Article

Viral Receptor-Binding Site Antibodies with Diverse Germline Origins Graphical Abstract

Authors Aaron G. Schmidt, Matthew D. Therkelsen, ..., Barton F. Haynes, Stephen C. Harrison

Correspondence [email protected]

In Brief A structure-based signature motif found in many human antibodies indicates recogition of the receptor-binding site of influenza virus hemagglutinin. These antibodies arise from diverse germline origins and affinity maturation pathways and suggest potential routes to a universal flu vaccine.

Highlights d

Identified a signature motif for influenza RBS-directed antibodies

d

Influenza RBS-antibodies are nearly unrestricted in gene usage

d

Multiple donors have RBS-directed antibodies with receptor mimicry

d

Viral resistance to one RBS antibody does not confer resistance to all

Schmidt et al., 2015, Cell 161, 1–9 May 21, 2015 ª2015 Elsevier Inc. http://dx.doi.org/10.1016/j.cell.2015.04.028

Accession Numbers 4YK4 4YJZ

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Article Viral Receptor-Binding Site Antibodies with Diverse Germline Origins Aaron G. Schmidt,1 Matthew D. Therkelsen,1,6 Shaun Stewart,2,7 Thomas B. Kepler,3 Hua-Xin Liao,4 M. Anthony Moody,4 Barton F. Haynes,4 and Stephen C. Harrison1,5,* 1Laboratory

of Molecular Medicine, Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA Vaccines and Diagnostics, Cambridge MA, USA 3Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA 4Duke Human Vaccine Institute, Duke University Medical School, Durham, NC 27710, USA 5Howard Hughes Medical Institute, Boston, MA 02115, USA 6Present Address: Life Science Program, Purdue University, West Lafayette, IN 47907; 7Present Address: Global Biotherapeutics Technologies, Pfizer Inc., Cambridge, MA 02139 *Correspondence: [email protected] http://dx.doi.org/10.1016/j.cell.2015.04.028 2Novartis

SUMMARY

Vaccines for rapidly evolving pathogens will confer lasting immunity if they elicit antibodies recognizing conserved epitopes, such as a receptor-binding site (RBS). From characteristics of an influenza-virus RBS-directed antibody, we devised a signature motif to search for similar antibodies. We identified, from three vaccinees, over 100 candidates encoded by 11 different VH genes. Crystal structures show that antibodies in this class engage the hemagglutinin RBS and mimic binding of the receptor, sialic acid, by supplying a critical dipeptide on their projecting, heavy-chain third complementarity determining region. They share contacts with conserved, receptor-binding residues but contact different residues on the RBS periphery, limiting the likelihood of viral escape when several such antibodies are present. These data show that related modes of RBS recognition can arise from different germline origins and mature through diverse affinity maturation pathways. Immunogens focused on an RBS-directed response will thus have a broad range of B cell targets. INTRODUCTION For antibody protection against rapidly evolving pathogens, a conserved surface such as a viral receptor-binding site (RBS) is an appropriate target. Mutations at conserved sites generally compromise a critical step in the infectious cycle, lowering the frequency of escape from neutralization. A viral RBS is often poorly immunogenic, however, precisely by virtue of having evolved to evade a host immune response. Restrictions on antibody access can come from a recessed receptor-binding pocket or from a cryptic RBS, exposed only after a conformational rearrangement in the viral surface protein. Moreover, the surface area the RBS presents on most viruses is smaller than the footprint of a typical antibody, allowing escape through mutation of non-conserved, peripheral residues.

Despite these barriers, viral receptor sites do sometimes elicit antibodies that resist escape and neutralize a broad range of isolates (Burton et al., 2012; Corti and Lanzavecchia, 2013). For HIV-1, members of a class of RBS-directed antibodies known as ‘‘VRC01-like’’ mimic the binding ridge on the first Ig-like domain of the viral receptor, CD4, with its antibody heavy-chain homolog, the second heavy-chain complementarity determining region (CDR H2) (Wu et al., 2010). VRC01-like antibodies have received considerable attention because of the unusual breadth of their neutralizing activity. Only the heavy-chain gene segments VH12 and VH146 can readily encode them, however, because of the strong restrictions imposed by CD4 mimicry (Scheid et al., 2011; West et al., 2014). The VRC01-like antibodies also seem to require a large number of somatic mutations and generally appear only after several years of infection (Klein et al., 2013; Mascola and Haynes, 2013). The principal target of the adaptive immune response against influenza is the viral hemagglutinin (HA), one of two glycoproteins on the virion surface (Skehel and Wiley, 2000). HA mediates entry of the virus into a host cell by engaging its receptor, sialic acid, a key determinant of host tropism (Weis et al., 1988). HA is also the fusogen that catalyzes the merging of viral and cellular membranes necessary to establish infection (Harrison, 2008). Most HA-directed antibodies target its antigenically variable ‘‘head’’; many of them neutralize by interfering with receptor binding, and some block fusion by bridging adjacent heads in a trimer (Knossow et al., 2002; Wiley et al., 1981). These antibodies are generally strain specific, and seasonal drift of the virus leads readily to escape. Less frequently generated antibodies target a conserved patch on the ‘‘stem’’ of HA. These antibodies can have heterosubtypic neutralizing activities, but their potential repertoire is limited, as most derive from the same VH gene, VH169 (Corti et al., 2011; Ekiert et al., 2009; Sui et al., 2009). Moreover, they neutralize poorly, because dense packing of HA spikes on the virion surface impedes access to the stem. They primarily impart protection by limiting viral spread through a cell-mediated mechanism such as ADCC, and protection thus depends on the antibody subclass (Corti et al., 2011; DiLillo et al., 2014). These properties suggest that it may not be wise to focus work on ‘‘universal’’ influenza vaccines solely on stem antibodies. Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc. 1

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Figure 1. Properties of CH65-like Antibodies (A) Representative CDR H3s from antibodies identified from a lineage (top) and CDR H3s of orphan antibodies (bottom). The donor and VH gene usage are noted. The critical dipeptide that mimics sialic acid in the CDR H3 is boxed (or underlined) in red. Lineage 652 and antibody H2548 are separated from the rest of the list because of the length and positioning of the critical dipeptide in its CDR H3. (B and C) Lineage (B) and orphan (C) antibody VH gene summary. (D and E) Lineage (D) and orphan (E) antibody VL gene summary. (F) Critical residues present in CH65-like antibodies. The single amino acid codes and number of antibodies having the critical dipeptide are in parentheses. (G) Distribution of CDR H3 lengths from all antibodies. See also Figure S3 and Tables S2, S3, and S4.

Binding of the RBS-directed antibodies, CH65 and CH67, mimics contacts between HA and the influenza-virus receptor, sialic acid (Schmidt et al., 2013; Whittle et al., 2011). In effect, CH65 and CH67 are to influenza virus as VRC01 is to HIV-1. An aspartic acid side chain at the tip of the CDR H3 has the same hydrogen-bonding pattern as the sialic-acid carboxylate, and adjacent elements on CDR H3 have contacts that recapitulate those of the sialic-acid acetamido group. CH65 and CH67 neutralize a broad range of H1 influenza viruses. They were derived by isolation of single B cells and sequencing of their rearranged heavy- and light-chain genes in a blood sample from an individual (designated TIV01) administered the 2007-8 trivalent inactivated influenza vaccine (TIV) (Moody et al., 2011). The CH65 and CH67 sequences, two of a much larger number of mature antibodies identified in the original study, defined a simple lineage (‘‘clone 860’’) with one intermediate and with an unmutated common ancestor (UCA) at its root; the lineage included one other antibody (CH66), identical to CH65 save for a single amino-acid residue (Schmidt et al., 2013; Xu et al., 2014). 2 Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc.

We have summarized the properties of the CDR H3 in clone860 antibodies, and the interactions of CDR H3 with the RBS of influenza virus HA, with a ‘‘signature’’ for identifying potential RBS-directed antibodies from TIV vaccinees, and we have found, from three individuals, over 100 antibodies that conform to it. We have determined structures for two representative members of the set (in addition to CH65 and CH67 from clone 860), bound with the head of the H1 HA that was part of the 2007-8 vaccine. Unlike the broadly neutralizing antibodies that target the HA stem, RBS-directed antibodies appear to be relatively unrestricted in their gene usage. They include heavy chains encoded by eleven different VH genes. Many of them fall into distinct clonal lineages and thus have developed through diverse affinity maturation pathways. The characteristics of the sialicacid binding site on HA allow an antibody to approach from various directions, unlike the CD4 site on HIV gp120, and we show that four antibodies, for which structures with HA are known, have among them only nine common contacting residues on HA, all but one of which is also a sialic-acid contact. Indeed, a mutation selected for resistance to one of them does

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Table 1. Properties of CH65-like Antibodies Studied Biochemically or Structurally Antibody

Donor

CH65

TIV01

Lineage 860

No. of Members 3

VH

CDR H3 Length

Critical Dipeptide

Competes with CH67

12

19

VD

Y

CH67

TIV01

860

3

12

19

VD

Y

641 I-9

TIV01

641

19

459

19

VD, ID

Y

H671

TIV01

639

5

459

19

VD, VE

Y

H1825

TIV01

643

16

459

19

VD

Y Y

H1109

TIV01

1277

4

459

19

VD, VE

H1111

TIV01

687

5

459

19

VD, PD, TD

Y

H2227

TIV01

652

8

44

23

PD

Y

H2365

TIV21

orphan

N.A.

39

20

MD

N.A.

H2526

TIV24

orphan

N.A.

169

21

PD

Y

H2548

TIV24

orphan

N.A.

313

21

GD

Y

N.A., not-applicable. See also Table S4 for influenza strain reactivities of all CH65-like antibodies.

not confer resistance to another. We suggest that a small number of antibodies with sialic-acid like contacts should protect against infection by a wide range of influenza viruses and that immunogens to elicit them would be useful goals for influenza vaccine development. RESULTS CH65-like Signature Sequence We examined paired VH and VL sequences from four additional members (designated TIV04, TIV14, TIV21 and TIV24) of the cohort of subjects to which TIV01 belonged (Moody et al., 2011). Participants in the study were administered either the 2007-8 or the 2008-9 formulations of the trivalent inactivated seasonal influenza vaccine (TIV); the components of each vaccine and donor information are in Table S1. Plasmablasts from donors were sorted 7 days post-vaccination and influenza-specific antibodies identified, yielding a total of 251 unique antibody sequences. We searched this database for antibodies that might, like CH65, recapitulate sialic-acid contacts, using a signature based on sequences in lineage 860 and on interactions of the 860 antibodies with HA. The search criteria were: (i) a CDR H3 of 19 ± 4 residues; (ii) ‘‘X(D/E)’’ at a central position, where X is a hydrophobic amino acid; (iii) a cluster of aromatic residues between the X(D/E) motif and the C terminus of CDR H3, as those residues contribute to the projecting conformation of the loop in antibodies CH65 and CH67. We found 107 unique antibodies in the database that satisfied these criteria. Most (97%) of the 107 came from vaccinee TIV01, but two of the other four vaccinees also had at least one such antibody. About 65% of all flu-specific antibodies identified in the original study came from TIV01, partly explaining why that individual contributed such a large fraction of all the CH65-like antibodies we detected. A list of CDR H3 sequences for the 107 antibodies is in Figure 1A and Table S2. Properties of CH65-like Antibodies Isolated from TIV Donors The CH65-like signature search identified antibodies from eleven different VH families, with a clear overrepresentation of VH459 (Figures 1B and 1C). There was a strong bias for kappa light

chains, but we also found members of several lambda-chain gene families (Figures 1D and 1E). Despite diverse gene usage, many of the CH65-like antibodies encoded a valine-aspartic acid (VD) dipeptide at the tip of a CDR H3 nineteen amino acids in length (Figures 1F and 1G). All rearranged antibody genes encoding a CH65-like signature had a JH6 gene segment, which readily recombines to create a GAC codon. (The Asp-encoding, third-position variant, GAT, was nearly absent.) JH6 also encodes the cluster of tyrosines C-terminal to the VD dipeptide. Clonal Lineages of the CH65-like Antibodies Many of the antibodies that matched our CH65-like search criteria belonged to clonal lineages with unambiguously inferred maturation pathways and UCAs. We identified 22 distinct lineages comprising a total of 87 mature antibodies, all from TIV01. We show the clonal trees of some examples in Figure S1. We had 78 antibody sequences from the other four TIV donors, 42 of which fell into 13 lineages, but none had a CH65-like signature. We did, however, identify twenty CH65-like antibodies among the remaining 36 ‘‘orphans’’ (i.e., those for which there were no other lineage members among the sequences determined): 17 from TIV01, two from TIV24, and one from TIV21. Although TIV01 was evidently predisposed to produce CH65like antibodies, the presence of such antibodies in two other individuals shows that this kind of antibody is not a peculiarity of TIV01’s immune system. Structural Characterization of CH65-like Antibodies Shows Common Features of Receptor Mimicry We selected for biochemical and structural analysis one member from each of several TIV01 clonal lineages and the three orphan antibodies from the two other TIV donors. Our selection maximized gene diversity, varied CDR H3 length, and selected variants of the critical dipeptide in the middle of CDR H3 (Table 1). Because it included representative antibodies from several large clonal lineages, this set covered more than half (63/107) of the antibodies we found in our search. Lineage 641, isolated from TIV01, has 19 mature antibodies, which bind with high affinity (KD < 100 nM for the IgG) to a wide range of recombinant influenza H1 HAs, including Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc. 3

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Figure 2. Comparison of Receptor-Binding Site Antibodies (A–D) 641 I-9 (A) H2526 (B) CH67 (PDB 4HKX) (C) and 5J8 (PDB 4M5Z) (D) in complex with H1 Solomon Islands/03/2006 (for A–C) or H1 California/ 07/2009 HA (for D) (colored silver). The CDR H3 for each antibody is in magenta; their VH and VL domains are in blue and green, respectively. See also Table S3 for a list of antibody residues that contact HA.

pandemic A/California/04/2009 (Moody et al., 2011). It differs in gene usage from lineage 860, but its CDR H3 is identical in length and contains ‘‘VD’’ at its tip. We determined the crystal structure of a late intermediate in this lineage, 641 I-9, in complex with the HA head from H1 Solomon Islands/03/2006 (Figure 2A and Table S3). Like CH65 and CH67, 641 I-9 engages the RBS, and the critical VD dipeptide in CDR H3 mimics contacts made by the sialic acid receptor (Figures 3A and 3B). Unlike CH65 and CH67, 641 I-9 contacts HA only through its heavy chain, and the resulting footprint on HA is only 610 A˚2 of buried surface. A phage-displayed antibody, C05, has a similar footprint. It engages the influenza RBS with a long, 24 amino acid CDR H3, but it does so without mimicking receptor contacts (Ekiert et al., 2012). Different VH genes encode the two CH65-like antibodies (H2526 and H2547) identified from TIV24 (Table 1). These antibodies have variants of the dipeptide ‘‘VD,’’ at the tip of projecting CDR H3 loops that are longer than those in the 641 and 860 lineages. Both H2526 and H2547 compete with CH67 for HA binding (Table 1). We determined the crystal structure of H2526 in complex with the HA head of H1 Solomon Islands/ 03/2006. Its CDR H3 dipeptide motif, ‘‘PD,’’ indeed engages the receptor-binding pocket (Figure 2B and Table S3), but the orientation of the Fab with respect to HA is 180 away from those of CH67 and 641 I-9. It also has a larger footprint (936 A˚2) than do the other two (825 A˚2 and 610 A˚2 for CH67 and 641 I-9, respectively). The signature Asp, like those of CH67 and 641 I-9, accepts a hydrogen bond from the mainchain NH of residue 137, but the orientation of the Fab positions the carboxylate so that instead of receiving a second hydrogen bond from the side chain of conserved Ser/ Thr136, it receives one from Og of the less conserved Ser145 (Figure 3C). The main-chain NH of the Asp residue donates a hydrogen bond to the carbonyl of HA residue 135, the same group that accepts a hydrogen bond from the preceding valine main-chain NH in complexes with lineage 860 or 641 antibodies and from the acetamido NH of sialic acid. Like the valyl side chain of ‘‘VD’’ and the sialic-acid acetamido group, the proline of ‘‘PD’’ contacts Trp153 at the base of the RBS pocket and Leu194 at the rear. Its contacts are 4 Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc.

thus closely related to those of sialic acid, with less precise mimicry than the 860 and 641 lineage members. The H2526 heavy-chain gene derives from VH169, which encodes the heavy chain variable domain of most HAdirected, broadly neutralizing, stem antibodies. Previously described VH169 antibodies contact Trp153 in the RBS with Phe54 in CDR H2 (Xu et al., 2013). H2526 is the only example we know for a donor-derived, VH169 antibody that has sialic-acid like contacts. One other reported RBSbinding antibody, 5J8, with ‘‘PD’’ at the tip of its CDR H3, has RBS contacts very similar to those of H2526, despite quite different gene usage: VH4b, JH4 and Vl321, Jl2 or 3 (Hong et al., 2013; Krause et al., 2011) (Figures 2D and 3E). The aspartic acid side chain has the same hydrogen-bond pattern as do the aspartic acids in CH65 and 641 I-9, and like the valine in those antibodies, the proline in H2526 contacts Trp153 and Leu194. It lacks, however, a hydrogen bond with the mainchain carbonyl of residue 135. Antibody 5J8 appears to require lysine 133a, found in H1 isolates from much of the 20th century and again in the new pandemic H1, but not in recent seasonal H1s. The two crystal structures described here, our previously described structures of CH65 and CH67, and the published structure of 5J8 thus show that RBS-binding antibodies can derive from a variety of variabledomain genes and that more than one dipeptide sequence at the tip of the CDR H3 loop can satisfy the requirements for sialic acid-like contacts. Potential Variation of CDR H3 Dipeptide Sequences in CH65-like Antibodies We examined the range of sequence variation consistent with RBS contacts that recapitulate those of sialic acid. To determine which signature motifs resembling ‘‘VD’’ might be adequate, we replaced the two residues in CH67 and in 641 I-9 with a set of related dipeptides and measured binding of the variant Fabs with rHA (H1 Solomon Islands/03/2006) by biolayer interferometry. These positions indeed accommodate substantial variation (Table 2 and Figure S2). Mutation of the critical Asp to Ala eliminated binding, but Glu could substitute. Nearly all the mutants bound less tightly than did CH67 or 641 I-9, although ‘‘TD’’ had comparable affinity. Nonetheless, were any of the bindingcompatible motifs displayed at the tip of a comparably projecting CDR H3, somatic hypermutation could probably compensate, leading to affinities comparable to those of CH67 or 641 I-9.

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Figure 3. Comparison of Receptor-Binding Site Antibodies and Receptor Mimicry (A) Schematic of LSTc (modified from PDB 3UBE) docked into the RBS of H1 Solomon Islands/03/ 2006, highlighting the interaction between the carboxylate and acetamido groups with HA. (B–E) 641 I-9 Fab (B) H2526 (C) CH67 (D) and 5J8 (E) in complex with HA (colored silver). The CDR H3 for each antibody is colored magenta with the critical dipeptide and flanking residues colored yellow and in stick representation. Antibody and HA numbering follow their respective deposited PDB structures.

We therefore imagine that a large number of naive responses might converge on the same binding mode. Specificity of RBS-Directed Antibodies Most of the CH65-like antibodies we have identified in samples from the TIV cohort bind H1 Solomon Islands/03/2006, the H1 component of the 2007–2008 vaccine (Table S4). Several bind the B-component of the vaccine, among them, members of lineage 1261 and orphan antibody, H2365, from TIV01 and TIV21, respectively. These have the same gene usage (VH39 and Vk133) and nearly identical CDR H3 sequences (Figures S3A and S3B); these features might be common characteristics of type-B reactive CH65-like antibodies. We did not find any CH65-like antibodies specific for the H3 vaccine components, but one antibody each from lineages 639 and 1277 (out of 5 and 4 total mature antibodies, respectively) appeared to crossreact with H3 Johannesburg/33/1994. Among sequences in a published study of 153 antibodies with H3N2 specificities from a single individual (Okada et al., 2010), we found 12 candidate CH65-like antibodies, all VH349 with a 19aa long CDR H3 and the critical VD dipeptide (Figure S3C). Although they did not report structures, the authors did map these antibodies to the RBS (Okada et al., 2011). These data suggest that RBSdirected antibodies with H1, H3 or B specificities occur with relatively high frequency in the general population, even though only a small subset is likely to have heterosubtypic specificity. Resistance to RBS-Directed Antibodies Crystal structures of the two complexes presented here along with our previously characterized CH65 and CH67 complexes represent three different VH genes, two Vl genes and one Vk gene. The projecting CDR H3 has conserved contacts in the receptor binding site that are common to all four of these antibodies, while the remaining contacts with the periphery of the receptor pocket vary substantially (Figures 4A–4D, Table S5). Only nine HA residues interact with all members of the set (CH65, CH67, 641 I-9 and H2526), and of these nine, eight are either

conserved or are human receptor contacts (or both), making escape mutations in HA at these positions unlikely. We obtained a resistance mutation, G189D, by passaging H1 Solomon Islands/03/2006 in the presence of CH65 (Figure 4E). This escape mutant, while conferring resistance to CH65, had only a modest effect on neutralization by 641 I-9 (Figure 4F). These data, and the structures we have analyzed, suggest that no single mutation will allow escape from binding and neutralization by all antibodies that approach the RBS from different directions, without severely compromising fitness. We suggest that a small number of CH65-like antibodies such as the ones identified in this study and others would neutralize a wide range of H1 viruses. RBS-Directed Antibodies in Sera of TIV Vaccinees All antibodies described here were isolated from blood plasmablasts seven days post vaccination. Were they present in the serum? We carried out CH65 blocking experiments with sera from members of the TIV cohort, pre- and post-vaccination, and analyzed the vaccination-elicited increase in the titer of antibodies blocked by CH65 (Figure 5). We detected the largest increases in those individuals (TIV01, TIV21 and TIV24) from whom we found B cells expressing CH65-like signature antibodies. We cannot conclude that these antibodies all have a CH65-like signature sequence (indeed, some may bind the RBS periphery and not mimic the receptor at all), but the correlation of blocking activity with detection of CH65-like signatures does suggest that at least a component of the blocking activity reflects circulating antibodies interacting with HA in the receptor-binding pocket. DISCUSSION From the structures and other data presented here, we draw the following three broad conclusions. First, antibodies directed against the influenza virus RBS, with contacts that recapitulate many of those made by sialic acid, have a variety of potential germline origins. JH6 is the only shared germline feature among the antibodies we have characterized, and the 5J8 antibody uses JH4. Second, B cells expressing more than one type of RBS antibody can be present in a single individual, as shown by the various lineages and orphan antibodies isolated from TIV01. Third, these RBS antibodies may not be a particularly rare Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc. 5

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Table 2. Kinetic and Affinity Measurements of Mutants of the CH65-like Antibody Critical Dipeptide KD (mM)

ka*104 (1/Ms)

0.259 ± 0.06

20.32 ± 4.33

0.053 ± .004

1.79 ± 0.12

4.24 ± 0.27

0.076 ± .002

koff (1/s)

CH67 VD (wildtype) ID

8.5 ± 6.89

5.62 ± 4.43

0.478 ± 0.088

TD

MD

0.316 ± 0.02

12.5 ± 0.58

0.050 ± 0.001

VE

2.53 ± 0.14

3.03 ± 0.16

0.077 ± 0.002

IE

3.6 ± 1.20

6.3 ± 2.00

0.227 ± 0.024

AD

4.38 ± 0.76

3.05 ± 0.51

0.134 ± 0.007

ME

N.B.

N.B.

N.B.

LE

N.B.

N.B.

N.B.

WD

N.B.

N.B.

N.B.

VN

N.B.

N.B.

N.B.

DV

N.B.

N.B.

N.B.

AA

N.B.

N.B.

N.B.

641 I-9 VD (wildtype)

0.364 ± 0.01

5.53 ± 0.61

0.0213 ± 0.0002

ID

0.569 ± 0.02

6.34 ± 0.23

0.0361 ± 0.001 0.0518 ± 0.001

MD

1.14 ± 0.09

4.53 ± 0.35

VE

2.02 ± 0.21

5.43 ± 0.55

0.109 ± 0.003

IE

2.32 ± 0.15

5.14 ± 0.31

0.119 ± 0.002

ME

7.73 ± 2.44

3.74 ± 1.14

0.289 ± 0.024

The wildtype ‘‘VD’’ in the CDR H3 of CH67 and 641 I-9, from lineages 860 and 641, respectively were mutated to the listed dipeptide. Affinities were calculated by applying a 1:1 binding isotherm to data from biolayer interferometry, using global fit parameters in the vendor-supplied software, from titrations with at least four concentrations of H1 Solomon Islands/ 03/2006 head. N.B., no binding. See also Figure S2 for critical dipeptides in clonal lineages.

response, as B cells expressing them were present in three of the five individuals in the TIV cohort from whom paired sequence data were obtained, and as sequences corresponding to the signature motif are present in the published studies. The antibodies compared here (CH65, CH67, 641 I-9, H2526, and 5J8) approach the sialic-acid binding site from different directions and have quite different peripheral contacts. The first three mimic sialic acid rather closely; the last two, somewhat less perfectly, but still with enough interactions in common with sialic acid to restrict the range of escape mutations that would retain viral fitness. The degree of variability afforded by the shallowness of the sialic-acid pocket and its exposure on the outward-facing surface of HA implies that a resistance mutation consistent with retention of receptor affinity would probably affect only a subset of the antibodies in a similar mixture and that escape would require an unlikely coincidence of several mutations. An influenza virus vaccine that could elicit several different RBS-directed antibodies might then need less frequent redesign and less frequent administration. We propose that such a broadly neutralizing response might be a more practical first step to the goal of ‘‘universality’’ than one or more broadly neutralizing, heterosubtypic antibodies. 6 Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc.

The variety of germline origins that can lead to an RBSdirected antibody suggests that most individuals will have representatives in their naive repertoire. We can make a rough estimate of the proportion of germline antibodies that could, in principle, mature into CH65-like binding. In humans, the frequency of CDR H3 loops between 18 and 20 residues long is about 0.1. We estimate the likelihood that the tip of the CDR H3 will bear a hydrophobic residue and an aspartic acid, in that order, as (0.07)*(0.05), from the approximate frequency of appearance of those residues in protein sequences. (Aspartic acid at the second position would probably occur more than 5% of the time, as JH6 encodes the heavy-chain junction segment of about one-third of the human naive repertoire (Brezinschek et al., 1995) Thus, a signature or signature-like CDR H3 should occur about once in every 30,000 germline heavy chains. If 10% of all light-chain possibilities is also consistent with CH65-like binding, as suggested by the absence of evident light-chain preference in our data, then we get an overall frequency of about 1 in 300,000, high enough that there should be many suitable BCRs in a naive human repertoire. That is, the ‘‘raw material’’ should indeed be present for several different, RBS-directed antibodies with sialic acid-like contacts. The structures and data presented here show that many different gene rearrangements can evolve by somatic hypermutation to function equivalently as influenza virus RBS-directed antibodies. What modifications to HA as an immunogen might exploit this redundancy to make the RBS immunodominant? A viable strategy needs to be reasonably independent of previous exposure. For example, the RBS on the H1 component of the TIV vaccine was (by a criterion of B cell prevalence) an immunodominant epitope for TIV01, considerably less so for TIV21 and TIV24, and fully subdominant for TIV04 and TIV14. Because of the number of somatic mutations accumulated by the CH65-like antibodies we identified from this cohort, the naive response that gave rise to them probably came from some earlier exposure, either to a vaccine or to an infection. The notion of ‘‘B-cell lineage vaccine design’’ as originally proposed suggests that targeting the UCAs of lineages with broadly neutralizing activity, by administering a modified antigen that binds those UCAs with high affinity, should increase the likelihood of eliciting an otherwise subdominant response (Haynes et al., 2012). This description had in mind potential HIV vaccines to generate a naive response in uninfected recipients. The wide variety of evolved solutions to the problem of RBS recognition and sialic-acid mimicry, documented here, and the confounding effects of previous exposures suggest that for influenza vaccines, a more appropriate version of lineage-based design might be to focus response on the RBS (e.g., by using a chimeric (Hai et al., 2012) or hyperglycosylated immunogen or a succession of different immunogens), rather than seeking to elicit any particular type of germline antibody.

EXPERIMENTAL PROCEDURES Donors Donors were recruited at Duke University and given the trivalent inactivated seasonal influenza vaccine (TIV) and blood drawn on day 0, 7 and 21, as previously described (Moody et al., 2011). TIV01 and TIV04 were

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Figure 4. Contact Residues of CH65-like Antibodies and Resistance to RBS-Directed Antibodies (A–D) Antibody footprints of (A) CH65 (green), (B) H2526 (magenta), (C) CH67 (yellow) and (D) 641 I-9 (cyan); overlapping contacts with sialic acid and conserved residues are in dark blue. (E) Conserved and sialic acid contacts (dark blue) with resistance mutation G189D (red) highlighted. (F) Neutralization and binding data for H1 Solomon Islands/03/2006 wild-type or a virus with resistance mutation G189D. Neutralization IC50 values are expressed in mg/ml. For antibodies CH67, 641 I-9 and H2526, see Table 1; for Cr6261, see Ekiert et al. (2009). See also Table S5 for antibody-antigen contacts.

given 2007–2008 FluzoneH vaccine containing A/Solomon Islands/3/2006 (H1N1), A/Wisconsin/ 67/2005 (H3N2), and B/Malaysia/2506/2004; TIV 14, TIV21 and TIV24 were given 2008–2009 FluzoneH vaccine containing A/Brisbane/59/2007 (H1N1), A/Uruguay/716/2007 (H3N2) and B/Florida/ 04/2006. All participants were recruited, vaccinated, and sampled during the 2008 calendar year, before the emergence and circulation of the pandemic 2009 influenza strain (Dawood et al., 2009). See Table S1 for summary. Lineage Analysis After isolation and sequencing of influenza-specific antibodies (Liao et al., 2009) lineages following methods previously described (Moody et al., 2011). Briefly, we used the dnaml program from the Phylip 3.69 package (Felsenstein, 2005) to estimate the maximum-likelihood phylogenetic tree for the clone, aligned the inferred sequence at the root to germline gene libraries to obtain the UCA, and recomputed the tree using the UCA at the root and inferred the intermediate sequences. See Figure S1 for lineage trees.

Fab Expression and Purification The genes for the heavy- and light-chain variable and constant domains of 641 I-9 and the single chain Fv (scFv) of H2526 with a (GGGGS)3 linker all with a noncleavable C-terminal His6X tag, were synthesized and codon optimized for mammalian cell expression by GenScript. Constructs were produced by transient transfection in mammalian 293T cells as previously described (Schmidt et al., 2013). Proteins were purified using Co-NTA agarose (Clontech) followed by gel filtration chromatography on Superdex 200 (GE Healthcare). All point mutations were made using the QuikChange Mutagenesis (Agilent), following the manufacturer’s suggested protocol. Sequencing reactions were carried out with an ABI3730xl DNA analyzer at the DNA Resource Core of Dana-Farber/Harvard Cancer Center (funded in part by NCI Cancer Center support grant 2P30CA006516-48). Expression and Purification of HA HA A/Solomon Islands/03/2006 globular ‘‘head’’ was cloned into pFastBac vector for insect cell expression and purified, as previously described (Schmidt et al., 2013). Briefly, head was purified by Co-NTA agarose (Clontech) followed by gel filtration chromatography on Superdex 200 (GE Healthcare). The C-terminal His6X tag was removed by treatment with PreScission protease (MolBioTech) and the protein repurified by orthogonal Co-NTA agarose chromatography. Interferometry and Binding Experiments Interferometry experiments were performed using a BLItz instrument (forteBIO, Pall Corporation) as previously described (Xu et al., 2014). Purified Fab was immobilized on a Ni-NTA biosensor, and cleaved HA Solomon Islands/ 03/2006 globular head was added to obtain binding kinetics. The ka and kd parameters were determined and KD was obtained by applying a 1:1 binding isotherm to the plateau value at each concentration. All experiments were carried out using a minimum of four different concentrations of HA head and each run was incubated until binding reached saturation.

Figure 5. Serum Blocking from TIV Donors Serum pre-immunization (gray) and post-immunization (black) from TIV donors described in this study were assayed for competition with CH65 antibody from lineage 860; fold change from pre- to post-vaccination is noted in parentheses. Red asterisk denotes TIV donors for whom we identified CH65-like antibodies.

Crystallization The 641 I-9 Fab and H2526 were incubated at a 2:1 molar ratio with HA head, and the resulting 1:1 complex was purified from excess Fab by gel filtration chromatography on Superdex 200 in 10 mM Tris-HCl, 150 mM NaCl (pH7.5). The complexes were concentrated to 10–12 mg/ml. Crystals of 641 I-9 complex were grown in hanging drops over a reservoir of 100 mM HEPES (pH7.5), 6% polyethylene glycol 2 K, 300 mM magnesium nitrate, 50 mM lithium sulfate and 2% MPD, were harvested, cryoprotected with additional MPD-supplemented crystallization buffer added directly to the drop, and flash cooled in liquid nitrogen. The H2526 complex were grown in hanging drops over a reservoir of 100 mM HEPES (pH 7.0), 30% polyethylene glycol 400 and 100 mM ammonium sulfate. Crystals were harvested directly from drop and flash cooled in liquid nitrogen.

Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc. 7

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Structure Determination and Refinement We recorded diffraction data at beamlines 24-ID-E at the Advanced Photon Source and 8.2.2 at the Advanced Light Source for 641 I-9 complex and H2526 complex, respectively. Data sets from individual crystals were processed with HKL2000 (Otwinowski, 1997). Molecular replacement (MR) was carried out with PHASER (McCoy et al., 2007), refinement with PHENIX (Terwilliger et al., 2008), and all model modifications with COOT (Emsley and Cowtan, 2004). For the 641 I-9-HA complex, we separated the search model into three fragments: the 641 I-9 Fab VHVL and CHCL fragments and the HA head. There were two copies in the asymmetric unit (ASU). Simulated annealing refinement of atomic positions and B-factors was followed by TLS and positional refinement and placement of water molecules. Non-crystallographic symmetry (NCS) restraints were imposed throughout refinement. For the H2526 complex, search models of a VHVL from PDB 3GBN with the CDR H3 removed and the HA head from PDB 4HKX were used for MR. A single copy of the complex was present in the ASU. The CDR H3 was rebuilt de novo along with mutations in the VHVL using COOT and refined using PHENIX. A similar refinement procedure was followed as for the 641 I-9 complex. The structures were validated with the MolProbity server (Chen et al., 2010). Coordinates and diffraction data have been deposited at the PDB: 4YK4 and 4YJZ. The data collection and refinement statistics can be found in Table S6. Structure Analysis All images were created using PyMol (Schro¨dinger), and interface buried surface was calculated using PISA (Krissinel and Henrick, 2007). Neutralization Studies Madin-Darby canine kidney cells (MDCK 33016-PF) were maintained in chemically defined medium (CDM, Lonza 00194243) at 37 C in flasks shaken at 125 RPM in a humidified 5% CO2 environment. Serial 4-fold dilutions of mAb in Dulbecco’s modified Eagle medium (DMEM, Lonza 12-604F) supplemented with 1% BSA (Sigma A9576), 100 U/ml penicillin, and 100 mg/ml streptomycin (P/S, Lonza 17-602E) were mixed in equal volumes with 200 focus-forming units per well (FFU/well) of virus to a final volume of 100 ml and incubated at 37 C for 2 hr. 9 3 103 MDCK 33016-PF cells/well in 100 ml DMEM supplemented with 1% BSA and P/S were added to antibody/virus mixtures and incubated 18 hr at 37 C. Infected cells were fixed with 50/50 ice cold acetone/methanol at 20 C for 30 min, washed once with wash buffer (PBS with 0.1% Tween 20), and blocked in blocking buffer (PBS with 2% BSA [Rockland BSA-30]) for 30 min at RT. Cells were incubated for 1 hr at RT in the presence of a 1:6,000 dilution of anti-influenza A or B nucleoprotein (NP) mAb cocktail (Millipore MAB8251 or MAB8661) in blocking buffer, washed 33 with wash buffer, and stained 1 hr at RT in the presence of 4 mg/ml goat anti-mouse IgG-conjugated Alexa Fluor 488 (Invitrogen A11001) in blocking buffer. Stained cells were scanned and quantified with a BioSpot Analyzer (Cellular Technology). Infectivity was calculated as the percent of FFU that were present at given mAb concentrations relative to the average FFU of control wells incubated without mAb on each respective plate. A four-parameter fit was calculated with GraphPad Prism 6 (GraphPad Software) and used to determine the half maximal inhibitory concentration (IC50) in mg/ml. CH65 Blocking Experiments Blocking experiments were performed by competitive ELISA as described (Tomaras et al., 2008). Briefly, mAb CH65 was biotinylated using standard techniques; ELISA plates (Costar) were coated with H1 A/Solomon Islands/ 03/2006 protein. Triplicate samples of plasma (1:50 dilution) were incubated for 1 hr at room temperature (RT), washed, and then followed by biotinylated target mAb at 50% effective concentration (determined by direct binding in the absence of plasma). Binding of biotinylated mAb was detected by streptavidin-horseradish peroxidase and developed using substrate followed by optical density (OD) reading at 450 nm. Background wells (containing no biotinylated mAb) were averaged and subtracted from other readings on the same plate; the fraction of binding inhibition is calculated from the formula

8 Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc.

 % of mAb inhibition = 1

 mean OD of wells with blocking 3 100: mean OD of wells with no blocking

ACCESSION NUMBERS The accession numbers for the coordinates and structure factors reported in this paper are Protein Data Bank: 4YK4 and 4YJZ for 641 I-9 and H2526 complexes, respectively. SUPPLEMENTAL INFORMATION Supplemental Information includes Supplemental Experimental Procedures, three figures, and six tables and can be found with this article online at http://dx.doi.org/10.1016/j.cell.2015.04.028. AUTHOR CONTRIBUTIONS A.G.S., S.S., M.A.M, and S.C.H. designed research; A.G.S., M.D.T., S.S., T.B.K., H.-X.L, M.A.M., performed research; A.G.S., M.D.T., S.S., T.B.K., H.-X.L, M.A.M., B.F.H., and S.C.H. analyzed data; A.G.S. and S.C.H. wrote the paper. ACKNOWLEDGMENTS We thank members of the Harrison Laboratory for discussions. We thank the beamline staff at Advanced Light Source 8.2.2 and Advanced Photon Source NE-CAT ID-24E. The work at Harvard Medical School and Boston Children’s Hospital was supported by NIH Grant P01AI089618. S.C.H. is an investigator of the Howard Hughes Medical Institute. The authors note that S.C.H., A.G.S., B.F.H., H.X.L., M.A.M., and T.B.K. hold a patent on CH65 lineage antibodies. Received: January 27, 2015 Revised: March 6, 2015 Accepted: April 9, 2015 Published: May 7, 2015 REFERENCES Brezinschek, H.P., Brezinschek, R.I., and Lipsky, P.E. (1995). Analysis of the heavy chain repertoire of human peripheral B cells using single-cell polymerase chain reaction. J. Immunol. 155, 190–202. Burton, D.R., Poignard, P., Stanfield, R.L., and Wilson, I.A. (2012). Broadly neutralizing antibodies present new prospects to counter highly antigenically diverse viruses. Science 337, 183–186. Chen, V.B., Arendall, W.B., 3rd, Headd, J.J., Keedy, D.A., Immormino, R.M., Kapral, G.J., Murray, L.W., Richardson, J.S., and Richardson, D.C. (2010). MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21. Corti, D., and Lanzavecchia, A. (2013). Broadly neutralizing antiviral antibodies. Annu. Rev. Immunol. 31, 705–742. Corti, D., Voss, J., Gamblin, S.J., Codoni, G., Macagno, A., Jarrossay, D., Vachieri, S.G., Pinna, D., Minola, A., Vanzetta, F., et al. (2011). A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 333, 850–856. Dawood, F.S., Jain, S., Finelli, L., Shaw, M.W., Lindstrom, S., Garten, R.J., Gubareva, L.V., Xu, X., Bridges, C.B., and Uyeki, T.M.; Novel Swine-Origin Influenza A (H1N1) Virus Investigation Team (2009). Emergence of a novel swineorigin influenza A (H1N1) virus in humans. N. Engl. J. Med. 360, 2605–2615. DiLillo, D.J., Tan, G.S., Palese, P., and Ravetch, J.V. (2014). Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgR interactions for protection against influenza virus in vivo. Nat. Med. 20, 143–151. Ekiert, D.C., Bhabha, G., Elsliger, M.A., Friesen, R.H., Jongeneelen, M., Throsby, M., Goudsmit, J., and Wilson, I.A. (2009). Antibody recognition of a highly conserved influenza virus epitope. Science 324, 246–251.

Please cite this article in press as: Schmidt et al., Viral Receptor-Binding Site Antibodies with Diverse Germline Origins, Cell (2015), http:// dx.doi.org/10.1016/j.cell.2015.04.028

Ekiert, D.C., Kashyap, A.K., Steel, J., Rubrum, A., Bhabha, G., Khayat, R., Lee, J.H., Dillon, M.A., O’Neil, R.E., Faynboym, A.M., et al. (2012). Cross-neutralization of influenza A viruses mediated by a single antibody loop. Nature 489, 526–532.

Okada, J., Ohshima, N., Kubota-Koketsu, R., Iba, Y., Ota, S., Takase, W., Yoshikawa, T., Ishikawa, T., Asano, Y., Okuno, Y., and Kurosawa, Y. (2011). Localization of epitopes recognized by monoclonal antibodies that neutralized the H3N2 influenza viruses in man. J. Gen. Virol. 92, 326–335.

Emsley, P., and Cowtan, K. (2004). Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132.

Otwinowski, Z.a.M., W. (1997). Processing of X-ray Diffraction Data Collected in Oscillation Mode, Vol 276 (Methods In Enzymology: Academic Press).

Felsenstein, J. (2005). PHYLIP (Phylogeny Inference Package) (University of Washington: Seattle, WA: distributed by the author, Department of Genome Sciences).

Scheid, J.F., Mouquet, H., Ueberheide, B., Diskin, R., Klein, F., Oliveira, T.Y., Pietzsch, J., Fenyo, D., Abadir, A., Velinzon, K., et al. (2011). Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science 333, 1633–1637.

Hai, R., Krammer, F., Tan, G.S., Pica, N., Eggink, D., Maamary, J., Margine, I., Albrecht, R.A., and Palese, P. (2012). Influenza viruses expressing chimeric hemagglutinins: globular head and stalk domains derived from different subtypes. J. Virol. 86, 5774–5781. Harrison, S.C. (2008). Viral membrane fusion. Nat. Struct. Mol. Biol. 15, 690–698. Haynes, B.F., Kelsoe, G., Harrison, S.C., and Kepler, T.B. (2012). B-cell-lineage immunogen design in vaccine development with HIV-1 as a case study. Nat. Biotechnol. 30, 423–433. Hong, M., Lee, P.S., Hoffman, R.M., Zhu, X., Krause, J.C., Laursen, N.S., Yoon, S.I., Song, L., Tussey, L., Crowe, J.E., Jr., et al. (2013). Antibody recognition of the pandemic H1N1 Influenza virus hemagglutinin receptor binding site. J. Virol. 87, 12471–12480. Klein, F., Diskin, R., Scheid, J.F., Gaebler, C., Mouquet, H., Georgiev, I.S., Pancera, M., Zhou, T., Incesu, R.B., Fu, B.Z., et al. (2013). Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell 153, 126–138. Knossow, M., Gaudier, M., Douglas, A., Barre`re, B., Bizebard, T., Barbey, C., Gigant, B., and Skehel, J.J. (2002). Mechanism of neutralization of influenza virus infectivity by antibodies. Virology 302, 294–298. Krause, J.C., Tsibane, T., Tumpey, T.M., Huffman, C.J., Basler, C.F., and Crowe, J.E., Jr. (2011). A broadly neutralizing human monoclonal antibody that recognizes a conserved, novel epitope on the globular head of the influenza H1N1 virus hemagglutinin. J. Virol. 85, 10905–10908. Krissinel, E., and Henrick, K. (2007). Inference of macromolecular assemblies from crystalline state. J. Mol. Biol. 372, 774–797. Liao, H.X., Levesque, M.C., Nagel, A., Dixon, A., Zhang, R., Walter, E., Parks, R., Whitesides, J., Marshall, D.J., Hwang, K.K., et al. (2009). High-throughput isolation of immunoglobulin genes from single human B cells and expression as monoclonal antibodies. J. Virol. Methods 158, 171–179. Mascola, J.R., and Haynes, B.F. (2013). HIV-1 neutralizing antibodies: understanding nature’s pathways. Immunol. Rev. 254, 225–244. McCoy, A.J., Grosse-Kunstleve, R.W., Adams, P.D., Winn, M.D., Storoni, L.C., and Read, R.J. (2007). Phaser crystallographic software. J. Appl. Cryst. 40, 658–674. Moody, M.A., Zhang, R., Walter, E.B., Woods, C.W., Ginsburg, G.S., McClain, M.T., Denny, T.N., Chen, X., Munshaw, S., Marshall, D.J., et al. (2011). H3N2 influenza infection elicits more cross-reactive and less clonally expanded anti-hemagglutinin antibodies than influenza vaccination. PLoS ONE 6, e25797. Okada, J., Ohshima, N., Kubota-Koketsu, R., Ota, S., Takase, W., Azuma, M., Iba, Y., Nakagawa, N., Yoshikawa, T., Nakajima, Y., et al. (2010). Monoclonal antibodies in man that neutralized H3N2 influenza viruses were classified into three groups with distinct strain specificity: 1968-1973, 1977-1993 and 19972003. Virology 397, 322–330.

Schmidt, A.G., Xu, H., Khan, A.R., O’Donnell, T., Khurana, S., King, L.R., Manischewitz, J., Golding, H., Suphaphiphat, P., Carfi, A., et al. (2013). Preconfiguration of the antigen-binding site during affinity maturation of a broadly neutralizing influenza virus antibody. Proc. Natl. Acad. Sci. USA 110, 264–269. Skehel, J.J., and Wiley, D.C. (2000). Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu. Rev. Biochem. 69, 531–569. Sui, J., Hwang, W.C., Perez, S., Wei, G., Aird, D., Chen, L.M., Santelli, E., Stec, B., Cadwell, G., Ali, M., et al. (2009). Structural and functional bases for broadspectrum neutralization of avian and human influenza A viruses. Nat. Struct. Mol. Biol. 16, 265–273. Terwilliger, T.C., Grosse-Kunstleve, R.W., Afonine, P.V., Moriarty, N.W., Zwart, P.H., Hung, L.W., Read, R.J., and Adams, P.D. (2008). Iterative model building, structure refinement and density modification with the PHENIX AutoBuild wizard. Acta Crystallogr. D Biol. Crystallogr. 64, 61–69. Tomaras, G.D., Yates, N.L., Liu, P., Qin, L., Fouda, G.G., Chavez, L.L., Decamp, A.C., Parks, R.J., Ashley, V.C., Lucas, J.T., et al. (2008). Initial Bcell responses to transmitted human immunodeficiency virus type 1: virionbinding immunoglobulin M (IgM) and IgG antibodies followed by plasma anti-gp41 antibodies with ineffective control of initial viremia. J. Virol. 82, 12449–12463. Weis, W., Brown, J.H., Cusack, S., Paulson, J.C., Skehel, J.J., and Wiley, D.C. (1988). Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature 333, 426–431. West, A.P., Jr., Scharf, L., Scheid, J.F., Klein, F., Bjorkman, P.J., and Nussenzweig, M.C. (2014). Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648. Whittle, J.R., Zhang, R., Khurana, S., King, L.R., Manischewitz, J., Golding, H., Dormitzer, P.R., Haynes, B.F., Walter, E.B., Moody, M.A., et al. (2011). Broadly neutralizing human antibody that recognizes the receptor-binding pocket of influenza virus hemagglutinin. Proc. Natl. Acad. Sci. USA 108, 14216–14221. Wiley, D.C., Wilson, I.A., and Skehel, J.J. (1981). Structural identification of the antibody-binding sites of Hong Kong influenza haemagglutinin and their involvement in antigenic variation. Nature 289, 373–378. Wu, X., Yang, Z.Y., Li, Y., Hogerkorp, C.M., Schief, W.R., Seaman, M.S., Zhou, T., Schmidt, S.D., Wu, L., Xu, L., et al. (2010). Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329, 856–861. Xu, R., Krause, J.C., McBride, R., Paulson, J.C., Crowe, J.E., Jr., and Wilson, I.A. (2013). A recurring motif for antibody recognition of the receptor-binding site of influenza hemagglutinin. Nat. Struct. Mol. Biol. 20, 363–370. Xu, H., Schmidt, A.G., O’Donnell, T., Therkelsen, M.D., Kepler, T.B., Moody, M.A., Haynes, B.F., Liao, H., Harrison, S.C., and Shaw, D.E. (2014). Key mutations stabilize antigen-binding conformation during affinity maturation of a broadly neutralizing influenza antibody lineage. Proteins 83, 771–780.

Cell 161, 1–9, May 21, 2015 ª2015 Elsevier Inc. 9

Viral receptor-binding site antibodies with diverse germline origins.

Vaccines for rapidly evolving pathogens will confer lasting immunity if they elicit antibodies recognizing conserved epitopes, such as a receptor-bind...
2MB Sizes 0 Downloads 6 Views