Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Aug 2.
Published in final edited form as: Nat Struct Mol Biol. 2018 Feb 2;25(2):115–121. doi: 10.1038/s41594-018-0025-9

Structural insights into the design of novel anti-influenza therapies

Nicholas C Wu 1, Ian A Wilson 1,2,§
PMCID: PMC5930012  NIHMSID: NIHMS962801  PMID: 29396418

Abstract

A limited arsenal of therapies is available to tackle the emergence of a future influenza pandemic or even to effectively deal with the continual outbreaks of seasonal influenza. However, recent findings hold great promise for design of novel vaccines and therapeutics, including the possibility of more universal treatments. A major contribution to those advances comes from structural biology, in particular from the many studies on influenza hemagglutinin (HA), the major surface antigen. The HA primary function is to enable the virus to enter host cells, and structural work has revealed the various HA conformational forms generated during the entry process. Other studies have explored how human broadly neutralizing antibodies (bnAbs), designed proteins, peptides and small molecules, can inhibit and neutralize the virus. Here we review milestones in HA structural biology and how the recent insights from broadly neutralizing antibodies are leading to design of novel vaccines and therapeutics.

Introduction

Influenza virus is a negative-strand RNA virus that contains eight RNA segments, encoding at least 12 proteins (PB2, PB1, PB1-F2, PA, PA-X, HA, NA, NP, M1, M2, NS1, and NS2). Two of these proteins, hemagglutinin (HA) and neuraminidase (NA), are cell surface glycoproteins that enable the virus to enter and to escape from host cells, respectively. HA is a homotrimer that is synthesized as a single polypeptide chain (HA0), which is subsequently cleaved by host cell proteases to attain its fusion-competent state. The mature HA trimer is therefore composed of HA1 and HA2 subunits that remain cross-linked after cleavage through a single disulfide bond. The HA trimer can be divided into two structural as well as functional domains, the head and the stem, that comprise the receptor-binding site (RBS) and the fusion machinery, respectively. NA is an enzyme that cleaves the sialoside receptor off from the cell surface and enables progeny virus to escape from the infected cell to subsequently infect new cells. Both HA and NA activities are essential for viral infection. However, HA greatly outnumbers NA on the virus surface and consequently is the main target of the humoral immune response. Nevertheless, NA is the primary target for the anti-influenza drugs oseltamivir and zanamivir1 due to the ability to more readily target the NA active site compared to the much shallower HA RBS.

Here we review progress on the structural and functional characterization of HA in particular with human broadly neutralizing antibodies (bnAbs), which have provided exciting new insights and stimulated structure-based design of novel vaccines and new classes of therapeutics to target influenza virus.

Hemagglutinin structure and function

Influenza A viruses have 18 different HA subtypes (H1–18), whereas influenza B viruses have two different lineages (Yamagata and Victoria lineages). The natural reservoir for influenza viruses are wild aquatic birds, and 16 of these 18 HA subtypes (H1–H16) are resident in the bird population. Genomic RNAs of the other two influenza A subtypes (H17 and H18) have recently been found in bats2, 3, although live virus of these two subtypes has yet to be isolated. The influenza virus HA structure (H3 subtype from the 1968 influenza pandemic) was first determined in 1981, and was also the first viral antigen from an enveloped virus to be described4, 5 (Fig. 1). The identification of substitutions in HA that account for the differential recognition of avian-type versus human-type receptors (α2–3 versus α2–6 linked sialosides) in 19836 facilitated structural determination of HA-bound receptor complexes with sialic acid analogues in 19887. Another unknown was whether the precursor HA0 undergoes substantial conformational changes when converting to its fusion-competent form, as HA1 and HA2, and that was answered in 1988: the HA0 structure revealed surprisingly few differences between the cleaved and uncleaved forms8, other than at the cleavage site, which seem to differ from the larger changes suggested recently for some other viral envelope proteins, such as HIV-1 Env9. The next burning question was what conformational changes in HA lead to its membrane fusion activity in the low pH of endosomal compartments. The structure of a fragment of the HA stem in 1994 showed the massive rearrangements that HA undergoes to acquire its post-fusion form10.

Figure 1. Milestones of influenza HA structural biology.

Figure 1

Time line plotting crystal structures that represent important contributions to our understanding of HA structure and function, and key discoveries of human heterosubtypic bnAbs that have led to development of HA-targeted antivirals and vaccines. HA stalk is shown in cyan; HA head in dark gray; broadly neutralizing antibodies (bnAbs) in pink. Sialic acid receptors, TBHQ, and Arbidol are shown in sphere representation (carton: yellow, oxygen: red, nitrogen: blue, sulfur: orange).

Further questions arose as to whether there are substantial structural differences in the HAs from subtypes in influenza viruses responsible for the other pandemics, such as the 1918 H1N1 Spanish influenza11, 12, 1957 H2N2 Asian influenza13, 14, and 2009 H1N1 swine-origin pandemic15, 16, 17, or in the more recent emerging viruses of concern to human health, such as H5N118, 19, H7N920, 21, 22, H10N823, 24, 25, and H6N124, 26, 27, as well as influenza B HA and other influenza A subtypes3, 19, 28, 29, 30, 31, 32. This compendium of structures indicated that the HA architecture is highly conserved, but the surface properties and glycosylation patterns differ extensively among influenza subtypes and types. Furthermore, as the virus evolves after entering the human population, the glycans cover more of the HA surface33, 34. Influenza A viruses can also be classified phylogenetically into group 1 (H1, H2, H5, H6, H7, H8, H9, H11, H12, H13, H16, H17, and H18) and group 2 (H3, H4, H7, H10, H14, and H15). The main functional differences between these groups arise in the different ways that they interact with their receptor35 and in the definition of their epitopes35, 36, especially in the head region.

HA stem binders

Despite these advances, it was not known until relatively recently whether human antibodies could be found that are able to broadly neutralize influenza virus. The big breakthrough came in 2008–2009 with the isolation and characterization of bnAbs that could neutralize most influenza A group 1 viruses37, 38, 39 (Fig. 1). Surprisingly, those antibodies, such as CR6261 and F10, did not bind to the hypervariable head, as do the strain-specific antibodies that are normally elicited by infection and vaccination; instead, they were found to target the much more highly conserved and less accessible stem region using only the variable heavy chain for interaction39, 40. Thus, the HA stem suddenly became of major interest as a bnAb target and suggested that a more universal vaccine might be possible. Indeed, back in 1993, a mouse antibody with some heterosubtypic breadth (C179) had been reported to target the HA stem region41, but this antibody was largely overlooked by researchers in the influenza field. A crystal structure of C179 some 20 years later confirmed that it bound to the same stem region as these human bnAbs42.

Further extensive searches then ensued for other human antibodies that could target the HA stem with binding profiles that differed from the first set of bnAbs, which were all derived from the same VH1–69 germline family and exclusively targeted group 1 HAs. The Crucell group discovered another antibody (CR8020), whose heavy chain is encoded by VH1–18 germline gene43. CR8020 binds slightly lower down the stem than the original bnAbs and targets group 2 viruses (Fig. 2). The next major development was the isolation by the Lanzavecchia group of antibody FI6v3, which can target all group 1 and group 2 HAs44. FI6v3 arose from a different germline gene family, VH3–30, and targets the same stem region as the VH1–69 antibodies, but with a completely different binding mode and in more conventional way with interactions from both the light and heavy chain variable regions (Fig. 2). In another major step forward, Crucell then identified an antibody (CR9114) that can bind to both influenza A and influenza B viruses45 (Fig. 2). Such an antibody represents the epitome of what one would want to elicit in a universal vaccine. All of these broadly neutralizing antibodies inhibit the membrane fusion activity of HA by stabilizing the pre-fusion form of the HA trimer and preventing the conformational changes induced by low pH that release the fusion peptide39, 41, 43, 44, 45.

Figure 2. Anti-HA broadly neutralizing antibodies (bnAbs).

Figure 2

(a) Structures of representative RBS-targeted (C05 (PDB 4FP8)50 and F045–092 (PDB 4O58)51) and stem-binding (CR9114 (PDB 4FQI)45 and FI6v3 (PDB 3ZTJ)44) bnAbs in complex with HA are shown. (b) The epitopes of four stem-binding bnAbs, namely CR8020 (PDB 3SDY)43, CR6261 (PDB 3GBM)40, FI6v3 (PDB 3ZTJ)44, and CR9114 (PDB 4FQI)45 are compared. The epitope of the indicated bnAb is colored lime. For comparison, regions that correspond to the epitope of the other three bnAbs are colored white. (c) The binding and neutralization breadths of CR802043, CR626138, 40, FI6v344, CR911445, F045–09249, 51, and C0550 are shown. The phylogenetic relationship of HAs from influenza B virus and from different influenza A virus subtypes is shown on the left. A check mark indicates that binding, neutralization activity, or in vivo protection have been demonstrated; otherwise, a cross is shown.

HA head binders

We will return later to stem binders but now focus on exciting new developments on bnAbs to the head region. It was not clear initially whether bnAbs could be found against the head, as that region is so hypervariable. However, a number of groups have now identified antibodies to the head that have greater breadth than the isolate-specific antibodies normally elicited by natural infection and vaccination. In 2009, the Takada group reported an antibody (S139/1) that cross-reacts with different influenza virus subtypes (H1, H2, H3, H5, H9, and H13) close to antigenic site B46. A flurry of papers in 2011 also reported on head antibodies that have considerable breadth within the H1 subtype (e.g. 5J847 and CH6548), and an exceptional pan-H3 antibody F045–092 that also had some activity against H1, H2, H5, and H13 subtypes49 (Fig. 2). These head antibodies interact with the conserved RBS by inserting their H3 or H2 complementarity determining loops (CDRs) into the same site as the sialic acid receptor, explaining their breadth. Perhaps the archetype antibody against the RBS is C05, which essentially uses a single, long CDR H3 to engage the RBS, thereby minimizing contact with the hypervariable residues that surround the RBS50 (Fig. 2). Indeed, the most effective of these bnAbs to the RBS have smaller binding footprints that emulate to some extent the limited surface engaged by the sialic acid receptor51. Despite the quite different angles of approach of these antibodies to the HA surface, analysis of CH6548, CH6752, 5J853, F045–09251, 641 I–954, and H252654 revealed that a dipeptide corresponding to an aspartic acid-hydrophobic motif was the primary antibody motif for recognition of the receptor binding site54, 55. Specifically, this dipeptide is located at the tip of the CDR loop that is inserted into the RBS. This observation further underscores that it is feasible to target the RBS for vaccine and therapeutic design.

Design of a more universal vaccine

A number of different families of antibodies have now been found to target the stem. Originally, most of these antibodies were from the VH1–69 germline family, including CR6261 and CR9114, with a very hydrophobic CDR H2 that inserts two hydrophobic residues (Ile, Phe) at the tip into hydrophobic stem pockets. A conserved Tyr residue in CDR H3 provides the basis for the interaction with the HA stem. Together, these recognition elements, arising from CDR H2 (IF) and H3 (Y), form the so-called IFY motif56, 57. Two other stem-binding bnAbs, FI6v344 and 39.2958 of the VH3–30 germline family, which bind the HA at a very different angle and use both light and heavy chains for interaction, compared to VH1–69, where the heavy chain makes the majority, if not all, of the stem interactions. Nonetheless, VH1–69 and VH3–30 antibodies have very similar binding footprints and fill the same set of hydrophobic pockets in the HA stem59. However, the VH3–30 family uses an extended CDR H3 to occupy the same pockets as the IFY motif of the VH1–69 antibodies. More recently, a number of vaccine-induced antibodies that arise from other heavy chain germline families have been shown to target this same site60. Thus, these finding have led to the concept of a headless HA as an immunogen that would focus the immune response on the HA stem and away from the more immunogenic head region61.

The main problem was how to design and engineer such a construct, given that the headless HA region comprises three segments: the N and C- terminal regions of the HA1 chain, and the entire HA2 chain. In 2015, two such designs, namely mini-HA and headless HA, were reported by Janssen62 and the NIH Vaccine Research Institute (VRC)63, respectively (Fig. 3a). These engineered immunogens were tested in mice, monkeys and ferrets. The immunized animals survived challenge with a lethal dose of virus, thus providing the all-important proof-of-principle that a protective response could indeed be generated against the HA stem by vaccination. Furthermore, the response was relatively broad: for example, in the immunization studies with the mini-HA #4900 design, a heterologous response was produced in mice against an H1 subtype (A/Puerto Rico/8/1934) that was quite distant from the template (A/Brisbane/59/2007) used for design of the immunogen62. Heterosubtypic responses were also elicited against viruses from another subtype (H5, A/Hong Kong/156/1997). Similar results were observed in cynomolgus monkeys: the mini-HA was able to protect against both heterologous and heterosubtypic viruses, in contrast to a trivalent seasonal vaccine that could generate only strain-specific responses62. Passive protection against heterosubtypic challenge (H5), by introducing serum from the immunized mice into naïve mice, was also observed. The VRC headless HA construct was also displayed on ferritin nanoparticles particles to enhance its immunogenicity63 (Fig. 3b).

Figure 3. HA stem immunogen design.

Figure 3

(a) Structures of stem-binding bnAbs (CR6261 or CR9114) in complex with HA (PDB 3GBM)40, Gen4 HA-SS construct (PDB 5C0S)63, and mini-HA #4900 construct (PDB 5CJQ)62 are shown. (b) EM reconstruction of a nanoparticle that displays the Gen6 HA-SS construct (EMDB EMD-6332)63. (c–d) The structural conformations of HA, Gen4 HA-SS (without the trimerizing gp41 peptide region), and mini-HA #4900 (without the trimerizing GCN4 region) are aligned by minimizing their RMSD. (c) Alignment was performed on a single protomer. (d) Alignment was also performed on the trimeric form. Although the protomers from both Gen4 HA-SS and mini-HA #4900 align well with HA, their trimeric forms adopt a more open, splayed conformation at the base as compared to native full-length HA.

Crystal structures of both mini-HA and headless HA designs indicated some splaying of the headless HA trimers at the base compared to the full-length HA (Fig. 3c–d), which is likely attributable to the engineering that involved additional trimerization domains (GCN462 or a trimerizing peptide from HIV-1 gp4163) to help stabilize the triple helices in the center of the stem. Notwithstanding, the neutralizing epitopes were faithfully preserved in these designs, as indicated by their strong binding to known bnAbs, such as CR6261 and CR9114.

Besides the mini-HA and VRC headless HA constructs, other stem-based immunogens have been designed and reported64, 65, 66, 67, 68, 69. Another proposed approach to elicit heterologous and heterosubtypic immunity to the HA stem was the use of HA chimeras, where head domains from different subtypes are fused with a single stem domain, to direct the antibody response to the conserved stem region by sequential immunization with different chimeras70, 71.

Overall, these stem immunogens are currently the most promising for development of a more universal vaccine and current work is in progress to increase the breadth of the response by further design.

Design of small proteins and peptides against the HA stem domain

Shortly after the first structures of stem bnAbs were determined, Baker and colleagues designed small proteins, inspired by the interactions between the antibodies and the HA stem. The concept was to select side chains to fill some of same pockets in the HA stem targeted by antibodies, and then identify protein scaffolds onto which those amino-acid side chains could be attached in an appropriate conformation and configuration to optimize binding. Two such designs, HB36 and HB80, bound to H1 and H5 HAs with low nanomolar affinity. The crystal structure of HB36 in complex with 1918/H1 HA showed the remarkable fidelity of the design72 (Fig. 4). The designs were then further optimized to produce a 51-residue protein (HB80.4) that was able to bind to all influenza A group 1 HAs and neutralize H1N1 viruses with potencies akin to the best bnAbs73. The most recent advance in these small protein designs came from using a massively parallel approach, whereby 22,600 mini-proteins with different backbone scaffolds of 37–43 residues were screened against influenza HA74. Binders with very high affinity (Kd < 10 nM) and stability (Tm > 95 ºC) were identified and, importantly, those designed proteins were not found to be immunogenic even after repeated injections in mice. One mini-protein design, HB1.6928.2.3, is smaller than the previous designs (Fig. 4) and represents an excellent alternative to bnAbs for prophylaxis and therapy.

Figure 4. Protein and peptide design that target the HA stem region.

Figure 4

Four different designs that target the HA stem region, namely HB36.3 (PDB 3R2X)72, HB80.4 (PDB 4EEF)73, HB1.6928.2.3 (PDB 5VLI)74, and P7 (PDB 5W6T)59, along with CR9114 Fab (PDB 3GBM)40 are shown in burgundy. HA is colored white. Their binding surfaces are colored lime in the bottom panel. Figure inspired in part from Whitehead 201775.

In another recent breakthrough, small peptides were designed and characterized against this same site in the HA stem, also based on how bnAbs target the stem, in a collaboration between Janssen and our laboratory59. Using the CDR interacting loops of two bnAbs FI6v3 and CR9114 as templates, cyclic peptides that also incorporated non-proteinogenic amino acids were designed to fill the same hydrophobic pockets in the HA stem as the stem-binding bnAbs59. The crystal structure of one of these peptides, P7, confirmed that it indeed targets exactly the same highly conserved region in the stem as the bnAbs. These 11-residue peptides bind with low nanomolar affinity to group 1 HAs and also inhibit the low-pH induced conformational changes that lead to membrane fusion. This design of a peptide mimic of antibody loops is perhaps the most successful design of an antibody surrogate75.

Design of a small protein against the HA head

The mode of binding of bnAbs to the HA head has also inspired design of small proteins that mimic the key interactions of antibody C0550 with the RBS. The peptide binding loop from C05 H3 was the basis for the design, where a scaffold (cystatin) that could incorporate the sequence of this loop in a similar conformation was used as the template76 (Fig. 5). The design was further optimized by fusing three of these small protein binders with a natural homotrimer. In this way, the design (HSB.2A) matched the geometry of the HA trimer and substantially increased the avidity and reduced the off-rate by more than three-orders of magnitude. HSB.2A also had similar breadth and potency as C05 IgG and protected mice in a challenge study. This study now also provides further proof-of-concept that the information derived from how bnAbs target the RBS can be used to design a small protein that inhibits influenza virus.

Figure 5. Protein and peptide design that targets the HA receptor-binding site.

Figure 5

(a) Crystal structures of HA-C05 Fab (PDB 4FP8)50 and HA-HSB.2A76 complexes are shown. C05 Fab and HSB.2A are colored burgundy. (b) C05 and HSB.2A are colored pink. The loop region that interacts with the HA receptor-binding site is colored red. HA is colored white. HA receptor-binding site is colored light blue.

Small molecule antivirals against the HA stem

On the small molecule front, a small molecule named tert-butyl hydroquinone (TBHQ) had been shown to bind influenza HAs (with Kd of 5 to 50 μM) and inhibit its fusion activity77. The crystal structure of TBHQ bound to H3 and H14 HAs revealed that it interacted with a hydrophobic pocket further up the stem region than the regions targeted by the bnAbs, but could still stabilize protomer interactions in the trimer, accounting for its inhibition of membrane fusion78.

In 2017, the crystal structure of a broad-spectrum antiviral that has been sold as an over-the-counter drug in some countries since 1990, in Russia as Arbidol and in China as Enerxin, was determined in complex with HA79. This drug is currently manufactured by Pharmstandard in Russia. Our laboratory found that Arbidol indeed interacts with influenza HA and binds in the pocket that was only partially occupied by TBHQ78 (Figs. 1 and 6). This compound has modest binding affinity (Kd ~40–100 μM) to group 1 and group 2 influenza A viruses, and also acts as a fusion inhibitor. Crystal structures of H3 and H7 HAs with Arbidol showed that a water molecule was also present in the binding pocket79. Structure-based design of Arbidol analogs to incorporate a hydroxyl at the position corresponding to the water molecule improved binding to 90–500 nM, depending on the HA80. Thus, Arbidol analogs represent promising lead compounds for further improvement of drug binding and stability.

Figure 6. Inhibition of HA-mediated membrane fusion by small molecules.

Figure 6

The structural basis of two HA-mediated membrane fusion inhibitors, namely TBHQ (left, PDB 3EYK)78 and Arbidol (right, PDB 5T6S)79, has been characterized. HA1 is colored grey. HA2 is colored teal. TBHQ and Arbidol are shown as spheres representation and colored by atom type (yellow: carbon, red: oxygen, blue: nitrogen). The receptor-binding site and CR9114 epitope are colored lime and pink, respectively. The Cα of each resistance mutation is shown as a purple sphere. The change in pH of fusion, as measured by hemolysis assay81, 82, is indicated for each resistance mutation.

Some resistance mutations have been described that do not seem to directly interfere with either TBHQ or Arbidol binding81, 82 (Fig. 6). Instead, these mutations alter the pH of fusion and slightly raise the susceptible pH for triggering the conformational rearrangements that lead to fusion. Thus, increasing the affinity of Arbidol may make it more difficult for the virus to escape. Such inhibitors now provide yet another target for therapeutic development and further options for combating influenza virus.

Conclusions

In the last ten years, immense progress has been made on developing more effective strategies to combat influenza virus either by vaccination or by therapy. The discovery of human bnAbs to influenza virus proved that it was indeed possible to contemplate design of a more universal vaccine. The determination of a large number of bnAb structures against the HA head and stem has enabled common modes of recognition to be deciphered, even if the antibodies differ in their germline origin and angle of approach to the HA surface33, 55, 83. These bnAbs therefore provide a blueprint for designing candidate immunogens that focus the immune response against the HA stem and the RBS. The headless HA designs and their characterization have convincingly demonstrated the feasibility of this approach.

These bnAbs have also inspired design of small proteins and peptides to specifically target the HA and emulate the neutralization properties and mechanisms of the bnAbs. The finding that small molecules can further target an additional conserved hydrophobic pocket in the HA stem provides yet another opportunity for further design of small molecules as therapeutics against influenza virus. The next few years therefore hold great promise both for design of new vaccines and therapies against influenza virus and hopefully these will be realized before any new pandemic emerges on the scale of the devastating H1N1 1918 influenza virus.

Acknowledgments

We appreciate support from NIH R56 AI127371 (I.A.W) and a Croucher Foundation Fellowship (N.C.W.). The authors have no competing financial interests.

References

  • 1.Jefferson T, et al. Neuraminidase inhibitors for preventing and treating influenza in healthy adults and children. Cochrane Database Syst Rev. 2014:CD008965. doi: 10.1002/14651858.CD008965.pub3. [DOI] [PubMed] [Google Scholar]
  • 2.Tong S, et al. A distinct lineage of influenza A virus from bats. Proc Natl Acad Sci U S A. 2012;109:4269–4274. doi: 10.1073/pnas.1116200109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tong S, et al. New world bats harbor diverse influenza A viruses. PLoS Pathog. 2013;9:e1003657. doi: 10.1371/journal.ppat.1003657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wilson IA, Skehel JJ, Wiley DC. Structure of the haemagglutinin membrane glycoprotein of influenza virus at 3 Å resolution. Nature. 1981;289:366–373. doi: 10.1038/289366a0. [DOI] [PubMed] [Google Scholar]
  • 5.Wiley DC, Wilson IA, Skehel JJ. Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature. 1981;289:373–378. doi: 10.1038/289373a0. [DOI] [PubMed] [Google Scholar]
  • 6.Rogers GN, Paulson JC, Daniels RS, Skehel JJ, Wilson IA, Wiley DC. Single amino acid substitutions in influenza haemagglutinin change receptor binding specificity. Nature. 1983;304:76–78. doi: 10.1038/304076a0. [DOI] [PubMed] [Google Scholar]
  • 7.Weis W, Brown JH, Cusack S, Paulson JC, Skehel JJ, Wiley DC. Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature. 1988;333:426–431. doi: 10.1038/333426a0. [DOI] [PubMed] [Google Scholar]
  • 8.Chen J, Lee KH, Steinhauer DA, Stevens DJ, Skehel JJ, Wiley DC. Structure of the hemagglutinin precursor cleavage site, a determinant of influenza pathogenicity and the origin of the labile conformation. Cell. 1998;95:409–417. doi: 10.1016/s0092-8674(00)81771-7. [DOI] [PubMed] [Google Scholar]
  • 9.Ringe RP, et al. Cleavage strongly influences whether soluble HIV-1 envelope glycoprotein trimers adopt a native-like conformation. Proc Natl Acad Sci U S A. 2013;110:18256–18261. doi: 10.1073/pnas.1314351110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bullough PA, Hughson FM, Treharne AC, Ruigrok RW, Skehel JJ, Wiley DC. Crystals of a fragment of influenza haemagglutinin in the low pH induced conformation. J Mol Biol. 1994;236:1262–1265. doi: 10.1016/0022-2836(94)90027-2. [DOI] [PubMed] [Google Scholar]
  • 11.Stevens J, Corper AL, Basler CF, Taubenberger JK, Palese P, Wilson IA. Structure of the uncleaved human H1 hemagglutinin from the extinct 1918 influenza virus. Science. 2004;303:1866–1870. doi: 10.1126/science.1093373. [DOI] [PubMed] [Google Scholar]
  • 12.Gamblin SJ, et al. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science. 2004;303:1838–1842. doi: 10.1126/science.1093155. [DOI] [PubMed] [Google Scholar]
  • 13.Xu R, McBride R, Paulson JC, Basler CF, Wilson IA. Structure, receptor binding, and antigenicity of influenza virus hemagglutinins from the 1957 H2N2 pandemic. J Virol. 2010;84:1715–1721. doi: 10.1128/JVI.02162-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Liu J, et al. Structures of receptor complexes formed by hemagglutinins from the Asian Influenza pandemic of 1957. Proc Natl Acad Sci U S A. 2009;106:17175–17180. doi: 10.1073/pnas.0906849106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Xu R, Ekiert DC, Krause JC, Hai R, Crowe JE, Jr, Wilson IA. Structural basis of preexisting immunity to the 2009 H1N1 pandemic influenza virus. Science. 2010;328:357–360. doi: 10.1126/science.1186430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Zhang W, et al. Crystal structure of the swine-origin A (H1N1)-2009 influenza A virus hemagglutinin (HA) reveals similar antigenicity to that of the 1918 pandemic virus. Protein Cell. 2010;1:459–467. doi: 10.1007/s13238-010-0059-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang H, Carney P, Stevens J. Structure and Receptor binding properties of a pandemic H1N1 virus hemagglutinin. PLoS Curr. 2010;2:RRN1152. doi: 10.1371/currents.RRN1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Stevens J, Blixt O, Tumpey TM, Taubenberger JK, Paulson JC, Wilson IA. Structure and receptor specificity of the hemagglutinin from an H5N1 influenza virus. Science. 2006;312:404–410. doi: 10.1126/science.1124513. [DOI] [PubMed] [Google Scholar]
  • 19.Ha Y, Stevens DJ, Skehel JJ, Wiley DC. X-ray structures of H5 avian and H9 swine influenza virus hemagglutinins bound to avian and human receptor analogs. Proc Natl Acad Sci U S A. 2001;98:11181–11186. doi: 10.1073/pnas.201401198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Xu R, et al. Preferential recognition of avian-like receptors in human influenza A H7N9 viruses. Science. 2013;342:1230–1235. doi: 10.1126/science.1243761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Shi Y, et al. Structures and receptor binding of hemagglutinins from human-infecting H7N9 influenza viruses. Science. 2013;342:243–247. doi: 10.1126/science.1242917. [DOI] [PubMed] [Google Scholar]
  • 22.Yang H, Carney PJ, Chang JC, Villanueva JM, Stevens J. Structural analysis of the hemagglutinin from the recent 2013 H7N9 influenza virus. J Virol. 2013;87:12433–12446. doi: 10.1128/JVI.01854-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhang H, et al. A human-infecting H10N8 influenza virus retains a strong preference for avian-type receptors. Cell Host Microbe. 2015;17:377–384. doi: 10.1016/j.chom.2015.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yang H, Carney PJ, Chang JC, Villanueva JM, Stevens J. Structure and receptor binding preferences of recombinant hemagglutinins from avian and human H6 and H10 influenza A virus subtypes. J Virol. 2015;89:4612–4623. doi: 10.1128/JVI.03456-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang M, et al. Structural basis for preferential avian receptor binding by the human-infecting H10N8 avian influenza virus. Nat Commun. 2015;6:5600. doi: 10.1038/ncomms6600. [DOI] [PubMed] [Google Scholar]
  • 26.Tzarum N, et al. Structure and receptor binding of the hemagglutinin from a human H6N1 influenza virus. Cell Host Microbe. 2015;17:369–376. doi: 10.1016/j.chom.2015.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wang F, et al. Adaptation of avian influenza A (H6N1) virus from avian to human receptor-binding preference. EMBO J. 2015;34:1661–1673. doi: 10.15252/embj.201590960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Song H, et al. Avian-to-human receptor-binding adaptation by influenza A virus hemagglutinin H4. Cell Rep. 2017;20:1201–1214. doi: 10.1016/j.celrep.2017.07.028. [DOI] [PubMed] [Google Scholar]
  • 29.Tzarum N, McBride R, Nycholat CM, Peng W, Paulson JC, Wilson IA. Unique structural features of influenza virus H15 hemagglutinin. J Virol. 2017;91:e00046–00017. doi: 10.1128/JVI.00046-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang Q, Tian X, Chen X, Ma J. Structural basis for receptor specificity of influenza B virus hemagglutinin. Proc Natl Acad Sci U S A. 2007;104:16874–16879. doi: 10.1073/pnas.0708363104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sun X, et al. Bat-derived influenza hemagglutinin H17 does not bind canonical avian or human receptors and most likely uses a unique entry mechanism. Cell Rep. 2013;3:769–778. doi: 10.1016/j.celrep.2013.01.025. [DOI] [PubMed] [Google Scholar]
  • 32.Lu X, et al. Structure and receptor binding specificity of hemagglutinin H13 from avian influenza A virus H13N6. J Virol. 2013;87:9077–9085. doi: 10.1128/JVI.00235-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wu NC, Wilson IA. A perspective on the structural and functional constraints for immune evasion: insights from influenza virus. J Mol Biol. 2017;429:2694–2709. doi: 10.1016/j.jmb.2017.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cherry JL, Lipman DJ, Nikolskaya A, Wolf YI. Evolutionary dynamics of N-glycosylation sites of influenza virus hemagglutinin. PLoS Curr. 2009;1:RRN1001. doi: 10.1371/currents.RRN1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nobusawa E, Aoyama T, Kato H, Suzuki Y, Tateno Y, Nakajima K. Comparison of complete amino acid sequences and receptor-binding properties among 13 serotypes of hemagglutinins of influenza A viruses. Virology. 1991;182:475–485. doi: 10.1016/0042-6822(91)90588-3. [DOI] [PubMed] [Google Scholar]
  • 36.Air GM. Sequence relationships among the hemagglutinin genes of 12 subtypes of influenza A virus. Proc Natl Acad Sci U S A. 1981;78:7639–7643. doi: 10.1073/pnas.78.12.7639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kashyap AK, et al. Combinatorial antibody libraries from survivors of the Turkish H5N1 avian influenza outbreak reveal virus neutralization strategies. Proc Natl Acad Sci U S A. 2008;105:5986–5991. doi: 10.1073/pnas.0801367105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Throsby M, et al. Heterosubtypic neutralizing monoclonal antibodies cross-protective against H5N1 and H1N1 recovered from human IgM+ memory B cells. PLoS One. 2008;3:e3942. doi: 10.1371/journal.pone.0003942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sui J, et al. Structural and functional bases for broad-spectrum neutralization of avian and human influenza A viruses. Nat Struct Mol Biol. 2009;16:265–273. doi: 10.1038/nsmb.1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ekiert DC, et al. Antibody recognition of a highly conserved influenza virus epitope. Science. 2009;324:246–251. doi: 10.1126/science.1171491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Okuno Y, Isegawa Y, Sasao F, Ueda S. A common neutralizing epitope conserved between the hemagglutinins of influenza A virus H1 and H2 strains. J Virol. 1993;67:2552–2558. doi: 10.1128/jvi.67.5.2552-2558.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dreyfus C, Ekiert DC, Wilson IA. Structure of a classical broadly neutralizing stem antibody in complex with a pandemic H2 influenza virus hemagglutinin. J Virol. 2013;87:7149–7154. doi: 10.1128/JVI.02975-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ekiert DC, et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science. 2011;333:843–850. doi: 10.1126/science.1204839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Corti D, et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science. 2011;333:850–856. doi: 10.1126/science.1205669. [DOI] [PubMed] [Google Scholar]
  • 45.Dreyfus C, et al. Highly conserved protective epitopes on influenza B viruses. Science. 2012;337:1343–1348. doi: 10.1126/science.1222908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yoshida R, et al. Cross-protective potential of a novel monoclonal antibody directed against antigenic site B of the hemagglutinin of influenza A viruses. PLoS Pathog. 2009;5:e1000350. doi: 10.1371/journal.ppat.1000350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Krause JC, Tsibane T, Tumpey TM, Huffman CJ, Basler CF, Crowe JE., Jr A broadly neutralizing human monoclonal antibody that recognizes a conserved, novel epitope on the globular head of the influenza H1N1 virus hemagglutinin. J Virol. 2011;85:10905–10908. doi: 10.1128/JVI.00700-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Whittle JR, et al. Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin. Proc Natl Acad Sci U S A. 2011;108:14216–14221. doi: 10.1073/pnas.1111497108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ohshima N, Iba Y, Kubota-Koketsu R, Asano Y, Okuno Y, Kurosawa Y. Naturally occurring antibodies in humans can neutralize a variety of influenza virus strains, including H3, H1, H2, and H5. J Virol. 2011;85:11048–11057. doi: 10.1128/JVI.05397-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ekiert DC, et al. Cross-neutralization of influenza A viruses mediated by a single antibody loop. Nature. 2012;489:526–532. doi: 10.1038/nature11414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lee PS, et al. Receptor mimicry by antibody F045–092 facilitates universal binding to the H3 subtype of influenza virus. Nat Commun. 2014;5:3614. doi: 10.1038/ncomms4614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Schmidt AG, et al. Preconfiguration of the antigen-binding site during affinity maturation of a broadly neutralizing influenza virus antibody. Proc Natl Acad Sci U S A. 2013;110:264–269. doi: 10.1073/pnas.1218256109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hong M, et al. Antibody recognition of the pandemic H1N1 Influenza virus hemagglutinin receptor binding site. J Virol. 2013;87:12471–12480. doi: 10.1128/JVI.01388-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schmidt AG, et al. Viral receptor-binding site antibodies with diverse germline origins. Cell. 2015;161:1026–1034. doi: 10.1016/j.cell.2015.04.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lee PS, Wilson IA. Structural characterization of viral epitopes recognized by broadly cross-reactive antibodies. Curr Top Microbiol Immunol. 2015;386:323–341. doi: 10.1007/82_2014_413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lang S, Xie J, Zhu X, Wu NC, Lerner RA, Wilson IA. Antibody 27F3 broadly targets influenza A group 1 and 2 hemagglutinins through a further variation in VH1–69 antibody orientation on the HA stem. Cell Rep. 2017;20:2935–2943. doi: 10.1016/j.celrep.2017.08.084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Avnir Y, et al. Molecular signatures of hemagglutinin stem-directed heterosubtypic human neutralizing antibodies against influenza A viruses. PLoS Pathog. 2014;10:e1004103. doi: 10.1371/journal.ppat.1004103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Nakamura G, et al. An in vivo human-plasmablast enrichment technique allows rapid identification of therapeutic influenza A antibodies. Cell Host Microbe. 2013;14:93–103. doi: 10.1016/j.chom.2013.06.004. [DOI] [PubMed] [Google Scholar]
  • 59.Kadam RU, et al. Potent peptidic fusion inhibitors of influenza virus. Science. 2017;358:496–502. doi: 10.1126/science.aan0516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Joyce MG, et al. Vaccine-induced antibodies that neutralize group 1 and group 2 influenza A viruses. Cell. 2016;166:609–623. doi: 10.1016/j.cell.2016.06.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Steel J, et al. Influenza virus vaccine based on the conserved hemagglutinin stalk domain. mBio. 2010;1:e00018–00010. doi: 10.1128/mBio.00018-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Impagliazzo A, et al. A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science. 2015;349:1301–1306. doi: 10.1126/science.aac7263. [DOI] [PubMed] [Google Scholar]
  • 63.Yassine HM, et al. Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection. Nat Med. 2015;21:1065–1070. doi: 10.1038/nm.3927. [DOI] [PubMed] [Google Scholar]
  • 64.Valkenburg SA, et al. Stalking influenza by vaccination with pre-fusion headless HA mini-stem. Sci Rep. 2016;6:22666. doi: 10.1038/srep22666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Wohlbold TJ, Nachbagauer R, Margine I, Tan GS, Hirsh A, Krammer F. Vaccination with soluble headless hemagglutinin protects mice from challenge with divergent influenza viruses. Vaccine. 2015;33:3314–3321. doi: 10.1016/j.vaccine.2015.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mallajosyula VV, et al. Influenza hemagglutinin stem-fragment immunogen elicits broadly neutralizing antibodies and confers heterologous protection. Proc Natl Acad Sci U S A. 2014;111:E2514–2523. doi: 10.1073/pnas.1402766111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mallajosyula VV, et al. Hemagglutinin sequence conservation guided stem immunogen design from influenza A H3 subtype. Front Immunol. 2015;6:329. doi: 10.3389/fimmu.2015.00329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bommakanti G, et al. Design of an HA2-based Escherichia coli expressed influenza immunogen that protects mice from pathogenic challenge. Proc Natl Acad Sci U S A. 2010;107:13701–13706. doi: 10.1073/pnas.1007465107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bommakanti G, et al. Design of Escherichia coli-expressed stalk domain immunogens of H1N1 hemagglutinin that protect mice from lethal challenge. J Virol. 2012;86:13434–13444. doi: 10.1128/JVI.01429-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Krammer F, et al. Assessment of influenza virus hemagglutinin stalk-based immunity in ferrets. J Virol. 2014;88:3432–3442. doi: 10.1128/JVI.03004-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Krammer F, Pica N, Hai R, Margine I, Palese P. Chimeric hemagglutinin influenza virus vaccine constructs elicit broadly protective stalk-specific antibodies. J Virol. 2013;87:6542–6550. doi: 10.1128/JVI.00641-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Fleishman SJ, et al. Computational design of proteins targeting the conserved stem region of influenza hemagglutinin. Science. 2011;332:816–821. doi: 10.1126/science.1202617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Whitehead TA, et al. Optimization of affinity, specificity and function of designed influenza inhibitors using deep sequencing. Nat Biotechnol. 2012;30:543–548. doi: 10.1038/nbt.2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chevalier A, et al. Massively parallel de novo protein design for targeted therapeutics. Nature. 2017;550:74–79. doi: 10.1038/nature23912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Whitehead TA. A peptide mimic of an antibody. Science. 2017;358:450–451. doi: 10.1126/science.aap9608. [DOI] [PubMed] [Google Scholar]
  • 76.Strauch EM, et al. Computational design of trimeric influenza-neutralizing proteins targeting the hemagglutinin receptor binding site. Nat Biotechnol. 2017;35:667–671. doi: 10.1038/nbt.3907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bodian DL, Yamasaki RB, Buswell RL, Stearns JF, White JM, Kuntz ID. Inhibition of the fusion-inducing conformational change of influenza hemagglutinin by benzoquinones and hydroquinones. Biochemistry. 1993;32:2967–2978. doi: 10.1021/bi00063a007. [DOI] [PubMed] [Google Scholar]
  • 78.Russell RJ, et al. Structure of influenza hemagglutinin in complex with an inhibitor of membrane fusion. Proc Natl Acad Sci U S A. 2008;105:17736–17741. doi: 10.1073/pnas.0807142105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kadam RU, Wilson IA. Structural basis of influenza virus fusion inhibition by the antiviral drug Arbidol. Proc Natl Acad Sci U S A. 2017;114:206–214. doi: 10.1073/pnas.1617020114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Wright ZVF, Wu NC, Kadam RU, Wilson IA, Wolan DW. Structure-based optimization and synthesis of antiviral drug Arbidol analogues with significantly improved affinity to influenza hemagglutinin. Bioorg Med Chem Lett. 2017;27:3744–3748. doi: 10.1016/j.bmcl.2017.06.074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Leneva IA, Russell RJ, Boriskin YS, Hay AJ. Characteristics of arbidol-resistant mutants of influenza virus: implications for the mechanism of anti-influenza action of arbidol. Antiviral Res. 2009;81:132–140. doi: 10.1016/j.antiviral.2008.10.009. [DOI] [PubMed] [Google Scholar]
  • 82.Hoffman LR, Kuntz ID, White JM. Structure-based identification of an inducer of the low-pH conformational change in the influenza virus hemagglutinin: irreversible inhibition of infectivity. J Virol. 1997;71:8808–8820. doi: 10.1128/jvi.71.11.8808-8820.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Xu R, Krause JC, McBride R, Paulson JC, Crowe JE, Jr, Wilson IA. A recurring motif for antibody recognition of the receptor-binding site of influenza hemagglutinin. Nat Struct Mol Biol. 2013;20:363–370. doi: 10.1038/nsmb.2500. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES