You are on page 1of 9

JOURNAL OF VIROLOGY, July 2008, p. 6200–6208 Vol. 82, No.

13
0022-538X/08/$08.00⫹0 doi:10.1128/JVI.00187-08
Copyright © 2008, American Society for Microbiology. All Rights Reserved.

Comparative Efficacy of Neutralizing Antibodies Elicited by


Recombinant Hemagglutinin Proteins from Avian
H5N1 Influenza Virus䌤
Chih-Jen Wei,1† Ling Xu,1† Wing-Pui Kong,1 Wei Shi,1 Kevin Canis,2 James Stevens,3‡
Zhi-Yong Yang,1 Anne Dell,2 Stuart M. Haslam,2 Ian A. Wilson,3 and Gary J. Nabel1*
Vaccine Research Center, NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC-3005, 40 Convent Drive, Bethesda,
Maryland 20892-30051; Division of Molecular Biosciences, Faculty of Natural Sciences, Imperial College London,
London SW7 2AZ, United Kingdom2; and Department of Molecular Biology & Skaggs Institute for
Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road,
BCC206, La Jolla, California 920373
Received 25 January 2008/Accepted 1 April 2008

Although the human transmission of avian H5N1 virus remains low, the prevalence of this highly pathogenic
infection in avian species underscores the need for a preventive vaccine that can be made without eggs. Here,
we systematically analyze various forms of recombinant hemagglutinin (HA) protein for their potential efficacy
as vaccines. Monomeric, trimeric, and oligomeric H5N1 HA proteins were expressed and purified from either

Downloaded from jvi.asm.org by on January 8, 2009


insect or mammalian cells. The immunogenicity of different recombinant HA proteins was evaluated by
measuring the neutralizing antibody response. Neutralizing antibodies to H5N1 HA were readily generated in
mice immunized with the recombinant HA proteins, but they varied in potency depending on their multimeric
nature and cell source. Among the HA proteins, a high-molecular-weight oligomer elicited the strongest
antibody response, followed by the trimer; the monomer showed minimal efficacy. The coexpression of another
viral surface protein, neuraminidase, did not affect the immunogenicity of the HA oligomer, as expected from
the immunogenicity of trimers produced from insect cells. As anticipated, HA expressed in mammalian cells
without NA retained the terminal sialic acid residues and failed to bind ␣2,3-linked sialic acid receptors. Taken
together, these results suggest that recombinant HA proteins as individual or oligomeric trimers can elicit
potent neutralizing antibody responses to avian H5N1 influenza viruses.

Since 1889, at least five influenza virus pandemics have oc- of H5N1 avian influenza virus are strongly associated with
curred, the most catastrophic of which was the Spanish influ- exposure to infected domestic fowl (21).
enza of 1918, which resulted in 20 to 50 million deaths world- Effective vaccination is a critical tool that supports public
wide (4, 8). Today, an average of about 200,000 influenza health efforts to reduce influenza virus morbidity and mortal-
virus-related hospitalizations and about 36,000 influenza virus- ity. Each year, the World Health Organization selects three
related deaths occur in a typical winter-seasonal epidemic in influenza virus strains as targets for inactivated vaccine devel-
the United States (14). First appearing in 1997, the highly opment. While the trivalent inactivated influenza virus vac-
pathogenic avian influenza H5N1 virus continues to spread cines currently used in the United States are manufactured
globally (19). The current global outbreak of H5N1 avian in- using embryonated eggs, it will be difficult to rapidly scale up
fluenza virus among domestic and wild birds, and its potential this technology for the mass production of vaccine in the event
adaptation to humans, has accelerated influenza H5N1 virus of a potential pandemic (18). Recently, a new cell culture-
research and pandemic preparedness. More than 300 cases of based approach for influenza virus vaccine development, in-
human H5N1 influenza virus infection had been confirmed. Of volving the production of influenza virus in cell culture fol-
these cases, nearly 200 individuals have died as a consequence lowed by virus inactivation and purification, has been proposed
of infection (22). Although a few instances of human-to-human and tested (1). While offering advantages over egg-based ap-
H5N1 influenza virus transmission have been documented, the proaches, e.g., cell culture technology can be scaled up in
current H5N1 virus has not yet acquired the ability to spread shorter periods of time, cell culture-based approaches for
efficiently within the human population, and most human cases H5N1 manufacture still require the production of a potentially
hazardous virus (1).
It has been demonstrated that protection provided by the
trivalent influenza virus vaccine is mediated primarily by anti-
* Corresponding author. Mailing address: Vaccine Research Center,
NIAID, National Institutes of Health, Bldg. 40, Room 4502, MSC- hemagglutinin (HA) neutralizing antibodies. Thus, a recombi-
3005, 40 Convent Drive, Bethesda, MD 20892-3005. Phone: (301) nant protein-based approach utilizing purified HA proteins
496-1852. Fax: (301) 480-0274. E-mail: gnabel@nih.gov. expressed in different mammalian systems offers another alter-
† These authors contributed equally to this work. native for influenza virus vaccine development. This platform
‡ Present address: Molecular Virology and Vaccines Branch, Influ-
enza Division, NCIRD, CCID, Centers for Disease Control and Pre-
provides advantages over current approaches, including well-
vention, 1600 Clifton Rd., Mail stop G-16, Atlanta, GA 30333. described technologies for mass production and reduced bio-

Published ahead of print on 16 April 2008. hazards during manufacturing. Various prototypes produced

6200
VOL. 82, 2008 INFLUENZA VACCINATION WITH RECOMBINANT HA PROTEINS 6201

in a baculovirus-insect cell expression system have proven safe reader (Wyatt Technology, Santa Barbara, CA). Additionally, mammalian cell-
and effective in clinical studies for both H1N1 and H3N2 expressed HAs were produced by the cotransfection of a 1/10 ratio (wt/wt) of
NA(KAN-1) or NA(New Caledonia/99) expression vector for the glycan array
influenza viruses (7, 10, 11, 15–17). In this study, we systemat- analysis, the mass spectrometry (MS) analysis of HA N-glycan composition, or
ically tested various recombinant HA proteins as alternatives the NA-coexpressed HA proteins that also were used for immunization. The
to egg-based vaccine candidates against influenza virus infec- molecular weights of the HA oligomer, trimer, or monomer proteins were de-
tion. H5N1 HA proteins were expressed and purified from termined by density gradient sedimentation as previously described (9).
Vaccination. Female BALB/c mice (6 to 8 weeks old; Jackson Laboratories)
either insect or mammalian cells. The immunogenicity of dif-
were immunized intramuscularly with 20 ␮g of inactivated influenza virus sub-
ferent recombinant HA proteins was evaluated by antibody virion vaccine [rgA/Vietnam/1203/2004 (H5N1); Biodefense and Emerging In-
neutralization. The data suggest that stable, trimeric viral fections Research Resources Repository, NIAID, NIH) or 20 ␮g of purified
spikes serve as the optimal protein immunogens to elicit neu- protein in 50 ␮l of phosphate-buffered saline (PBS) (pH 7.4) and mixed with 50
tralizing antibodies against H5N1 isolates, an approach that ␮l of Ribi adjuvant (Sigma, St. Louis, MO) in PBS, pH 7.4, as recommended, at
weeks 0 and 3. Blood was collected 14 days after each immunization, and serum
may be applicable to seasonal influenza and other viruses. was isolated. Animal experiments were conducted in full compliance with all
relevant federal regulations and NIH guidelines.
ELISA and isotyping of anti-HA antibodies. The mouse anti-HA immuno-
MATERIALS AND METHODS globulin G (IgG) and IgM enzyme-linked immunosorbent assay (ELISA) titers
Genes and expression vectors. Based on H3 numbering (20), a cDNA corre- were measured by a previously described method (23). Purified trimeric HA was
sponding to residues 11 to 500 of the HA from A/Thailand/KAN-1/2004 (KAN-1; used to coat the plate, and anti-HA antibodies were detected by peroxidase-
GenBank accession no. AAS65615) was synthesized using human-preferred conjugated goat anti-mouse IgG and IgM antibody (Jackson ImmunoResearch,
codons as described previously (6) by using GeneArt (Regensburg, Germany). West Grove, PA). The subclasses of anti-HA antibodies also were determined by
This construct terminates at the bromelain cleavage site (12). Alternatively, the ELISA using antibodies to IgA, IgG1, IgG2a, IgG2b, IgG3, and IgM (Calbio-
14 amino acids (EISGVKLESIGIYQ) between the bromelain cleavage site and chem, Gibbstown, NJ).
the transmembrane domain of HA were inserted to produce the ⌬TM construct. Production of pseudotyped lentiviral vectors and measurement of neutralizing
The original viral protease cleavage site PQRERRRKKRG was changed to antibodies. The recombinant lentiviral vectors expressing a luciferase reporter

Downloaded from jvi.asm.org by on January 8, 2009


PQRETRG in order to retain the uncleaved and unprocessed proteins. The gene were produced as previously described (6, 24).
purified protein contains additional residues at the C terminus (ISGRLVPRGS Glycan array analysis. The glycan microarray analysis of the HA proteins was
PGSGYIPEAPRDGQAYVRKDGEWVLLSTFLGHHHHHH), in which the performed as previously described (24).
thrombin cleavage site is in italics, the bacteriophage T4 fibritin foldon trimer- MS of HA N-glycans. The HA N-glycans were prepared for MS analysis as
ization sequence is underlined, and the His tag is in boldface (12). The inserts previously described (5). Briefly, purified HA glycoproteins were reduced, car-
were cloned into the cytomegalovirus/human T-cell leukemia virus type 1 repet- boxymethylated, and digested with L-1-tosylamido-2-phenylmethyl chloromethyl
itive sequence (CMV/R) 8␬B expression vector for efficient expression in mam- ketone (TPCK) bovine pancreas trypsin (EC 3.4.21.4). The N-glycans were
malian cells (6) or into the baculovirus transfer vector pAcGP67A (BD Bio- enzymatically released by digestion with PNGase F (EC 3.5.1.52; Roche Molec-
sciences, Bedford, MA). Genes for NA(KAN-1)(H5N1) and NA(New ular Biochemicals) and purified by reverse-phase C18 Sep-Pak (Waters Corp.)
Caledonia/99)(H1N1) (GenBank accession nos. AY555150 and AJ518092, chromatography. Prior to MS analyses, the released N-glycans were permethyl-
respectively) also were synthesized using human-preferred codons (GeneArt, ated and purified using a reverse-phase C18 Sep-Pak (Waters Corp.). Matrix-
Regensburg, Germany) and were cloned into the expression vector CMV/R 8␬B. assisted laser desorption ionization–time of flight (MALDI-TOF) data were
Baculovirus production. HA proteins were produced by the cotransfection of acquired on a Voyager-DE STR mass spectrometer (PerSeptive Biosystems,
baculovirus transfer vector with BaculoGold-linearized baculovirus DNA (BD Framingham, MA) in the reflectron mode with delayed extraction. Permethyl-
Biosciences, Bedford, MA) into Spodoptera frugiperda (Sf9) cells (Invitrogen, ated samples were dissolved in 10 ␮l of 80% (vol/vol) methanol in water, and 1
Carlsbad, CA) using the BaculoGold transfection buffer set (BD Biosciences, ␮l of dissolved sample was premixed with 1 ␮l of matrix (10 mg/ml 2,5-dihy-
Bedford, MA) and subsequently was amplified in the same cells according to the droxybenzoic acid [DHB] in 80% [vol/vol] aqueous methanol) before being
manufacturer’s instructions. loaded onto a metal plate. MALDI-TOF/TOF experiments were performed on
Protein expression and purification. Plasmids expressing a secreted HA were a 4800 Proteomics Analyzer (Applied Biosystems, Framingham, MA) operated
transfected into the human embryonic kidney cell line 293F using 293fectin in the reflectron positive ion mode.
(Invitrogen, Carlsbad, CA) according to the manufacturer’s instructions. 293F
cells were cultured in Freestyle 293 expression medium (Invitrogen, Carlsbad,
CA), and supernatant was collected 72 to 96 h posttransfection and cleared by RESULTS
centrifugation and filtration. HA proteins were purified as previously described
(6), with minor modifications. Briefly, HA was recovered from the cell superna- Expression, purification, and characterization of HA pro-
tant by metal affinity chromatography using Ni Sepharose high-performance teins. To evaluate the efficacy of recombinant HA proteins as
resin (GE Healthcare, Piscataway, NJ). Fractions containing HA were combined
potential vaccine candidates, cDNAs encoding the ectodomain
and subjected to ion-exchange chromatography using a MonoQ HR10/10 column
(GE Healthcare, Piscataway, NJ). HA oligomers, trimers, and monomers then of HA (A/Thailand/KAN-1/2004) were cloned into a baculo-
were separated by gel filtration chromatography using a Hi-Load 16/60 Superdex virus transfer vector, pAcGP67A, or a mammalian expres-
200-pg column (GE Healthcare, Piscataway, NJ). To remove the foldon se- sion vector, CMV/R 8␬B (6), to allow the efficient secretion
quence and His tag, HA proteins were subjected to thrombin digestion (EMD of HA proteins (Fig. 1A). The multibasic protease cleavage
Chemicals, Inc., San Diego, CA) at 3 U/mg at 4°C overnight. Insect-expressed
HA proteins (KAN-1) were purified as previously described (12). Trichoplusia ni
site PQRERRRKKRG between HA1 and HA2 was mutated
(Hi5) cells were infected at a multiplicity of infection of 10 and cultured in to PQRETRG to reduce the efficiency of processing. To
Express Five serum-free medium (Invitrogen, Carlsbad, CA). The cell culture stabilize the trimeric conformation of HA proteins, a bac-
was maintained at 27°C with gentle shaking. The culture suspension was col- teriophage-trimerizing foldon sequence was engineered into
lected 96 h after infection, and the HA proteins were purified using the same
the constructs, and a His tag was introduced at the COOH
method as that described for mammalian cell-expressed proteins, except that,
after using the MonoQ column, HA protein was left overnight at 4°C to precip- terminus for purification purposes (12). A thrombin cleav-
itate ferritin (12). HA protein from A/Vietnam/1203/2004 (VN1203) was purified age site was inserted between the HA and foldon sequence
as previously described (12). The expression of the HA proteins was confirmed to ensure the cleavage of the foldon and His tag, if neces-
by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and sary. After the generation of baculovirus vectors expressing
Western blotting using a mouse monoclonal anti-Penta His antibody (Qiagen,
Hilden, Germany), mouse monoclonal anti-HA antibody 10D10 (24), or a rabbit
HA proteins, the expression of HA proteins was carried out
polyclonal anti-HA antibody (Immune Technology, New York, NY). Protein by either infecting Hi5 cells or transfecting 293F cells with
purity also was examined by dynamic light scattering using a DynaPro plate mammalian expression vectors. The expression of secreted
6202 WEI ET AL. J. VIROL.

Downloaded from jvi.asm.org by on January 8, 2009


FIG. 1. H5 HA expression vectors, biochemical purification, and characterization of insect- and mammalian cell-expressed HA proteins.
(A) Vectors encoding H5 HA (A/Thailand/KAN-1/2004) with a mammalian codon preference were prepared in the mammalian expression vector
CMV/R 8␬B or the baculovirus transfer vector pAcGP67A. The cleavage site between HA1 and HA2 was mutated in order to obtain full-length
proteins. The clones were engineered to contain a trimeric foldon sequence and hexa-His tag (see Materials and Methods) at the COOH terminus,
which could be removed by thrombin digestion. These vectors were introduced into the 293F renal epithelial cells or Hi5 cells, as described in
Materials and Methods, to generate recombinant protein that was purified in monomeric, trimeric, or oligomeric forms for immunization studies.
(B) The HA proteins purified from insect or mammalian cells existed as a high-molecular-mass oligomer (⬎700 kDa) and trimer (uncleaved trimer;
⬎200 kDa) after gel filtration (red line). After thrombin digestion, HA protein eluted as a trimer (cleaved trimer) and a small fraction of monomer
(cleaved monomer) also could be detected (black line). The figures present superimposed elution profiles of insect- or mammalian cell-expressed
HA proteins overlaid with calibration standards (blue lines). (C) Insect or mammalian cell-derived proteins purified in the oligomer, trimer, and
cleaved trimer forms were analyzed by SDS-PAGE (left). Uncleaved trimer and cleaved trimer also were analyzed by Western blotting using a
polyclonal anti-HA antibody (middle), and the removal of the foldon domain and His tag from the trimer was confirmed by the decrease in the
size of the HA band and by using an anti-His tag antibody (right). HA-F, uncleaved HA with a foldon trimerization domain and His tag.

proteins was first confirmed by Western blotting using anti- pressed HA also was confirmed by SDS-PAGE (Fig. 1C) and
His tag or anti-HA antibodies. The secreted HA proteins by Western blot analysis using an antibody against HA, and
then were purified using a nickel affinity and MonoQ anion- this revealed a slightly lower molecular size after the cleav-
exchange column, followed by using a Superdex200 gel fil- age of the trimerization motif and His tag (Fig. 1C). Its
tration column to separate HA oligomer, trimer, and mono- removal was confirmed by Western blotting using an anti-
mer. In Hi5 cells, HA was expressed as two major species, a His tag antibody (Fig. 1C). The mammalian cell-expressed
high-molecular-weight oligomer and an uncleaved trimer HA also appeared as a high-molecular-mass oligomer and
(Fig. 1B). The molecular sizes of insect-expressed HA oligo- trimer (Fig. 1B) with molecular masses of 1,394 and 222
mer and trimer were estimated to be 1,321 and 214 kDa, kDa, respectively, as determined by density gradient sedi-
respectively, as determined by density gradient sedimenta- mentation. In contrast to the insect-produced protein, the
tion. When the foldon sequence was removed by thrombin peak size of the trimer showed a higher molecular mass due
digestion, the majority of the HA proteins appeared as a to the more extensive glycosylation in mammalian cells. In
cleaved trimer, and a small fraction of cleaved monomer addition, unlike its insect-expressed counterpart, the high-
also was present (Fig. 1B). The expression of insect-ex- molecular-mass oligomer remained intact after thrombin
VOL. 82, 2008 INFLUENZA VACCINATION WITH RECOMBINANT HA PROTEINS 6203

Downloaded from jvi.asm.org by on January 8, 2009


FIG. 2. Ability of the trimeric HAs from mammalian or insect cells to elicit neutralizing antibodies: higher neutralizing antibody responses were
elicited by oligomers than by trimers, and the titers increased with a repeat immunization. (A) The neutralization by antisera from five mice
immunized with insect-expressed HAs prepared from insect cells was assessed by the incubation of mouse sera with HA/NA-pseudotyped lentiviral
reporter vectors encoding luciferase. The percent neutralization was calculated by the reduction of luciferase activity relative to the values achieved
in the absence of sera. Among the insect cell-expressed proteins, oligomer, uncleaved trimer, and cleaved trimer from the VN1203 strain all elicited
potent neutralizing antibodies against KAN-1 HA/NA pseudovirus, while the cleaved trimer induced only modest neutralizing activity. (B) Mam-
malian high-molecular-mass oligomers induced the highest titer of neutralizing antibody response, followed by cleaved trimer, uncleaved trimer,
and cleaved monomer. Statistically significant differences were observed between oligomer and trimer (P ⬍ 0.0001), oligomer and cleaved trimer
(P ⬍ 0.0001), and oligomer and cleaved monomer (P ⬍ 0.0001) forms. (C) Mice received a single injection of mammalian cell-expressed oligomer
or a secondary boost, as indicated, 3 weeks after the initial injection. The neutralizing antibody responses were measured in serum samples
collected 14 days after each vaccination using the HA/NA-pseudotyped lentivirus reporter assay. All animals were immunized with 20 ␮g/injection
of HA protein in 50 ␮l PBS and an equal volume of Ribi as the adjuvant. (D) Total HA antibodies in mice immunized with insect- or mammalian
cell-expressed HA proteins were measured by ELISA. (E) Antisera from mice immunized with insect- or mammalian cell-expressed HA proteins
were characterized for IgG1, IgG2a, and IgM responses. Similarly to mice immunized with inactivated H5N1 subvirion vaccine (inact. H5 vaccine),
both insect- and mammalian cell-expressed oligomeric and trimeric HA induced the highest levels of IgG2a, followed by IgG1. Naı̈ve mouse serum
was used as a negative control. IgG3 and IgA also were tested but were negative for all sera (not shown).

cleavage, although trimeric and monomeric species were pseudotyped, lentiviral vector assay (24). To analyze the ability
detected (Fig. 1B). The expression of HA proteins of the of insect-expressed HA to induce neutralizing antibodies, HA/
expected size also was confirmed by SDS-PAGE and West- NA-pseudotyped reporters were incubated with antisera from
ern blot analysis (Fig. 1C). For subsequent immunogenicity immunized mice, and the neutralizing antibody activity was
studies, only the peak fractions of each species were col- measured using a luciferase assay. Sera from animals immu-
lected in 2-ml aliquots. An analysis of these fractions by nized with insect-expressed oligomer and uncleaved trimer in-
dynamic light scattering confirmed that each immunogen hibited pseudovirus entry effectively, indicating the presence of
was of ⬎97% homogeneity (data not shown). neutralizing antibodies to H5 HA (Fig. 2A). The thrombin-
Humoral immune responses elicited by insect-expressed re- digested trimer elicited only a modest level of neutralizing
combinant HAs. Humoral immunity in mice was evaluated by antibody against KAN-1 HA (Fig. 2A), although a cleaved
vaccination with different forms of HA expressed in insect trimer from a closely related strain (VN1203) elicited levels
cells. Mice were immunized with 20 ␮g of oligomers, trimers, comparable to those observed with uncleaved trimer (Fig. 2A).
or monomers in Ribi adjuvant twice at an interval of 3 weeks. These differences likely were related to the relative stability of
Antisera from the immunized animals were collected 14 days the cleaved HA trimer of these two strains when mixed with
after the second immunization and analyzed for neutralization adjuvant. The total HA antibodies were measured by ELISA
activity using a previously described HA/neuraminidase (NA)- (Fig. 2D) and used to determine the ratio of neutralizing to
6204 WEI ET AL. J. VIROL.

total binding antibodies. Among the insect cell-expressed pro- nary core fucosylated structures. The sialylation of the complex
teins, uncleaved trimer elicited the highest percentage of neu- glycans is a predominant feature, with fully sialylated bi-, tri-,
tralizing antibodies, followed by oligomer and cleaved trimer. and tetra-antennary structures being observed at m/z 2966,
Isotypes of antibodies elicited by insect-expressed oligomer 3776, and 4586, respectively. The spectra derived from the
and trimer were mostly IgG1 and IgG2a and were similar N-glycans of HA coexpressed with NA(KAN-1)(H5N1) again
whether the immunogen was generated in mammalian or showed compositions consistent with those of bi-, tri-, and
insect cells (Fig. 2E). tetra-antennary core fucosylated structures (m/z 1835 to 3503;
Comparison of the immunogenicity of mammalian cell-ex- Hex3HexNAc4Fuc-NeuAc1Hex7HexNAc6Fuc) (Fig. 4). How-
pressed oligomers, trimers, and monomers. Mammalian cell- ever, the sialylation of these glycans has been greatly reduced,
expressed HA oligomers, trimers, and monomers were ana- as clearly observed by a comparison of the core fucosylated
lyzed for their ability to elicit neutralizing antibody against H5 tetra-antennary glycans. Without the coexpression of NA, core
HA. Vaccination in mice was performed similarly, and antisera fucosylated tetra-antennary glycans with 1, 2, 3, and 4 sialic
were collected and analyzed for their neutralizing activities. acid residues were observed in abundance at m/z 3503, 3864,
Neutralizing antibody titers were significantly higher for ani- 4225, and 4586, respectively (Fig. 4A, Table 1). The coexpres-
mals immunized with oligomers (Fig. 2B). Cleaved trimers also sion of NA caused a loss of the signals at m/z 3864, 4225, and
elicited a neutralizing antibody response, although the titer 4586 and a concurrent increase in the abundance of the non-
was lower than that of the oligomers in this lentiviral neutral- sialylated core fucosylated tetra-antennary glycans at m/z 3142
ization assay (Fig. 2B). While uncleaved trimers also induced (Fig. 4A, Table 1). Similarly, a dramatic reduction in HA
neutralizing antibody, no detectable antibody responses were N-glycan sialylation was observed by the coexpression of NA
found in the animals immunized with cleaved monomers (Fig. (New Caledonia/99)(H1N1) (Fig. 4C, Table 1).
2B). When the ratio of neutralizing to total HA binding anti- Given that the mammalian cell-expressed oligomers elic-

Downloaded from jvi.asm.org by on January 8, 2009


bodies (Fig. 2D) was calculated, antisera from mice immunized ited the strongest neutralizing antibody response, we tested
with mammalian cell-expressed oligomer showed the highest whether the inclusion of NA during protein expression
percentage of neutralizing antibodies, suggesting that these would affect its immunogenicity. In the construct that ter-
complexes better preserve the physiologic trimeric spike struc- minates at the bromelain cleavage site, the inclusion of NA
ture. The ratios of neutralizing to total HA binding antibodies during protein expression reduced the ability of HA to elicit
in mice immunized with uncleaved and cleaved trimer were antibodies that neutralize HA/NA-pseudotyped virus (Fig.
⬃70% lower than those of mice immunized with oligomer. 5A). Since these proteins appeared similarly on purification
While a single dose of mammalian cell-expressed oligomers after gel filtration, this difference most likely was due to
elicited only modest levels of neutralizing antibody, as shown their stability in the Ribi adjuvant. In particular, the coex-
by the lentiviral neutralization assay, neutralizing titers against pression of NA with the bromelain site HA construct was
H5 (KAN-1) pseudovirus were enhanced substantially after a noted to reduce protein precipitation. In contrast, the in-
secondary boost 3 weeks after the initial injection (Fig. 2C). clusion of NA in the ⌬TM construct, which has 14 additional
Like insect-expressed HA, mammalian cell-expressed oligomer amino acids between the bromelain cleavage site and the
and trimer injected with Ribi adjuvant elicited antibodies of transmembrane domain, did not affect the neutralizing an-
IgG1 and IgG2a subclasses that were similar to the antibodies tibody response (Fig. 5B), suggesting that the additional
elicited by the inactivated influenza H5N1 subvirion vaccine sequence stabilized the protein in the presence of adjuvant.
(Fig. 2E). The rationale for testing the ⌬TM construct was that it
NA is not required for HA-elicited neutralizing antibody contained more of the HA protein ectodomain and might,
responses. NA has been shown to play a role in viral release therefore, represent a more native protein with additional
from cells (2). This viral enzyme cleaves terminal sialic acid determinants as an immunogen. However, the ⌬TM con-
residues from carbohydrate moieties on the surfaces of in- struct primarily formed oligomers that lacked trimers, sug-
fected cells and, therefore, promotes the release of progeny gesting that the inclusion of the additional amino acids
viruses (2). NA also cleaves sialic acid residues from HA, destabilized the trimeric form of the protein. Taking these
thereby preventing the aggregation of viruses (2). HA proteins results together, the coexpression of NA is not required to
made with or without NA coexpression behaved differently in elicit neutralizing antibodies by transmembrane-deleted,
the glycan array binding analysis (Fig. 3). HA trimers ex- stabilized oligomers of the HA protein, suggesting that this
pressed without NA showed no prominent binding to any of form of HA serves as a preferred immunogen.
the glycans tested (Fig. 3A). In contrast, mammalian cell-ex-
pressed HA trimers that coexpressed NA preferentially bound
DISCUSSION
to ␣2,3-linked sialic acid oligosaccharides (Fig. 3B).
The effect of the coexpression of NA on HA glycosylation Although egg-based vaccines have been used to combat sea-
was investigated by MS analysis (Fig. 4, Table 1). The N-linked sonal flu, the lengthy production cycles and limited manufac-
glycans from HA (Fig. 4A) and HA coexpressed with NA- turing capacity of egg-based vaccines are not conducive to
(KAN-1)(H5N1) (Fig. 4B) or NA(New Caledonia/99)(H1N1) facilitating a rapid response during a potential influenza virus
(Fig. 4C) were purified, derivatized, and subjected to MALDI- pandemic. Several clinical studies have shown that recombi-
TOF (MS). The spectra derived from HA without the coex- nant HA-based vaccines purified from baculovirus expression
pression of NA showed predominantly complex type N-glycans systems are safe and effective against H1N1 and H3N2 influ-
(m/z 1835 to 4586; Hex3HexNAc4Fuc-NeuAc4Hex7HexNAc6 enza viruses (7, 10, 11, 15–17). The protein-based approach
Fuc) (Fig. 4), which is consistent with bi-, tri-, and tetra-anten- represents an attractive alternative to egg-based technology,
VOL. 82, 2008 INFLUENZA VACCINATION WITH RECOMBINANT HA PROTEINS 6205

Downloaded from jvi.asm.org by on January 8, 2009


FIG. 3. Glycan array analysis of the specificity of HA trimers expressed with or without NA. Glycan microarray analysis of H5 HA proteins
expressed without (A) or with (B) the coexpression of NA. Glycans with related linkages are grouped by color and include selected glycoproteins
(orange), predominantly ␣2,3 sialosides (yellow) or ␣2,6 sialosides (green), ␣2,8 ligands (blue), and others (purple). This analysis was performed
by the Core H of the Consortium for Functional Genomics at Emory University.

since it uses HA proteins as antigens and does not require the a nickel affinity column followed by anion-exchange and gel
production of potentially dangerous live virus. Also, with this filtration chromatography. The entire purification process can
approach, vaccine production is not limited by the supply of be completed in 2 to 3 days, and protein production can easily
eggs and can be easily scaled up in a good-manufacturing- be scaled up. In both Hi5 insect cells and 293F mammalian
practices facility. cells, HA proteins were expressed as high-molecular-weight
Recently, a recombinant HA vaccine against avian H5N1 oligomers and stabilized trimers, demonstrating that the tri-
influenza virus has demonstrated tolerability in humans (16). merizing foldon sequence indeed prevented the HA from dis-
However, this vaccine only induced protective neutralizing an- sociating into monomers. Upon the removal of the foldon
tibody titers in 50% of the subjects receiving the highest dose sequence by thrombin digestion, only trimers and monomers
(two doses of 90 ␮g vaccine). Since it has been previously were present in the insect-expressed proteins, whereas in the
reported that recombinant HA proteins expressed in insect mammalian cell-expressed proteins only a small portion of
cells tend to form monomers (13), the suboptimal immunoge- monomer was observed after the removal of the foldon se-
nicity of this H5 HA vaccine may be due in part to recombinant quence. The discrepancy may be due to the different glyco-
HA protein not being presented in its native trimeric confor- sylation states of proteins derived from insect cells versus pro-
mation. In this study, we cloned the ectodomain of HA from an teins produced in mammalian cells, although it is certainly not
H5N1 virus (KAN-1) and expressed the HA proteins in mam- conclusive.
malian or insect cells. HA proteins initially were purified using We then evaluated the immunogenicity of these different
Downloaded from jvi.asm.org by on January 8, 2009

6206
VOL. 82, 2008 INFLUENZA VACCINATION WITH RECOMBINANT HA PROTEINS 6207

TABLE 1. Assignments of major molecular ions observed in


MALDI spectra of permethylated N-glycans from HA
trimers expressed with or without NA
(m/z) for:

N-glycan composition HA/NA


HA/NA
HA (New
(KAN-1)
Caledonia)

Hex5HexNAc2 1,580 1,580 1,580


Hex3HexNAc4Fuc1 1,835 1,835 1,835
Hex4HexNAc4Fuc1 2,040 2,040 2,040
Hex3HexNAc5Fuc1 2,081 2,081 2,081
Hex5HexNAc4Fuc1 2,244 2,244 2,244
Hex4HexNAc5Fuc1 2,285 2,285 2,285
FIG. 5. HA-elicited antibody response is independent of NA coex-
Hex5HexNAc5Fuc1 2,489 2,489 2,489
pression in oligomers from transmembrane-deleted, but not bromelain
Hex5HexNAc4Fuc1NeuAc1 2,605 NDa ND
site-truncated, HA. Neutralizing antibody titers in sera from mice
Hex5HexNAc5Fuc1 2,693 2,693 2,693
immunized with HA proteins produced with or without the coexpres-
Hex5HexNAc6Fuc1 2,734 2,734 2,734
sion of NA were examined. (A) In the bromelain site construct, the
Hex5HexNAc5Fuc1NeuAc1 2,851 ND ND
inclusion of NA [bromelain site oligomer (⫹NA)] reduced HA’s ability
Hex5HexNAc6Fuc1 2,938 2,938 2,938
to induce neutralizing antibodies compared to that of the bromelain
Hex5HexNAc4Fuc1NeuAc2 2,966 ND ND
site construct containing no NA [bromelain site oligomer (⫺NA)].
Hex6HexNAc5Fuc1NeuAc1 3,054 ND ND
(B) In ⌬TM oligomers, coexpression with NA [oligomer (⌬TM,
Hex7HexNAc6Fuc1 3,142 3,142 3,142
⫹NA)] did not alter the immunogenicity of the HA proteins compared
Hex7HexNAc6Fuc2 3,316 3,316 3,316
to ⌬TM oligomer expression without NA [oligomer (⌬TM, ⫺NA)].
Hex7HexNAc7Fuc1 3,387 3,387 3,387

Downloaded from jvi.asm.org by on January 8, 2009


Hex6HexNAc5Fuc1NeuAc2 3,415 ND ND
Hex7HexNAc6Fuc1NeuAc1 3,503 3,503 3,503
Hex7HexNAc6Fuc2NeuAc1 3,677 ND ND
Hex6HexNAc5Fuc1NeuAc3 3,776 ND ND cell-expressed oligomers or cleaved trimers were examined,
Hex7HexNAc6Fuc1NeuAc2 3,864 ND ND and their ability to elicit neutralizing antibodies against differ-
Hex7HexNAc6Fuc2NeuAc2 4,038 ND ND ent H5N1 strain pseudoviruses was similar (unpublished data),
Hex7HexNAc6Fuc1NeuAc3 4,225 ND ND though we cannot exclude the possibility of differences in their
Hex7HexNAc6Fuc1NeuAc4 4,586 ND ND
fine specificity. It should be noted that, although Ribi adjuvant
a
ND, not determined. does not contain any denaturants or reducing agents, its effect
on the stability and conformation of HA proteins is unknown.
We attempted to analyze this effect biochemically but were
forms of HA derived from either insect or mammalian cells unable to extract HA proteins from the lipid-rich components
using an HA/NA-pseudotyped lentiviral system (24). In this of this adjuvant. Although NA plays an essential role in viral
assay, the neutralization activity can be determined easily by replication and infection, the trimming of terminal sialic acid
measuring the ability of antisera from mice immunized with from the HA proteins by NA did not affect the immunogenicity
recombinant HA proteins to inhibit pseudovirus entry. It has of recombinant HA oligomers. However, the addition of NA
been shown that this pseudotype inhibition assay correlates did prevent the precipitation of purified protein and facilitated
highly with traditional microneutralization and hemagglutina- the production of the HA oligomers (data not shown). The
tion inhibition assays (6, 24) and can be easily performed in a removal of terminal sialic acids by NA appeared to be impor-
conventional biosafety level 2 laboratory with biosafety level 3 tant for the receptor binding of HA. Glycan binding analyses of
practices. Among the proteins produced from mammalian HA expressed in the insect cell, which lacks sialic acids, have
cells, high-molecular-weight oligomers elicited the highest ti- revealed a similar ␣2-3 specificity (12) to the NA-coexpression
ters of neutralizing antibody, followed by the cleaved trimers mammalian HA protein, which bound to ␣2,3-linked sialic acid
and uncleaved trimers. Cleaved monomers failed to induce oligosaccharides. These findings are consistent with the obser-
significant neutralizing antibodies against H5N1 virus, even vation that insect-produced, stabilized trimers elicited substan-
though anti-H5 antibodies were detected by ELISA. This may tial levels of neutralizing antibodies (Fig. 2A) despite the lack
be due to the preferential induction of antibodies against of the sialylation of HA in this cell type.
epitopes present in the monomeric form and not in the trimer, Although previous studies have shown that recombinant HA
similarly to that observed with human immunodeficiency virus proteins derived from insect cells elicit immune responses (7,
type 1 gp120 monomers and trimers (reviewed in reference 3). 10, 11, 15–17), our data provide evidence that oligomeric or
It also is possible that the monomeric form is less immunogenic trimeric HA produced in mammalian cells are comparable or
than the trimer/oligomer forms of the same protein. In a sep- slightly better in eliciting neutralizing antibodies against avian
arate study, antisera from animals immunized with mammalian H5N1 virus. Further testing will be required to determine

FIG. 4. MALDI-TOF mass spectra of permethylated N-glycans from HA trimers expressed with or without NA. Shown are profiles of N-glycans
from HA (A), HA coexpressed with NA isolated from the KAN-1 strain (B), or HA coexpressed with NA isolated from the New Caledonia (NC)
strain (C) from the 50% (vol/vol) acetonitrile fraction from C18 Sep-Paks. All molecular ions are [M ⫹ Na]⫹. Structural assignments are based on
monosaccharide composition, MALDI-TOF/TOF (MS/MS) analysis, and knowledge of N-glycan biosynthetic pathways.
6208 WEI ET AL. J. VIROL.

whether other adjuvants, such as alum, QS-21, or MF-59, can 8. Morens, D. M., and A. S. Fauci. 2007. The 1918 influenza pandemic: insights
for the 21st century. J. Infect. Dis. 195:1018–1028.
improve the immunogenicity of recombinant HA proteins. Not 9. Pancera, M., J. Lebowitz, A. Schon, P. Zhu, E. Freire, P. D. Kwong, K. H.
only could the potency of these adjuvants differ but also their Roux, J. Sodroski, and R. Wyatt. 2005. Soluble mimetics of human immu-
effects on the stability of the trimer may vary. These results nodeficiency virus type 1 viral spikes produced by replacement of the native
trimerization domain with a heterologous trimerization motif: characteriza-
eventually will require validation with the most active and tion and ligand binding analysis. J. Virol. 79:9954–9969.
manufacturable forms in human clinical trials. Nonetheless, 10. Powers, D. C., J. E. McElhaney, O. A. Florendo, Jr., M. C. Manning, C. M.
our data demonstrate that recombinant mammalian cell- or Upshaw, D. W. Bentley, and B. E. Wilkinson. 1997. Humoral and cellular
immune responses following vaccination with purified recombinant hemag-
insect-expressed trimeric HA proteins represent a promising glutinin from influenza A (H3N2) virus. J. Infect. Dis. 175:342–351.
approach to the development of vaccines relevant to seasonal 11. Powers, D. C., G. E. Smith, E. L. Anderson, D. J. Kennedy, C. S. Hackett,
and pandemic influenza virus. B. E. Wilkinson, F. Volvovitz, R. B. Belshe, and J. J. Treanor. 1995. Influenza
A virus vaccines containing purified recombinant H3 hemagglutinin are well
tolerated and induce protective immune responses in healthy adults. J. In-
ACKNOWLEDGMENTS fect. Dis. 171:1595–1599.
12. Stevens, J., O. Blixt, T. M. Tumpey, J. K. Taubenberger, J. C. Paulson, and
We thank Ati Tislerics and Hamani Henderson for manuscript prep-
I. A. Wilson. 2006. Structure and receptor specificity of the hemagglutinin
aration, Brenda Hartman and Michael Cichanowski for the prepara- from an H5N1 influenza virus. Science 312:404–410.
tion of figures, and members of the G.J.N. laboratory for helpful advice 13. Stevens, J., A. L. Corper, C. F. Basler, J. K. Taubenberger, P. Palese, and
and discussions. We acknowledge The Consortium for Functional Gly- I. A. Wilson. 2004. Structure of the uncleaved human H1 hemagglutinin from
comics, funded by the NIGMS GM62116, and David F. Smith, Emory the extinct 1918 influenza virus. Science 303:1866–1870.
University School of Medicine, Atlanta, GA, for the glycan array 14. Thompson, W. W., D. K. Shay, E. Weintraub, L. Brammer, N. Cox, L. J.
analysis. We thank Jacob Lebowitz for performing the sedimentation Anderson, and K. Fukuda. 2003. Mortality associated with influenza and
equilibrium measurements. Monovalent influenza virus subvirion vac- respiratory syncytial virus in the United States. JAMA 289:179–186.
cine, rgA/Vietnam/1203/2004 (H5N1), NR-4143, was obtained through 15. Treanor, J. J., R. F. Betts, G. E. Smith, E. L. Anderson, C. S. Hackett, B. E.
Wilkinson, R. B. Belshe, and D. C. Powers. 1996. Evaluation of a recombi-
the NIH Biodefense and Emerging Infections Research Resources
nant hemagglutinin expressed in insect cells as an influenza vaccine in young
Repository, NIAID, NIH. and elderly adults. J. Infect. Dis. 173:1467–1470.

Downloaded from jvi.asm.org by on January 8, 2009


This research was supported by the Intramural Research Program, 16. Treanor, J. J., B. E. Wilkinson, F. Masseoud, J. Hu-Primmer, R. Battaglia,
Vaccine Research Center, NIAID, NIH, and by the Biotechnology and D. O’Brien, M. Wolff, G. Rabinovich, W. Blackwelder, and J. M. Katz. 2001.
Biological Sciences Research Council (BBSRC) of the Wellcome Trust Safety and immunogenicity of a recombinant hemagglutinin vaccine for H5
(A.D. and S.M.H.). A.D. was supported as a BBSRC Professorial influenza in humans. Vaccine 19:1732–1737.
Research Fellow. 17. Wang, K., K. M. Holtz, K. Anderson, R. Chubet, W. Mahmoud, and M. M.
Cox. 2006. Expression and purification of an influenza hemagglutinin–one
REFERENCES step closer to a recombinant protein-based influenza vaccine. Vaccine 24:
1. Alymova, I. V., S. Kodihalli, E. A. Govorkova, B. Fanget, C. Gerdil, and R. G. 2176–2185.
Webster. 1998. Immunogenicity and protective efficacy in mice of influenza 18. Webby, R. J., and R. G. Webster. 2003. Are we ready for pandemic influ-
B virus vaccines grown in mammalian cells or embryonated chicken eggs. enza? Science 302:1519–1522.
J. Virol. 72:4472–4477. 19. Webster, R. G., M. Peiris, H. Chen, and Y. Guan. 2006. H5N1 outbreaks and
2. Colman, P. M. 1994. Influenza virus neuraminidase: structure, antibodies, enzootic influenza. Emerg. Infect. Dis. 12:3–8.
and inhibitors. Protein Sci. 3:1687–1696. 20. Wilson, I. A., J. J. Skehel, and D. C. Wiley. 1981. Structure of the haemag-
3. Douek, D. C., P. D. Kwong, and G. J. Nabel. 2006. The rational design of an glutinin membrane glycoprotein of influenza virus at 3 A resolution. Nature
AIDS vaccine. Cell 124:677–681. 289:366–373.
4. Hampson, A. W. 1997. Surveillance for pandemic influenza. J. Infect. Dis. 21. World Health Organization. 2006. Basic health information on human avian
176(Suppl. 1):S8–S13. influenza A/H5N1. http://www.wpro.who.int/information_sources/databases
5. Jang-Lee, J., S. J. North, M. Sutton-Smith, D. Goldberg, M. Panico, H. /regional_statistics/rstat_Human_Avian_influenza.htm.
Morris, S. Haslam, and A. Dell. 2006. Glycomic profiling of cells and tissues 22. World Health Organization. 2007. Cumulative number of confirmed human
by mass spectrometry: fingerprinting and sequencing methodologies. Meth- cases of avian influenza A/(H5N1) reported to WHO, 15 June 2007. http:
ods Enzymol. 415:59–86. //www.who.int/csr/disease/avian_influenza/country/cases_table_2007_06_15/en
6. Kong, W.-P., C. Hood, Z.-Y. Yang, C. J. Wei, L. Xu, A. Garcia-Sastre, T. M. /index.html.
Tumpey, and G. J. Nabel. 2006. Protective immunity to lethal challenge of 23. Yang, Z.-Y., B. K. Chakrabarti, L. Xu, B. Welcher, W.-P. Kong, K. Leung, A.
the 1918 pandemic influenza virus by vaccination. Proc. Natl. Acad. Sci. USA Panet, J. R. Mascola, and G. J. Nabel. 2004. Selective modification of
103:15987–15991. variable loops alters tropism and enhances immunogenicity of human im-
7. Lakey, D. L., J. J. Treanor, R. F. Betts, G. E. Smith, J. Thompson, E. munodeficiency virus type 1 envelope. J. Virol. 78:4029–4036.
Sannella, G. Reed, B. E. Wilkinson, and P. F. Wright. 1996. Recombinant 24. Yang, Z.-Y., C. J. Wei, W.-P. Kong, L. Wu, L. Xu, D. F. Smith, and G. J.
baculovirus influenza A hemagglutinin vaccines are well tolerated and im- Nabel. 2007. Immunization by avian H5 influenza hemagglutinin mutants
munogenic in healthy adults. J. Infect. Dis. 174:838–841. with altered receptor binding specificity. Science 317:825–828.

You might also like