. J Immunol 2008;181:1563–72.
2005;11:1174–9. [29] Otten GR, Schaefer M, Doe B, Liu H, Megede JZ, Donnelly J, et al. Potent
[2] Calistri P, Giovannini A, Hubalek Z, Ionescu A, Monaco F, Savini G, et al. Epi- immunogenicity of an HIV-1 gag-pol fusion DNA vaccine delivered by in vivo
demiology of west Nile in Europe and in the Mediterranean basin. Open Virol electroporation. Vaccine 2006;24:4503–9.
J 2010;4:29–37. [30] Chang GJ, Hunt AR, Davis B. A single intramuscular injection of recombinant
[3] Murray KO, Mertens E, Desprès P. West Nile virus and its emergence in the plasmid DNA induces protective immunity and prevents Japanese encephalitis
United States of America. Vet Res 2010;41:67. in mice. J Virol 2000;74:4244–52.
[4] Krisztalovics K, Ferenczi E, Molnar Z, Csohan A, Ban E, Zoldi V, et al. West Nile [31] Ramanathan MP, Kutzler MA, Kuo YC, Yan J, Liu H, Shah V, et al. Coimmuniza-
virus infections in Hungary. August–September 2008. Euro Surveill 2008;13, tion with an optimized IL15 plasmid adjuvant enhances humoral immunity via
pii:19030. stimulating B cells induced by genetically engineered DNA vaccines expressing
[5] Barzon L, Franchin E, Squarzon L, Lavezzo E, Toppo S, Martello T, et al. Genome consensus JEV and WNV E DIII. Vaccine 2009;27:4370–80.
sequence analysis of the first human West Nile virus isolated in Italy in 2009. [32] Lu S. Heterologous prime-boost vaccination. Curr Opin Immunol
Euro Surveill 2009;14, pii:19384. 2009;21:346–51.

You might also like