Research Article
15 March 2014

Assessment of Influenza Virus Hemagglutinin Stalk-Based Immunity in Ferrets

ABSTRACT

Therapeutic monoclonal antibodies that target the conserved stalk domain of the influenza virus hemagglutinin and stalk-based universal influenza virus vaccine strategies are being developed as promising countermeasures for influenza virus infections. The pan-H1-reactive monoclonal antibody 6F12 has been extensively characterized and shows broad efficacy against divergent H1N1 strains in the mouse model. Here we demonstrate its efficacy against a pandemic H1N1 challenge virus in the ferret model of influenza disease. Furthermore, we recently developed a universal influenza virus vaccine strategy based on chimeric hemagglutinin constructs that focuses the immune response on the conserved stalk domain of the hemagglutinin. Here we set out to test this vaccination strategy in the ferret model. Both strategies, pretreatment of animals with a stalk-reactive monoclonal antibody and vaccination with chimeric hemagglutinin-based constructs, were able to significantly reduce viral titers in nasal turbinates, lungs, and olfactory bulbs. In addition, vaccinated animals also showed reduced nasal wash viral titers. In summary, both strategies showed efficacy in reducing viral loads after an influenza virus challenge in the ferret model.
IMPORTANCE Influenza virus hemagglutinin stalk-reactive antibodies tend to be less potent yet are more broadly reactive and can neutralize seasonal and pandemic influenza virus strains. The ferret model was used to assess the potential of hemagglutinin stalk-based immunity to provide protection against influenza virus infection. The novelty and significance of the findings described in this report support the development of vaccines stimulating stalk-specific antibody responses.

INTRODUCTION

In the United States, epidemics of seasonal influenza cause substantial morbidity (1) and significant mortality (2). Despite the proven ability of inactivated and live attenuated influenza virus vaccines to reduce the impact of influenza, the potential of currently licensed influenza vaccines is not fully manifested because of several factors. First, influenza vaccination coverage rates remain low (3). In particular, a recent survey of 11,963 adults (18 to 64 years of age) revealed that only 28.2% reported receiving the 2008-2009 influenza vaccine (4). Second, influenza vaccines induce immune responses that specifically neutralize influenza viruses that are closely related to the vaccine strain, yet the potency of these neutralizing responses diminishes with antigenic drift. Thus, annual influenza vaccination is required to maintain protective immune responses against a “moving target” (5). Third, the emergence of pandemic influenza virus strains is difficult to predict, and once an influenza pandemic emerges, it is even more difficult to redirect vaccine production in a timely fashion to respond to a pandemic, as happened during the 2009 H1N1 influenza pandemic (6, 7). Predictions of influenza pandemics is further complicated by the realization that several influenza virus subtypes possess pandemic potential, as evidenced by the emergence of avian influenza A (H7N9) virus in March 2013 (8) and sporadic human infections with H4, H5, H6, H7, H9, and H10 avian influenza viruses (914).
Hemagglutinin (HA)-specific universal influenza vaccines have the potential to mitigate these limitations by focusing humoral immune responses on its antigenically conserved stalk region. Approaches to developing stalk-focused universal vaccines have included headless HA (1517), recombinant soluble HA (1822), synthetic polypeptides (23), prime-boost regimens (24, 25), nanoparticles (26), and recombinant influenza viruses expressing chimeric HA (cHA) (19, 21). Stalk-specific vaccines would shift the humoral immune responses away from the immunodominant globular-head domain to the more conserved stalk domain. Universal vaccines stimulating stalk-specific antibody responses would have several desirable aspects, including (i) conferring protection against homologous and drifted influenza virus strains, (ii) obviating the need for annual influenza vaccinations with reformulated H1, H3, and B virus strains that antigenically match prevalent circulating strains, and (iii) conferring increased protection against newly emerging influenza viruses with pandemic potential (27, 28). Importantly, stalk-reactive antibodies occur naturally in humans, albeit in general at low frequencies, and have been detected in experimentally vaccinated mice (21, 2937). On the basis of sequence conservation, a universal influenza vaccine targeting the HA stalk would likely require three components to cover group 1 (H1, H2, H5, H6, H8, H9, H11, H12, H13, H16, H17) and group 2 (H3, H4, H7, H10, H14, H15) influenza A and B virus HAs.
In this study, we have examined in ferrets the level of protection conferred by group 1 HA stalk-specific antibodies against a challenge infection with pandemic H1N1 virus. Ferrets were passively immunized with stalk-reactive monoclonal antibodies (MAbs) or vaccinated with recombinant viral vectors expressing cHAs known to induce stalk-reactive antibodies in mice. These studies revealed that group 1 stalk-specific antibodies could reduce titers of infectious virus within the nasal cavity and also reduced pulmonary virus titers in immunized ferrets challenged with a pandemic H1N1 influenza virus that contains an HA head not present in the cHA vaccination regimen. These findings suggest that ferrets produce HA stalk-reactive antibodies following vaccination with cHAs and that stalk-reactive antibodies provide protection from heavy viral loads after a challenge infection in this influenza animal model.

MATERIALS AND METHODS

Cells and viruses.

Madin-Darby canine kidney (MDCK), 293T, 293, A549, and baby hamster kidney 21 (BHK-21) cells were propagated in Dulbecco's modified Eagle's medium (DMEM) or minimum essential medium (both from Gibco). A/Netherlands/602/09 pandemic H1N1 virus and the recombinant B-cH9/1 virus (a B/Yamagata/16/88 virus that expresses a cH9/1 HA as described in reference 35) were grown in embryonated chicken eggs, and titers were determined on MDCK cells in medium containing tosyl phenylalanyl chloromethyl ketone (TPCK)-treated trypsin as described before.

Generation of a VSV vector expressing cH5/1 protein.

The cH5/1 gene (an A/Viet Nam/1203/04 H5 head on top of an A/PR/8/34 H1 stalk domain [19, 21]) was amplified by PCR, and the SalI-NheI restriction enzyme-digested PCR product was then cloned into the XhoI and NheI sites of the pVSV-XN2 (38) vector to generate pVSV-cH5/1. Recombinant vesicular stomatitis virus (VSV) expressing cH5/1 HA (VSV-cH5/1) was recovered with the above plasmid with minor modifications to the previously described method (39). Briefly, BHK-21 cells were infected with the T7 polymerase-expressing vaccinia virus vTF7-3 (40) at a multiplicity of infection (MOI) of 20. At 1 h postinfection, the cells were transfected with the pVSV-cH5/1 plasmid and support plasmids pBS-N, pBS-P, pBS-G, and pBS-L. At 48 h posttransfection, the cell culture medium was collected, filtered through a 0.1-μm filter, and passaged onto BHK-21 cells. After a cytopathic effect (CPE) became evident, the culture medium was collected and virus was plaque purified and used to grow stocks. A VSV vector expressing green fluorescent protein (GFP) was used as a control.

Generation of an adenovirus 5 vector expressing cH6/1 protein.

Prior to virus generation, cH6/1 (an A/mallard/Sweden/81/02 H6 globular-head domain on top of an H1/PR8 stalk domain [21, 35]) was cloned into a previously described transfer plasmid (pE1A-CMV, lacking the HA epitope tag) (41). For virus generation, 2.0 × 106 human embryonic kidney 293 (HEK-293) cells (generously supplied by Patrick Hearing) were plated per well of a six-well dish and transfected the following day with a 3:1 ratio of X-tremeGENE 9 (Roche) to DNA according to the manufacturer's instructions. Cells were transfected with a total of 5.5 μg of DNA consisting of 5 μg of PvuI-linearized cH6/1 pE1A-CMX plasmid and 500 ng of dl309 viral DNA that had been digested with ClaI/XbaI to remove the left end of the adenoviral genome (bp 1 to 920). X-tremeGENE 9-transfected, ClaI/XbaI-digested viral DNA was used as a negative control. After 24 h of incubation, cells were overlaid with 2× DMEM-supplemented 1% agarose for plaque selection. Overlays were reapplied approximately every 3 days for 1 week, and then plaques were isolated for screening and used for 10 lysate generation. Once a CPE was evident (2 to 3 days), cells were harvested and frozen at −80°C. Cells underwent four freeze-thaw cycles, and then viral DNA was prepared by an established method for sequencing (42). Once the cH6/1 sequence was confirmed, virus stocks were amplified on HEK-293 cells and purified by consecutive banding on step and equilibrium cesium chloride gradients. Expression of the cH6/1 protein was confirmed by immunofluorescence staining on A549-infected cells with anti-stalk MAb 6F12 (43), and virus titers were determined by standard plaque assay on HEK-293 cells. The empty control adenovirus vector (in the same genomic background) was kindly provided by Patrick Hearing.

Immunostaining.

MDCK cells were infected at a MOI of 1 with B-cH9/1 or wild-type B/Yamagata/16/88 and fixed (0.5% paraformaldehyde) at 24 h postinfection. A subset of cells was permeabilized with 0.1% Triton X-100 and stained with an anti-influenza B virus nucleoprotein antibody (Abcam; 1:1,000). The rest of the cells were stained with anti-H1 stalk antibody 6F12 (10 μg/ml) or anti-H9 head antibody G1-26 (BEI Resources NR-9485; 1:1,000). 293T and A549 cells were infected/transduced with empty or cH6/1-expressing adenovirus at an MOI of about 100. Cells were permeabilized with 0.5% Triton X-100 and stained with an anti-hexon antibody (Abcam; 1:1,000), an anti-H1 stalk antibody 6F12 (10 μg/ml), or an anti-H6 head antibody NatalieC (10 μg/ml). An Alexa 488-conjugated anti-mouse antibody (Life Technologies; 1:1,000) was used as the secondary antibody for immunofluorescence analysis. Vero cells were infected at a low MOI with VSV expressing GFP or cH5/1 HA. Cells were fixed at 24 h postinfection and stained with mouse anti-VSV serum (1:1,000), MAb 6F12 (10 μg/ml), or anti-H5 head antibody VN4-10 (BEI Resources NR-2737; 1:1,000). A horseradish peroxidase (HRP)-linked anti-mouse antibody (Santa Cruz; 1:3,000) was used as the secondary antibody, and stained cells were visualized with aminoethyl carbazole substrate solution (Millipore).

Antibodies and recombinant proteins.

Mouse MAbs 6F12 (H1 stalk reactive, IgG2b) (43) and XY102 (A/Hong Kong/1/68 HA head reactive, hemagglutination inhibition [HI] active, IgG2b) (44) were purified from supernatants of hybridoma cultures as described before. Briefly, the supernatants were passed over a column loaded with protein G-Sepharose (GE Healthcare), washed, eluted, and concentrated, and the buffer was exchanged for phosphate-buffered saline (PBS; pH 7.4) with Amicon Ultra centrifugation units (Millipore). Protein concentrations were determined by the A280 method with a NanoDrop device. Recombinant HAs were expressed as ectodomains with a C-terminal trimerization domain and a hexahistidine tag with the baculovirus system as described before (20, 45). Protein concentrations were measured by the Bradford method.

Animals, passive transfer, immunization, and challenge.

Five-month-old male Fitch ferrets were confirmed to be seronegative for circulating H1N1, H3N2, and B influenza viruses prior to purchase from Triple F Farms (Sayre, PA). Ferrets were housed in PlasLabs poultry incubators with free access to food and water (4648). All of the animal experiments described here were conducted by using protocols approved by the Icahn School of Medicine at Mount Sinai Institutional Animal Care and Use Committee. Animals were anesthetized by intramuscular administration of ketamine/xylazine for all of the procedures described here, including bleeding, nasal washes, vaccination, infection, and passive transfer.
For passive-transfer experiments, animals were bled to obtain baseline serum samples 2 weeks before the transfer. On day −1, 30 mg/kg of mouse MAb 6F12 or XY102 (n = 2 per group) was transferred intravenously via the vena cava (Fig. 1A). At 24 h postinoculation, animals were bled and infected with 104 PFU of A/Netherlands/602/09 (pandemic H1N1) virus. Nasal washes were then taken on days 1 and 3 postinfection, and body weights were monitored daily. Animals were observed for approximately 30 min daily for signs of morbidity (e.g., sneezing). On day 4 postinfection, animals were sacrificed and exsanguinated and tissue samples were taken from the upper left and right lobes of the lungs, olfactory bulb, and nasal turbinates.
FIG 1
FIG 1 Persistence and distribution of MAb 6F12 in two passively immunized ferrets. (A) Schematic representation of the passive immunization and challenge study. A baseline serum sample was collected prior to the passive immunization of ferrets with 30 mg/kg of MAb 6F12 on day −1. On day 0 postimmunization, a serum sample was taken and ferrets were challenged by infection with 104 PFU of A/Netherlands/602/09. (B) Titers of MAb 6F12 in serum samples collected on days −1, 0, and 4 of passive immunization were measured by ELISA reactivity against baculovirus-produced H1 from A/California/04/09. (C) Titers of MAb 6F12 in nasal wash samples collected on days 1 and 3 of passive immunization were measured by ELISA reactivity against baculovirus-produced H1 from A/California/04/09. (D) Titers of MAb 6F12 in lung homogenate samples collected on day 4 of passive immunization were measured by ELISA reactivity against baculovirus-produced H1 from A/California/04/09. In the experiments whose results are shown in panels C and D, nasal wash or lung samples from ferrets passively immunized with MAb XY102, which specifically recognizes H3 of A/Hong Kong/1/1968, served as negative controls (n = 2 ferrets). OD, optical density.
For vaccination experiments, animals (n = 5) were intranasally infected with 2 × 107 PFU (in 1 ml of PBS) of influenza B virus vector B-cH9/1 HA (an H9 head on top of an H1 stalk domain [21, 35]) (see Fig. 4A). At 3 weeks postinfection, animals were boosted by the intramuscular administration of 2 × 105 PFU (in 0.5 ml) of recombinant VSV-cH5/1 HA (an H5 globular-head domain on top of an H1 stalk domain [19, 21]). A second boost consisting of a replication-deficient recombinant adenovirus 5 vector expressing the cH6/1 protein (an H6 globular-head domain on top of an H1 stalk domain) was given intranasally and intramuscularly (1.2 × 108 PFU in 0.5 ml per site) 3 weeks after the first boost. Control group animals received the same empty or GFP-expressing virus (VSV) vectors in the same sequence (n = 4). Four weeks after the last priming, animals were challenged with 104 PFU of A/Netherlands/602/09 (pandemic H1N1) virus. Nasal washes were then taken on days 1 and 3 postinfection, and weight was monitored daily. Animals were observed for approximately 30 min daily, and signs of morbidity (e.g., sneezing) were recorded. On day 4 postinfection, animals were sacrificed and tissue samples were taken from the lung (upper right lobe), olfactory bulb, and nasal turbinates.

HI assays.

HI assays were performed as described elsewhere (46, 49). Working stocks of each influenza virus strain were prepared by diluting the virus stock to a final HA titer of 8 HA units/50 μl. Each serum sample was serially diluted 2-fold in PBS (25 μl per well) in 96-V-well microtiter plates. Then, 25 μl of working stock of the influenza virus strain was added to each well so that all of the wells contained a final volume of 50 μl. The serum-virus samples were then incubated at room temperature for 45 min to allow HA head-specific antibodies to neutralize the influenza virus. To each well, 50 μl of a 0.5% suspension of turkey or chicken red blood cells was added. The assay plates were then incubated at 4°C until red blood cells in the PBS control sample formed a button and red blood cells hemagglutinated in control wells containing virus and no antibody. The HI titer was defined as the reciprocal of the highest dilution of antibody that inhibited red blood cell hemagglutination by influenza virus.

ELISAs.

Enzyme-linked immunosorbent assays (ELISAs) were performed as described before (20, 31). Briefly, plates were coated with 2 μg/ml of recombinant, baculovirus-produced H1 (from A/California/04/09, A/New Caledonia/20/99, and A/South Carolina/1/18), H2 (from A/Japan/305/57), or H17 (from A/yellow shouldered bat/Guatemala/06/10) HA protein (20). Wells were then incubated with serial (2-fold) dilutions of ferret sera, nasal washes, or lung homogenates for 1 h at room temperature. After extensive washes, plates were incubated with anti-mouse antibody (for MAbs; Santa Cruz) or anti-ferret (Alpha Diagnostics International) IgG HRP-labeled secondary antibody for another hour at room temperature. Plates were washed again and then developed with SigmaFast o-phenylenediamine dihydrochloride substrate and read on a Synergy H1 (BioTek) plate reader.

RESULTS

Persistence and tissue distribution of 6F12 in the ferret.

Previously, we have shown that HA head-reactive IgA but not IgG antibody is able to prevent transmission in the ferret and guinea pig models of influenza virus infection (49). We reasoned that at an especially low concentration (3 mg/kg), IgG is not efficiently transported to mucosal surfaces. This transport might be additionally inhibited by the lower Fc-Fc receptor interactions between mouse MAbs and the ferret host. In addition, the half-life of mouse IgG in ferrets has not been well characterized; however, a previous study that examined the therapeutic potential of a humanized MAb, m102, in the ferret model of Nipah virus infection reported an elimination half-life of 3.5 days following the intravenous administration of 25 mg of MAb (50). We were therefore curious if treatment with a large dose of MAb (30 mg/kg) would increase the Ab concentration on mucosal surfaces and protect from upper respiratory tract infection. Ferrets were passively immunized by the intravenous administration of 30 mg/kg of either H1 stalk-specific MAb 6F12 or H3-specific MAb XY102 (isotype control) (Fig. 1A). The persistence and tissue distribution of MAb 6F12 were examined by ELISA with baculovirus-produced H1 from A/California/04/09. MAb 6F12 could be easily detected by ELISA within serum samples on day 4 after passive immunization (Fig. 1B). In addition, MAb 6F12 was detected by ELISA in nasal wash samples collected on day 1, but the level of MAb declined by day 3 after a challenge infection (Fig. 1C). MAb 6F12 was also detected by ELISA in lung homogenates on day 4 postchallenge (Fig. 1D). These results suggest that passive immunization by intravenous administration of MAb 6F12 would confer a window of protection against a challenge infection within the ferret respiratory tract.

Prophylactic administration of 6F12 reduces viral loads in lungs, olfactory bulbs, and nasal turbinates.

Mouse MAb 6F12 is an H1 stalk domain-specific antibody that potently inhibits viral replication of H1N1 virus isolates spanning from 1930 to 2009 and efficiently protects mice prophylactically and therapeutically from a viral challenge (43). In order to investigate whether 6F12 would also be efficacious prophylactically in the ferret model of influenza disease, a 30-mg/kg dose of this MAb was administered to ferrets intravenously 24 h prechallenge and the animals were then challenged with pandemic H1N1 strain A/Netherlands/602/09. Control group ferrets received the same amount of an isotype control antibody (Fig. 1A). Viral titers from nasal wash samples taken on days 1 and 3 were slightly lower in the 6F12-treated animals than in the control group (Fig. 2A). The effect was more pronounced on day 1 than on day 3, which matches the lower 6F12 titers found in nasal washes on day 3 postchallenge. Furthermore, the day 4 nasal turbinate titers of 6F12-treated ferrets were lower than those of control animals (Fig. 2B). A reduction of approximately 2 logs in 6F12-treated animals was also observed in the olfactory bulb (Fig. 2C), and lung titers were approximately 1 log lower than those of control animals (Fig. 2D). Weight loss was only minimal and similar in both groups (data not shown). In summary, prophylactic treatment of ferrets with MAb 6F12 reduced the viral loads in challenged animals in all of the analyzed tissues. The readouts established for this experiment were then also used to compare and analyze the efficacy of a cHA vaccine regimen in ferrets.
FIG 2
FIG 2 Prophylactic administration of MAb 6F12 reduced viral titers following a challenge infection. Ferrets were passively immunized with MAb 6F12 (green bars; n = 2 ferrets) or isotype control MAb XY102 (black bars; n = 2 ferrets). On day 0 after passive immunization, ferrets were challenge infected with 104 PFU of A/Netherlands/602/09. (A) Virus titers in nasal wash samples collected on day 1 or 3 after a challenge infection were determined by plaque assay. On day 4 after the challenge infection, titers of influenza virus in nasal turbinate (B), olfactory bulb (C), and lung (D) samples were assessed by plaque assay.

Vaccination with cHAs induces stalk-reactive antibodies in the ferret.

We have previously shown that vaccination of inbred BALB/c mice with cHA constructs (HAs with a conserved stalk domain but divergent head domains) induces broadly neutralizing stalk-reactive antibodies (21). Here we wanted to test if vaccination of ferrets would also induce stalk-reactive antibodies. To this end, we used viral vectors expressing cHA constructs (Fig. 3). Prior to vaccination of ferrets with the viral vectors, the expression of the cHA was demonstrated by immunostaining. The expression of cHA by an influenza B virus vector expressing B-cH9/1 HA (21, 35) was demonstrated by immunofluorescence assay with infected MDCK cells (Fig. 3A). The expression of cHA by a VSV expressing cH5/1 HA (19, 21) was demonstrated by immunostaining of virus plaques in Vero cells (Fig. 3B). The expression of cHA by a replication-deficient adenovirus 5 vector expressing cH6/1 HA (an H6 head on top of an H1/PR8 stalk domain [19, 21]) was demonstrated by immunofluorescence assay with infected 293T and A549 cells (Fig. 3C).
FIG 3
FIG 3 Expression of cHAs by viral vectors. (A) An engineered influenza B virus expresses cH9/1 HA (H9 head on top of an H1 stalk domain) instead of influenza B HA. Shown is staining of B-cH9/1- or B-wt (wild-type influenza B virus)-infected cells with an anti-influenza B nucleoprotein antibody (anti-NP), anti-H1 stalk antibody 6F12 (anti-stalk), or an anti-H9 head antibody (anti-H9 head). (B) A recombinant VSV was engineered to express cH5/1 (H5 head on top of an H1 stalk domain) HA as a transgene. Shown is staining of VSV-cH5/1- or VSV-GFP-infected Vero cells with anti-VSV mouse serum (anti-VSV), anti-H1 stalk antibody 6F12 (anti-stalk), or an anti-H5 head antibody (anti-H5 head). (C) A replication-deficient adenovirus was engineered to express cH6/1 HA (H6 head on top of an H1 stalk domain). Shown are infected 293T cells stained for the presence of adenovirus (anti-hexon) and transduced A459 cells stained with anti-H1 stalk antibody 6F12 (anti-stalk) or an anti-H6 head antibody (anti-H6 head).
Ferrets were first vaccinated with B-cH9/1 HA and then boosted with VSV-cH5/1 HA (an H5 head on top of an H1 stalk domain [19, 21]) and then with a replication-deficient adenovirus 5 vector expressing cH6/1 HA (an H6 head on top of an H1/PR8 stalk domain [19, 21]) (Fig. 4A). This vaccination regimen was chosen in order to avoid the generation of antibodies against any antigen in pandemic H1N1 virus different from the HA stalk, which could also contribute to protection after a subsequent challenge. Vaccinated animals developed low seroreactivity against pandemic H1 HA after priming. This reactivity was boosted approximately 4-fold by the cH5/1 vaccination and then again 8-fold by the final cH6/1 vaccination. Sera from vector control animals exhibited only background reactivity that was comparable to the reactivity of pooled prevaccination sera of the ferrets used. Since cHA-vaccinated animals were naive to the H1 head domain and also tested HI negative against the pandemic H1N1 strain A/Netherlands/602/09, we conclude that any reactivity to H1 strains is based on cross-reactive antibodies to the conserved stalk domain. Furthermore, our cHA vaccine constructs are based on the stalk domain of A/PR/8/34 H1 HA. Therefore, reactivity to pandemic H1 HA already represents heterologous stalk reactivity within H1 HAs. We also tested reactivity to two more H1 HAs, the HA from prepandemic seasonal strain A/New Caledonia/20/99 and the HA from 1918 pandemic H1N1 strain A/South Carolina/1/18 (Fig. 4C and D). Sera from cHA-vaccinated animals reacted strongly with both proteins. In order to test if cHA vaccination induces cross-reactivity to other group 1 subtypes, we also tested reactivity against an H2 HA from A/Japan/305/57 virus (Fig. 4E) and against an H17 HA (from recently discovered bat H17N10 influenza virus strain A/yellow shouldered bat/Guatemala/06/10) (Fig. 4F). Sera from cHA-vaccinated ferrets reacted strongly with both HAs, while sera from vector control animals showed only background reactivity (Fig. 4E and F). Cross-reactivity against group 2 HA was not expected, since earlier studies with mice have shown that group 1 stalk-based cHA vaccination regimens do not protect from a group 2 virus challenge and vice versa (21, 22). Importantly, we did not detect any H1 head-specific antibody responses against the challenge virus following the vaccination regimen as measured by HI assay (data not shown). As positive controls, convalescent-phase reference sera from two ferrets infected with A/California/7/2009 were included in the HI assay, and each reference serum yielded an HI titer of 1,280.
FIG 4
FIG 4 Ferrets develop HA stalk-specific humoral responses after repeated immunization with viral vectors expressing cHAs. (A) Schematic representation of the HA stalk-based vaccination strategy used in this study. Ferrets (n = 5) were vaccinated with influenza B virus expressing cH9/1 HA, boosted with VSV-cH5/1 HA, and boosted a second time with an adenovirus 5 vector expressing the cH6/1 protein. Control ferrets (n = 4) were vaccinated with wild-type influenza B virus or VSV (expressing GFP) and adenovirus (empty) vectors. Ferrets were then challenged by infection with 104 PFU of A/Netherlands/602/09 virus. The development of broadly cross-reactive stalk-specific antibody responses was assessed by ELISA with baculovirus-produced H1 from A/California/04/09 (B), H1 from A/South Carolina/1/18) (C), H1 from A/New Caledonia/20/99 (D), H2 (from A/Japan/305/57) (E), or H17 (from A/yellow shouldered bat/Guatemala/06/10) (F). OD, optical density.

cHA vaccine constructs protect ferrets from a viral challenge.

In order to test the protection that cHA vaccination would confer on ferrets, we challenged the animals with the pandemic H1N1 strain A/Netherlands/602/09 (Fig. 5A). The readouts were the same as for the passive-transfer experiment; we measured virus titers in day 1 and 3 nasal washes and in the lungs, olfactory bulb, and nasal turbinates on day 4 postinfection. Interestingly, nasal wash titers were lower in cHA-vaccinated ferrets than in control animals on day 1 (approximately 5-fold), as well on day 3 (more than 10-fold), when the difference was highly significant (P = 0.0005) (Fig. 5A). This result is not surprising since we expected that the intranasally applied priming and second boost would induce stalk-reactive mucosal IgA antibodies. The reduction of virus titers in the nasal washes is also reflected by a significant reduction of virus titers in the nasal turbinates of about 10-fold (P = 0.0331) (Fig. 5B). Furthermore, the olfactory bulb virus titers of cHA-vaccinated animals were more than 2 logs lower than those of vector control animals (P = 0.0062) (Fig. 5C). In fact, we were unable to detect virus in the olfactory bulbs of four out of five cHA-vaccinated ferrets, whereas high virus titers were found in the olfactory bulbs of all four control ferrets. Finally, we also detected a reduction of approximately half a log of lung virus titers in cHA-vaccinated ferrets compared to those of vector control ferrets (Fig. 5D). In summary, the protective efficacy of the cHA vaccine was comparable to (nasal turbinates, olfactory bulbs, and lung titers) or better than (nasal wash titers) that of prophylactically administered MAb 6F12.
FIG 5
FIG 5 The HA stalk-based vaccination strategy confers protection against a challenge infection with A/Netherlands/602/09 virus. Ferrets (n = 5) were vaccinated with B-cH9/1 HA, boosted with VSV-cH5/1 HA, and boosted a second time with an adenovirus 5 vector expressing the cH6/1 protein. Control ferrets (n = 4) were vaccinated with wild-type influenza B virus or VSV (expressing GFP) and adenovirus (empty) vectors. (A) Titers of challenge virus in nasal wash samples collected on day 1 or 3 after the challenge infection were determined by plaque assay. On day 4 after the challenge infection, titers of influenza virus in nasal turbinate (B), olfactory bulb (C), and lung (D) samples were assessed by plaque assay. n.s., not significant.

DISCUSSION

In recent years, broadly neutralizing antibodies against the conserved stalk domain of the influenza virus HA have been isolated (30, 32, 36, 37, 43, 51, 52, 5356). These antibodies can be used for prophylactic and therapeutic treatments of influenza virus infections. Although the large amount of MAb needed for treatment might preclude the use of the antibodies in the general population, this approach might be useful for the therapy of severe influenza cases, especially when drug-resistant viruses in an immunocompromised host are involved (5763). We therefore wanted to evaluate MAb 6F12 in a prophylactic setting in the ferret model. This antibody has pan-H1 neutralizing activity in vitro and is able to protect mice from a challenge with H1N1 influenza viruses that span almost 100 years of antigenic drift (43). We show here that MAb 6F12 is indeed efficacious against a pandemic H1N1 strain in the ferret model as well. In particular, prophylactic administration of MAb 6F12 resulted in a more pronounced reduction of virus titers in olfactory bulbs and lungs. Unexpectedly, we could also detect this mouse IgG antibody at low titers in nasal wash samples from treated ferrets. These low levels of antibody found in the nasal washes correlated well with small reductions of nasal wash viral titers. Several factors could contribute to the pronounced reduction of virus titers in olfactory bulb and lung samples compared to the modest reduction of virus titers observed in nasal wash samples. On day 4 after intravenous injection, high levels of MAb 6F12 could be detected in serum and lung samples, which contrasts with the low level of MAb 6F12 detected in nasal wash samples. In addition, MAb 6F12 liberated by the homogenization of olfactory bulb and lung tissue samples would bind to and neutralize a small fraction of the virus present in the tissue samples prior to the determination of virus titers by plaque assay. We speculate that 6F12-like antibodies, if transported efficiently to mucosal surfaces (e.g., locally induced by intranasally administered vaccines) would be able to efficiently reduce nasal wash virus titers and possibly have an impact on transmission as well. We recently showed that this is the case for globular-head-reactive MAb 30D1, which efficiently blocks replication when administered to guinea pigs as IgA (efficiently transported to mucosal surfaces) but lacks efficacy when administered as IgG (not efficiently transported to mucosal surfaces) (49).
In an “antibody-guided” vaccine approach based on stalk-reactive antibodies, we have developed cHA vaccine constructs (19, 21). These constructs possess a conserved, structurally integrated stalk domain in combination with divergent globular-head domains from “exotic” subtypes (21). By sequentially immunizing mice with these constructs, we protected them from a challenge with heterologous (H1N1) and heterosubtypic (other group 1 HA-expressing viruses) influenza viruses (21). Here, we tested the efficacy of this vaccine approach in the ferret model. By immunizing ferrets with combinations of divergent globular heads and a conserved stalk domain, we hoped to get an immune response focused on broadly neutralizing epitopes in the stalk. This strategy is based on the observation that sequential infection/vaccination with seasonal H1N1 and pandemic H1N1 viruses (which have highly divergent globular-head domains and highly conserved stalk domains) induces high levels of stalk-reactive antibodies in humans (32, 3537, 64). Similar findings were also obtained in the mouse model (31). Here, in the ferret model, we show that a cHA-based immunization strategy confers protection against a pandemic H1N1 challenge. The observed level of protection was similar to or better than that conferred by inactivated, antigenically matched, unadjuvanted split vaccine administered once (65, 66) or twice (67) or an antigenically matched experimental vaccinia virus-vectored construct (68). It is of note that the cHA-based vaccine did not induce any HI-active antibodies, but vaccinated ferrets were able to produce a broadly reactive anti-stalk response against divergent group 1 HA subtypes. This proof-of-principle study focused on protection afforded by the stalk domain of HA. A human vaccine candidate based on the same principle would most likely consist of inactivated or attenuated cHA-expressing viruses that also have a neuraminidase (NA). We believe that the antibody titers against the more conserved NA would be boosted as well in the absence of an immunodominant globular-head domain (69, 70). These antibodies would then also contribute to broad protection. Furthermore, conserved internal proteins like the nucleoprotein induce strong protective T-cell responses that contribute to protection as well (7174). We have conclusively shown that such a vaccination strategy based on the H1 HA stalk domain is able to broadly protect against group 1 HA-expressing viruses in mice but was unable to protect against an H3N2 challenge virus (21). We therefore believe that a successful human vaccination strategy would need to contain a group 1, a group2, and an influenza B virus stalk component to induce broadly neutralizing stalk antibodies.
In summary, we have shown that treatment of ferrets with a stalk-reactive antibody and vaccination by a stalk-based vaccination strategy are efficacious in protecting against an influenza virus challenge. We believe that both strategies are valuable additions to the armamentarium for fighting seasonal and pandemic influenza virus infections in the human population.

ACKNOWLEDGMENTS

We thank Chen Wang and Richard Cadagan for excellent technical assistance.
This study was partially funded by a National Institutes of Health National Institute of Allergy and Infectious Diseases program project grant (P01AI097092), by PATH, and by R01-AI080781. Florian Krammer was supported by an Erwin Schrödinger fellowship (J 3232) from the Austrian Science Fund (FWF). Matthew S. Miller was supported by a Canadian Institutes of Health Research postdoctoral fellowship.

REFERENCES

1.
Molinari NA, Ortega-Sanchez IR, Messonnier ML, Thompson WW, Wortley PM, Weintraub E, and Bridges CB. 2007. The annual impact of seasonal influenza in the US: measuring disease burden and costs. Vaccine 25:5086–5096.
2.
Centers for Disease Control and Prevention. 2010. Estimates of deaths associated with seasonal influenza—United States, 1976-2007. MMWR Morb. Mortal. Wkly. Rep. 59:1057–1062. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm5933a1.htm.
3.
Centers for Disease Control and Prevention. 2011. Interim results: state-specific influenza vaccination coverage—United States, August 2010-February 2011. MMWR Morb. Mortal. Wkly. Rep. 60:737–743. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm6022a3.htm.
4.
Williams WW, Lu PJ, Lindley MC, Kennedy ED, Singleton JA, and Centers for Disease Control and Prevention. 2012. Influenza vaccination coverage among adults—National Health Interview Survey, United States, 2008-09 influenza season. MMWR Morb. Mortal. Wkly. Rep. 61(Suppl):65–72. http://www.cdc.gov/mmwr/preview/mmwrhtml/su6102a11.htm.
5.
Wang TT and Palese P. 2011. Biochemistry. Catching a moving target. Science 333:834–835.
6.
Centers for Disease Control and Prevention. 2009. Swine influenza A (H1N1) infection in two children—Southern California, March-April 2009. MMWR Morb. Mortal. Wkly. Rep. 58:400–402. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm5815a5.htm.
7.
Centers for Disease Control and Prevention. 2009. Update on influenza A (H1N1) 2009 monovalent vaccines. MMWR Morb. Mortal. Wkly. Rep. 58:1100–1101. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm5839a3.htm.
8.
Gao R, Cao B, Hu Y, Feng Z, Wang D, Hu W, Chen J, Jie Z, Qiu H, Xu K, Xu X, Lu H, Zhu W, Gao Z, Xiang N, Shen Y, He Z, Gu Y, Zhang Z, Yang Y, Zhao X, Zhou L, Li X, Zou S, Zhang Y, Yang L, Guo J, Dong J, Li Q, Dong L, Zhu Y, Bai T, Wang S, Hao P, Yang W, Han J, Yu H, Li D, Gao GF, Wu G, Wang Y, Yuan Z, and Shu Y. 2013. Human infection with a novel avian-origin influenza A (H7N9) virus. N. Engl. J. Med. 368:1888–1897.
9.
Centers for Disease Control and Prevention. 2012. Notes from the field: highly pathogenic avian influenza A (H7N3) virus infection in two poultry workers—Jalisco, Mexico, July 2012. MMWR Morb. Mortal. Wkly. Rep. 61:726–727. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm6136a4.htm.
10.
Kayali G, Barbour E, Dbaibo G, Tabet C, Saade M, Shaib HA, Debeauchamp J, and Webby RJ. 2011. Evidence of infection with H4 and H11 avian influenza viruses among Lebanese chicken growers. PLoS One 6:e26818.
11.
Koopmans M, Wilbrink B, Conyn M, Natrop G, van der Nat H, Vennema H, Meijer A, van Steenbergen J, Fouchier R, Osterhaus A, and Bosman A. 2004. Transmission of H7N7 avian influenza A virus to human beings during a large outbreak in commercial poultry farms in the Netherlands. Lancet 363:587–593.
12.
Peiris M, Yuen KY, Leung CW, Chan KH, Ip PL, Lai RW, Orr WK, and Shortridge KF. 1999. Human infection with influenza H9N2. Lancet 354:916–917.
13.
Tweed SA, Skowronski DM, David ST, Larder A, Petric M, Lees W, Li Y, Katz J, Krajden M, Tellier R, Halpert C, Hirst M, Astell C, Lawrence D, and Mak A. 2004. Human illness from avian influenza H7N3, British Columbia. Emerg. Infect. Dis. 10:2196–2199.
14.
Yuan J, Zhang L, Kan X, Jiang L, Yang J, Guo Z, and Ren Q. 2013. Origin and molecular characteristics of a novel 2013 avian influenza A(H6N1) virus causing human infection in Taiwan. Clin. Infect. Dis. 57:1367–1368.
15.
Bommakanti G, Citron MP, Hepler RW, Callahan C, Heidecker GJ, Najar TA, Lu X, Joyce JG, Shiver JW, Casimiro DR, ter Meulen J, Liang X, and Varadarajan R. 2010. Design of an HA2-based Escherichia coli expressed influenza immunogen that protects mice from pathogenic challenge. Proc. Natl. Acad. Sci. U. S. A. 107:13701–13706.
16.
Sagawa H, Ohshima A, Kato I, Okuno Y, and Isegawa Y. 1996. The immunological activity of a deletion mutant of influenza virus haemagglutinin lacking the globular region. J. Gen. Virol. 77(Pt 7):1483–1487.
17.
Steel J, Lowen AC, Wang TT, Yondola M, Gao Q, Haye K, Garcia-Sastre A, and Palese P. 2010. Influenza virus vaccine based on the conserved hemagglutinin stalk domain. mBio 1:e00018–10.
18.
Goff PH, Eggink D, Seibert CW, Hai R, Martínez-Gil L, Krammer F, and Palese P. 2013. Adjuvants and immunization strategies to induce influenza virus hemagglutinin stalk antibodies. PLoS One 8:e79194.
19.
Hai R, Krammer F, Tan GS, Pica N, Eggink D, Maamary J, Margine I, Albrecht RA, and Palese P. 2012. Influenza viruses expressing chimeric hemagglutinins: globular head and stalk domains derived from different subtypes. J. Virol. 86:5774–5781.
20.
Krammer F, Margine I, Tan GS, Pica N, Krause JC, and Palese P. 2012. A carboxy-terminal trimerization domain stabilizes conformational epitopes on the stalk domain of soluble recombinant hemagglutinin substrates. PLoS One 7:e43603.
21.
Krammer F, Pica N, Hai R, Margine I, and Palese P. 2013. Chimeric hemagglutinin influenza virus vaccine constructs elicit broadly protective stalk-specific antibodies. J. Virol. 87:6542–6550.
22.
Margine I, Krammer F, Hai R, Heaton NS, Tan GS, Andrews SA, Runstadler JA, Wilson PC, Albrecht RA, Garcia-Sastre A, and Palese P. 2013. Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J. Virol. 87:10435–10446.
23.
Wang TT, Tan GS, Hai R, Pica N, Ngai L, Ekiert DC, Wilson IA, Garcia-Sastre A, Moran TM, and Palese P. 2010. Vaccination with a synthetic peptide from the influenza virus hemagglutinin provides protection against distinct viral subtypes. Proc. Natl. Acad. Sci. U. S. A. 107:18979–18984.
24.
Wei CJ, Boyington JC, McTamney PM, Kong WP, Pearce MB, Xu L, Andersen H, Rao S, Tumpey TM, Yang ZY, and Nabel GJ. 2010. Induction of broadly neutralizing H1N1 influenza antibodies by vaccination. Science 329:1060–1064.
25.
Wei CJ, Yassine HM, McTamney PM, Gall JG, Whittle JR, Boyington JC, and Nabel GJ. 2012. Elicitation of broadly neutralizing influenza antibodies in animals with previous influenza exposure. Sci. Transl. Med. 4:147ra114.
26.
Kanekiyo M, Wei CJ, Yassine HM, McTamney PM, Boyington JC, Whittle JR, Rao SS, Kong WP, Wang L, and Nabel GJ. 2013. Self-assembling influenza nanoparticle vaccines elicit broadly neutralizing H1N1 antibodies. Nature 499:102–106.
27.
Krammer F and Palese P. 2013. Influenza virus hemagglutinin stalk-based antibodies and vaccines. Curr. Opin. Virol. 3:521–530.
28.
Krammer F and Palese P. 2013. Universal influenza virus vaccines: need for clinical trials. Nat. Immunol. 15:3–5.
29.
Corti D, Suguitan AL Jr, Pinna D, Silacci C, Fernandez-Rodriguez BM, Vanzetta F, Santos C, Luke CJ, Torres-Velez FJ, Temperton NJ, Weiss RA, Sallusto F, Subbarao K, and Lanzavecchia A. 2010. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J. Clin. Invest. 120:1663–1673.
30.
Ekiert DC, Friesen RH, Bhabha G, Kwaks T, Jongeneelen M, Yu W, Ophorst C, Cox F, Korse HJ, Brandenburg B, Vogels R, Brakenhoff JP, Kompier R, Koldijk MH, Cornelissen LA, Poon LL, Peiris M, Koudstaal W, Wilson IA, and Goudsmit J. 2011. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 333:843–850.
31.
Krammer F, Pica N, Hai R, Tan GS, and Palese P. 2012. Hemagglutinin stalk-reactive antibodies are boosted following sequential infection with seasonal and pandemic H1N1 influenza virus in mice. J. Virol. 86:10302–10307.
32.
Li GM, Chiu C, Wrammert J, McCausland M, Andrews SF, Zheng NY, Lee JH, Huang M, Qu X, Edupuganti S, Mulligan M, Das SR, Yewdell JW, Mehta AK, Wilson PC, and Ahmed R. 2012. Pandemic H1N1 influenza vaccine induces a recall response in humans that favors broadly cross-reactive memory B cells. Proc. Natl. Acad. Sci. U. S. A. 109:9047–9052.
33.
Miller MS, Gardner TJ, Krammer F, Aguado LC, Tortorella D, Basler CF, and Palese P. 2013. Neutralizing antibodies against previously encountered influenza virus strains increase over time: a longitudinal analysis. Sci. Transl. Med. 5:198ra107.
34.
Miller MS, Tsibane T, Krammer F, Hai R, Rahmat S, Basler CF, and Palese P. 2013. 1976 and 2009 H1N1 influenza virus vaccines boost anti-hemagglutinin stalk antibodies in humans. J. Infect. Dis. 207:98–105.
35.
Pica N, Hai R, Krammer F, Wang TT, Maamary J, Eggink D, Tan GS, Krause JC, Moran T, Stein CR, Banach D, Wrammert J, Belshe RB, García-Sastre A, and Palese P. 2012. Hemagglutinin stalk antibodies elicited by the 2009 pandemic influenza virus as a mechanism for the extinction of seasonal H1N1 viruses. Proc. Natl. Acad. Sci. U. S. A. 109:2573–2578.
36.
Thomson CA, Wang Y, Jackson LM, Olson M, Wang W, Liavonchanka A, Keleta L, Silva V, Diederich S, Jones RB, Gubbay J, Pasick J, Petric M, Jean F, Allen VG, Brown EG, Rini JM, and Schrader JW. 2012. Pandemic H1N1 influenza infection and vaccination in humans induces cross-protective antibodies that target the hemagglutinin stem. Front. Immunol. 3:87.
37.
Wrammert J, Koutsonanos D, Li GM, Edupuganti S, Sui J, Morrissey M, McCausland M, Skountzou I, Hornig M, Lipkin WI, Mehta A, Razavi B, Del Rio C, Zheng NY, Lee JH, Huang M, Ali Z, Kaur K, Andrews S, Amara RR, Wang Y, Das SR, O'Donnell CD, Yewdell JW, Subbarao K, Marasco WA, Mulligan MJ, Compans R, Ahmed R, and Wilson PC. 2011. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. J. Exp. Med. 208:181–193.
38.
Schnell MJ, Buonocore L, Whitt MA, and Rose JK. 1996. The minimal conserved transcription stop-start signal promotes stable expression of a foreign gene in vesicular stomatitis virus. J. Virol. 70:2318–2323.
39.
Lawson ND, Stillman EA, Whitt MA, and Rose JK. 1995. Recombinant vesicular stomatitis viruses from DNA. Proc. Natl. Acad. Sci. U. S. A. 92:4477–4481.
40.
Fuerst TR, Niles EG, Studier FW, and Moss B. 1986. Eukaryotic transient-expression system based on recombinant vaccinia virus that synthesizes bacteriophage T7 RNA polymerase. Proc. Natl. Acad. Sci. U. S. A. 83:8122–8126.
41.
Evans JD and Hearing P. 2003. Distinct roles of the adenovirus E4 ORF3 protein in viral DNA replication and inhibition of genome concatenation. J. Virol. 77:5295–5304.
42.
Schmid SI and Hearing P. 1999. Adenovirus DNA packaging, p 47–59. In Wold WSM (ed), Adenovirus methods and protocols. Humana Press, Totowa, NJ.
43.
Tan GS, Krammer F, Eggink D, Kongchanagul A, Moran TM, and Palese P. 2012. A pan-H1 anti-hemagglutinin monoclonal antibody with potent broad-spectrum efficacy in vivo. J. Virol. 86:6179–6188.
44.
Moran T, Liu YC, Schulman JL, and Bona CA. 1984. Shared idiotopes among monoclonal antibodies specific for A/PR/8/34 (H1N1) and X-31(H3N2) influenza viruses. Proc. Natl. Acad. Sci. U. S. A. 81:1809–1812.
45.
Margine I, Palese P, and Krammer F. 2013. Expression of functional recombinant hemagglutinin and neuraminidase proteins from the novel H7N9 Influenza virus using the baculovirus expression system. J. Vis. Exp. 81:e51112.
46.
Baker SF, Guo H, Albrecht RA, Garcia-Sastre A, Topham DJ, and Martínez-Sobrido L. 2013. Protection against lethal influenza with a viral mimic. J. Virol. 87:8591–8605.
47.
Martínez-Romero C, de Vries E, Belicha-Villanueva A, Mena I, Tscherne DM, Gillespie VL, Albrecht RA, de Haan CA, and Garcia-Sastre A. 2013. Substitutions T200A and E227A in the hemagglutinin of pandemic 2009 influenza A virus increase lethality but decrease transmission. J. Virol. 87:6507–6511.
48.
Seibert CW, Kaminski M, Philipp J, Rubbenstroth D, Albrecht RA, Schwalm F, Stertz S, Medina RA, Kochs G, Garcia-Sastre A, Staeheli P, and Palese P. 2010. Oseltamivir-resistant variants of the 2009 pandemic H1N1 influenza A virus are not attenuated in the guinea pig and ferret transmission models. J. Virol. 84:11219–11226.
49.
Seibert CW, Rahmat S, Krause JC, Eggink D, Albrecht RA, Goff PH, Krammer F, Duty JA, Bouvier NM, Garcia-Sastre A, and Palese P. 2013. Recombinant IgA is sufficient to prevent influenza virus transmission in guinea pigs. J. Virol. 87:7793–7804.
50.
Zhu Z, Bossart KN, Bishop KA, Crameri G, Dimitrov AS, McEachern JA, Feng Y, Middleton D, Wang LF, Broder CC, and Dimitrov DS. 2008. Exceptionally potent cross-reactive neutralization of Nipah and Hendra viruses by a human monoclonal antibody. J. Infect. Dis. 197:846–853.
51.
Dreyfus C, Laursen NS, Kwaks T, Zuijdgeest D, Khayat R, Ekiert DC, Lee JH, Metlagel Z, Bujny MV, Jongeneelen M, van der Vlugt R, Lamrani M, Korse HJ, Geelen E, Sahin Ö, Sieuwerts M, Brakenhoff JP, Vogels R, Li OT, Poon LL, Peiris M, Koudstaal W, Ward AB, Wilson IA, Goudsmit J, and Friesen RH. 2012. Highly conserved protective epitopes on influenza B viruses. Science 337:1343–1348.
52.
Ekiert DC, Bhabha G, Elsliger MA, Friesen RH, Jongeneelen M, Throsby M, Goudsmit J, and Wilson IA. 2009. Antibody recognition of a highly conserved influenza virus epitope. Science 324:246–251.
53.
Heaton NS, Leyva-Grado VH, Tan GS, Eggink D, Hai R, and Palese P. 2013. In vivo bioluminescent imaging of influenza a virus infection and characterization of novel cross-protective monoclonal antibodies. J. Virol. 87:8272–8281.
54.
Sui J, Hwang WC, Perez S, Wei G, Aird D, Chen LM, Santelli E, Stec B, Cadwell G, Ali M, Wan H, Murakami A, Yammanuru A, Han T, Cox NJ, Bankston LA, Donis RO, Liddington RC, and Marasco WA. 2009. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat. Struct. Mol. Biol. 16:265–273.
55.
Throsby M, van den Brink E, Jongeneelen M, Poon LL, Alard P, Cornelissen L, Bakker A, Cox F, van Deventer E, Guan Y, Cinatl J, ter Meulen J, Lasters I, Carsetti R, Peiris M, de Kruif J, and Goudsmit J. 2008. Heterosubtypic neutralizing monoclonal antibodies cross-protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS One 3:e3942.
56.
Wang TT, Tan GS, Hai R, Pica N, Petersen E, Moran TM, and Palese P. 2010. Broadly protective monoclonal antibodies against H3 influenza viruses following sequential immunization with different hemagglutinins. PLoS Pathog. 6:e1000796.
57.
DeVries A, Wotton J, Lees C, Boxrud D, Uyeki T, and Lynfield R. 2012. Neuraminidase H275Y and hemagglutinin D222G mutations in a fatal case of 2009 pandemic influenza A (H1N1) virus infection. Influenza Other Respir. Viruses 6:e85–8.
58.
Hu Y, Lu S, Song Z, Wang W, Hao P, Li J, Zhang X, Yen HL, Shi B, Li T, Guan W, Xu L, Liu Y, Wang S, Tian D, Zhu Z, He J, Huang K, Chen H, Zheng L, Li X, Ping J, Kang B, Xi X, Zha L, Li Y, Zhang Z, Peiris M, and Yuan Z. 2013. Association between adverse clinical outcome in human disease caused by novel influenza A H7N9 virus and sustained viral shedding and emergence of antiviral resistance. Lancet 381:2273–2279.
59.
Hurt AC, Leang SK, Tiedemann K, Butler J, Mechinaud F, Kelso A, Downie P, and Barr IG. 2013. Progressive emergence of an oseltamivir-resistant A(H3N2) virus over two courses of oseltamivir treatment in an immunocompromised paediatric patient. Influenza Other Respir. Viruses 7:904–908.
60.
Iioka F, Sada R, Maesako Y, Nakamura F, and Ohno H. 2012. Outbreak of pandemic 2009 influenza A/H1N1 infection in the hematology ward: fatal clinical outcome of hematopoietic stem cell transplant recipients and emergence of the H275Y neuraminidase mutation. Int. J. Hematol. 96:364–369.
61.
Shetty AK, Ross GA, Pranikoff T, Gubareva LV, Sechrist C, Guirand DM, Abramson J, and Lin JJ. 2012. Oseltamivir-resistant 2009 H1N1 influenza pneumonia during therapy in a renal transplant recipient. Pediatr. Transplant. 16:E153–E157.
62.
van der Vries E, Stittelaar KJ, van Amerongen G, Veldhuis Kroeze EJ, de Waal L, Fraaij PL, Meesters RJ, Luider TM, van der Nagel B, Koch B, Vulto AG, Schutten M, and Osterhaus AD. 2013. Prolonged influenza virus shedding and emergence of antiviral resistance in immunocompromised patients and ferrets. PLoS Pathog. 9:e1003343.
63.
van Kampen JJ, Bielefeld-Buss AJ, Ott A, Maaskant J, Faber HJ, Lutisan JG, and Boucher CA. 2013. Case report: oseltamivir-induced resistant pandemic influenza A (H1N1) virus infection in a patient with AIDS and Pneumocystis jirovecii pneumonia. J. Med. Virol. 85:941–943.
64.
Sangster MY, Baer J, Santiago FW, Fitzgerald T, Ilyushina NA, Sundararajan A, Henn AD, Krammer F, Yang H, Luke CJ, Zand MS, Wright PF, Treanor JJ, Topham DJ, and Subbarao K. 2013. The B cell response and hemagglutinin stalk-reactive antibody production in different age cohorts following 2009 H1N1 influenza vaccination. Clin. Vaccine Immunol. 20:867–876.
65.
Ellebedy AH, Fabrizio TP, Kayali G, Oguin TH, Brown SA, Rehg J, Thomas PG, and Webby RJ. 2010. Contemporary seasonal influenza A (H1N1) virus infection primes for a more robust response to split inactivated pandemic influenza A (H1N1) virus vaccination in ferrets. Clin. Vaccine Immunol. 17:1998–2006.
66.
Hossain MJ, Bourgeois M, Quan FS, Lipatov AS, Song JM, Chen LM, Compans RW, York I, Kang SM, and Donis RO. 2011. Virus-like particle vaccine containing hemagglutinin confers protection against 2009 H1N1 pandemic influenza. Clin. Vaccine Immunol. 18:2010–2017.
67.
Baras B, de Waal L, Stittelaar KJ, Jacob V, Giannini S, Kroeze EJ, van den Brand JM, van Amerongen G, Simon JH, Hanon E, Mossman SP, and Osterhaus AD. 2011. Pandemic H1N1 vaccine requires the use of an adjuvant to protect against challenge in naïve ferrets. Vaccine 29:2120–2126.
68.
Kreijtz JH, Süzer Y, Bodewes R, Schwantes A, van Amerongen G, Verburgh RJ, de Mutsert G, van den Brand J, van Trierum SE, Kuiken T, Fouchier RA, Osterhaus AD, Sutter G, and Rimmelzwaan GF. 2010. Evaluation of a modified vaccinia virus Ankara (MVA)-based candidate pandemic influenza A/H1N1 vaccine in the ferret model. J. Gen. Virol. 91:2745–2752.
69.
Kilbourne ED, Cerini CP, Khan MW, Mitchell JW, and Ogra PL. 1987. Immunologic response to the influenza virus neuraminidase is influenced by prior experience with the associated viral hemagglutinin. I. Studies in human vaccinees. J. Immunol. 138:3010–3013.
70.
Palese P and Wang TT. 2011. Why do influenza virus subtypes die out? A hypothesis. mBio 2:e00150–11.
71.
Bodewes R, Kreijtz JH, van Amerongen G, Hillaire ML, Vogelzang-van Trierum SE, Nieuwkoop NJ, van Run P, Kuiken T, Fouchier RA, Osterhaus AD, and Rimmelzwaan GF. 2013. Infection of the upper respiratory tract with seasonal influenza A(H3N2) virus induces protective immunity in ferrets against infection with A(H1N1)pdm09 virus after intranasal, but not intratracheal, inoculation. J. Virol. 87:4293–4301.
72.
Hillaire ML, van Trierum SE, Kreijtz JH, Bodewes R, Geelhoed-Mieras MM, Nieuwkoop NJ, Fouchier RA, Kuiken T, Osterhaus AD, and Rimmelzwaan GF. 2011. Cross-protective immunity against influenza pH1N1 2009 viruses induced by seasonal influenza A (H3N2) virus is mediated by virus-specific T-cells. J. Gen. Virol. 92:2339–2349.
73.
Kreijtz JH, Bodewes R, van den Brand JM, de Mutsert G, Baas C, van Amerongen G, Fouchier RA, Osterhaus AD, and Rimmelzwaan GF. 2009. Infection of mice with a human influenza A/H3N2 virus induces protective immunity against lethal infection with influenza A/H5N1 virus. Vaccine 27:4983–4989.
74.
Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, Lambkin-Williams R, Gilbert A, Oxford J, Nicholas B, Staples KJ, Dong T, Douek DC, McMichael AJ, and Xu XN. 2012. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 18:274–280.

Information & Contributors

Information

Published In

cover image Journal of Virology
Journal of Virology
Volume 88Number 615 March 2014
Pages: 3432 - 3442
Editor: T. S. Dermody
PubMed: 24403585

History

Received: 12 October 2013
Accepted: 23 December 2013
Published online: 15 March 2014

Permissions

Request permissions for this article.

Contributors

Authors

Florian Krammer
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Rong Hai
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Mark Yondola
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Present address: Mark Yondola, Avatar Biotechnologies, Brooklyn, NY.
Gene S. Tan
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Victor H. Leyva-Grado
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Alex B. Ryder
Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
Matthew S. Miller
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
John K. Rose
Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
Peter Palese
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Adolfo García-Sastre
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Global Health & Emerging Pathogens Institute at Icahn School of Medicine at Mount Sinai, New York, New York, USA
Randy A. Albrecht
Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
Global Health & Emerging Pathogens Institute at Icahn School of Medicine at Mount Sinai, New York, New York, USA

Editor

T. S. Dermody
Editor

Notes

Address correspondence to Randy A. Albrecht, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy