RSV IMMUNOGENS, ANTIBODIES AND COMPOSITIONS THEREOF

Abstract
The present invention provides immunogens that protect against RSV infection. The present invention also provides antibody proteins that protect against RSV infection. Such immunogens and antibody proteins are produced based on three-dimensional models also included in the invention. One model is of a complex between motavizumab and its antibody-binding domain on RSV fusion (F) protein. A second model is of a complex between 10 IF antibody and its antibody-binding domain on RSV F protein. The immunogens disclosed herein have been modified to elicit a humoral response against RSV F protein without eliciting a significant cell-mediated response against RSV. Such immunogens can comprise a scaffold into which RSV contact residues are embedded. The present invention also includes methods that utilize the disclosed three-dimensional models to produce immunogens and antibody proteins of the present invention. Also disclosed are methods of using the disclosed immunogens, for example to protect individuals from RSV infection. Also disclosed are methods of using the disclosed antibody proteins, for example to protect individuals from RSV infection.
Description
FIELD

The present invention relates to novel compositions that protect individuals from respiratory syncytial virus (RSV) infection. In particular, the present invention relates to vaccines that elicit antibodies having a high affinity for the RSV fusion (F) protein. The present invention also relates to therapeutic compositions comprising antibodies having a high affinity for the RSV F protein.


BACKGROUND

Respiratory syncytial virus (RSV) is a highly contagious member of the Paramyxoviridae family of viruses that causes significant worldwide morbidity and mortality each year, particularly in infants. RSV infects people repeatedly throughout life, and causes significant morbidity in healthy children and adults. The RSV fusion (F) protein (see, e.g., Lopez J A et al., 1998, J. Virol. 72, 6922-6928) and antibodies thereto, have been targets for vaccine efforts. There is currently no licensed RSV vaccine. A previous vaccine trial in the 1960s containing a formalin-inactivated RSV actually enhanced the severity of disease upon natural infection with RSV. This was thought to have occurred due to an imbalanced T-cell response and elicitation of low avidity antibodies. Since there is currently no licensed RSV vaccine, passive immunization is used to prevent RSV infection, especially in those infants with prematurity, bronchopulmonary dysplasia, or congenital heart disease. Originally, RSV-neutralizing polyclonal antibodies from pooled human sera (RESPIGAM®) were used (see, e.g., Groothuis J R et al., 1995, Pediatrics 95, 463-467). This treatment was followed by the development of palivizumab (SYNAGIS®) (see, e.g., Johnson S et al., 1997, J. Infect. Dis. 176, 1215-1224). Palivizumab was humanized from mouse antibody 1129, which binds a 24-amino acid, linear, conformational epitope on the RSV F protein (see, e.g., Beeler J A, et al., 1989, J. Virol. 63, 2941-2950; Arbiza J et al., J. Gen. Virol. 73, 2225-2234, Lopez J A et al., 1993, J. Gen. Virol. 74, 2567-2577). Palivizumab binds to the F protein and thereby neutralizes the virus. Such treatments are expensive, costing approximately $1000 per dose. Moreover, the antibodies must be administered on a monthly basis during the winter months, thereby adding to the cost of treatment. When administered at a dose of 15 mg/kg each month during the RSV season, palivizumab reduces RSV-related hospitalizations by 55% (see, e.g., The Impact-RSV Study Group, 1998, Pediatrics 102, 531-537). Thus, a vaccine requiring only one or two administrations would have advantages over the current preventative treatment for RSV.


SUMMARY

The present invention provides immunogens that protect against RSV infection. The present invention also provides antibody proteins that protect against RSV infection. Such immunogens and antibody proteins are produced based on three-dimensional models also included in the invention. One model is of a complex between motavizumab and its antibody-binding domain on RSV fusion (F) protein. A second model is of a complex between 101F antibody and its antibody-binding domain on RSV F protein. The immunogens disclosed herein have been modified to elicit a humoral response against RSV F protein without eliciting a significant cell-mediated response against RSV. Such immunogens can comprise a scaffold into which RSV contact residues are embedded. The present invention also includes methods that utilize the disclosed three-dimensional models to produce immunogens and antibody proteins of the present invention. Also disclosed are methods of using the disclosed immunogens, for example to protect individuals from RSV infection. Also disclosed are methods of using the disclosed antibody proteins, for example to protect individuals from RSV infection.


The present disclosure provides an RSV immunogen comprising an amino acid sequence of 1LP1_b (SEQ ID NO: 11) having from one to twenty amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of: (a) substitution of a serine at amino acid position N-25 in SEQ ID NO:11; (b) substitution of a leucine at amino acid position 1-28 in SEQ ID NO:11; (c) substitution of a serine at amino acid position Q-29 in SEQ ID NO: 11; (d) substitution of an isoleucine at amino acid position L-31 in SEQ ID NO:11; (e) substitution of an asparagine at amino acid position K-32 in SEQ ID NO:11; (f) substitution of an aspartic acid at amino acid position K-4 in SEQ ID NO:11; (g) substitution of a lysine at amino acid position Q-6 in SEQ ID NO:11; (h) substitution of a lysine at amino acid position Q-7 in SEQ ID NO:11; (i) substitution of a leucine at amino acid position N-8 in SEQ ID NO:11; (j) substitution of a serine at amino acid position F-10 in SEQ ID NO:11; and (k) substitution of an asparagine at amino acid position Y-11 in SEQ ID NO:11. Such an RSV immunogen can comprise all of the substitutions (a) through (k). The present disclosure also provides an RSV immunogen comprising an amino acid sequence of truncated 1S2X_a (SEQ ID NO:13) having from one to twenty-five amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of: (a) substitution of a serine at amino acid position 92 in SEQ ID NO:13; (b) substitution of a leucine at amino acid position 95 in SEQ ID NO:13; (c) substitution of a serine at amino acid position 96 in SEQ ID NO:13; (d) substitution of an isoleucine at amino acid position 98 in SEQ ID NO:13; (e) substitution of an asparagine at amino acid position 99 in SEQ ID NO:13; (f) substitution of an aspartic acid at amino acid position 100 in SEQ ID NO:13; (g) substitution of an asparagine at amino acid position 105 in SEQ ID NO:13; (h) substitution of an aspartic acid at amino acid position 106 in SEQ ID NO:13; (i) substitution of a lysine at amino acid position 108 in SEQ ID NO:13; (j) substitution of a lysine at amino acid position 109 in SEQ ID NO:13; (k) substitution of a leucine at amino acid position 110 in SEQ ID NO:13; (l) substitution of a serine at amino acid position 112 in SEQ ID NO:13; and (m) substitution of an asparagine at amino acid position 113 in SEQ ID NO:13. Such an RSV immunogen can comprise all of the substitutions (a) through (m). The present disclosure also provides a nucleic acid molecule comprising a nucleic acid sequence that encodes any of such RSV immunogens. Also provided is a recombinant molecule comprising such a nucleic acid molecule. Also provided is a recombinant cell comprising such a recombinant molecule. Also provided are uses of such nucleic acid molecules, recombinant molecules, and recombinant cells to produce any of such RSV immunogens. A recombinant molecule can also be a nucleic acid vaccine. Also provided is a composition that can include any of the RSV immunogens, nucleic acid molecules, recombinant molecules, or recombinant cells. Also provided is a vaccine comprising any of such RSV immunogens. The disclosure provides a method to elicit a neutralizing humoral immune response against RSV; such method comprises administering any of such RSV immunogens or vaccines, wherein such administration elicits a neutralizing humoral immune response against RSV. Also provided is a method to protect a patient from RSV infection; such method comprises administering to the patient any of such RSV immunogens or vaccines, wherein such administration protects the patient from RSV infection.


The disclosure provides an immunogen comprising an antibody-binding domain that binds an antibody selected from the group consisting of motavizumab and 101F antibody, wherein the three-dimensional structure of the antibody-binding domain of the immunogen spatially corresponds to a three-dimensional structure of an antibody-binding domain of a fusion (F) peptide derived from respiratory syncytial virus (RSV) fusion (F) protein in a complex selected from the group consisting of:


(a) a complex between a F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, the complex being set forth in the three-dimensional model defined by the atomic coordinates specified in Research Collaboratory for Structural Bioinformatics (RCSB) Protein Data Bank accession code 3IXT; and


(b) a complex between a F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, the complex being set forth in the three-dimensional model defined by the atomic coordinates specified in Research Collaboratory for Structural Bioinformatics (RCSB) Protein Data Bank accession code 3O41;


wherein the antibody-binding domain of the immunogen comprises less than 12 consecutive amino acids from a motavizumab-binding domain or a 101F antibody-binding domain of RSV F protein, and wherein the immunogen elicits a humoral immune response against RSV. In one embodiment, the antibody-binding domain of the immunogen comprises contact residues that have a spatial orientation represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms from the corresponding backbone atoms of contact residues in an RSV F peptide that contacts motavizumab or 101F antibody in the respective complex. In one embodiment, the immunogen comprises contact residues of the motavizumab-binding domain embedded in a protein scaffold comprising a three-dimensional structure having two alpha helices defined by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the two alpha helices of the peptide consisting of amino acid sequence SEQ ID NO:2 when complexed with motavizumab, the three-dimensional model of the complex being defined by the coordinates specified in Protein Data Bank accession code 3IXT. In one embodiment, the immunogen comprises contact residues of the 101F antibody-binding domain embedded in a protein scaffold comprising a three-dimensional structure with atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the peptide consisting of amino acid sequence SEQ ID NO:4 when complexed with 101F antibody, the 3-dimensional model of the complex being defined by the coordinates specified in Protein Data Bank accession code 3O41. The present disclosure also provides a nucleic acid molecule comprising a nucleic acid sequence that encodes any of such immunogens. Also provided is a recombinant molecule comprising such a nucleic acid molecule. Also provided is a recombinant cell comprising such a recombinant molecule. Also provided are uses of such nucleic acid molecules, recombinant molecules, and recombinant cells to produce any of such immunogens. A recombinant molecule can also be a nucleic acid vaccine. Also provided is a composition that can include any of the immunogens, nucleic acid molecules, recombinant molecules, or recombinant cells. Also provided is a vaccine comprising any of such immunogens. The disclosure provides a method to elicit a neutralizing humoral immune response against RSV; such method comprises administering any of such immunogens or vaccines, wherein such administration elicits a neutralizing humoral immune response against RSV. Also provided is a method to protect a patient from RSV infection; such method comprises administering to the patient any of such immunogens or vaccines, wherein such administration protects the patient from RSV infection.


The disclosure provides an antibody protein comprising a heavy chain comprising SEQ ID NO:5, except that the antibody protein comprises at least one amino acid substitution selected from the group consisting of: (a) substitution of the amino acid corresponding to position 32 of SEQ ID NO:5, is a histidine or a glutamic acid; (b) substitution of the amino acid at position 35 of SEQ ID NO:5 is substituted with an alanine; (c) substitution of the amino acid at position 52 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of lysine, histidine, threonine, serine and arginine; (d) substitution of the amino acid at position 53 of SEQ ID NO:5 is substituted with a histidine or a serine; (e) substitution of the amino acid at position 54 of SEQ ID NO:5 is substituted with a phenylalanine or an arginine; (f) substitution of the amino acid at position 56 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of isoleucine, serine, glutamic acid, and aspartic acid; (g) substitution of the amino acid at position 58 of SEQ ID NO:5 is substituted with a tyrosine or a tryptophan; (h) substitution of the amino acid at position 97 of SEQ ID NO:5 is substituted with an aspartic acid, a histidine or an arginine; (i) substitution of the amino acid at position 99 of SEQ ID NO:5 is substituted with aspartic acid; (j) substitution of the amino acid at position 100 of SEQ ID NO:5 is substituted with a tyrosine, a tryptophan or a histidine; and (k) substitution of the amino acid at position 100A of SEQ ID NO:5 is substituted with a serine or a threonine. The disclosure provides an antibody protein comprising a heavy chain comprising SEQ ID NO:6, except that the antibody protein comprises at least one amino acid substitution selected from the group consisting of: (a) substitution of the amino acid at position 32 of SEQ ID NO:6 is substituted with a phenylalanine; (b) substitution of the amino acid at position 49 of SEQ ID NO:6 is substituted with a histidine or an arginine; (c) substitution of the amino acid at position 92 of SEQ ID NO:6 is substituted with a lysine; (d) substitution of the amino acid at position 94 of SEQ ID NO:6 is substituted with a histidine; and (e) substitution of the amino acid at position 96 of SEQ ID NO:6 is substituted with a histidine. The present disclosure also provides a nucleic acid molecule comprising a nucleic acid sequence that encodes any of such antibody proteins. Also provided is a recombinant molecule comprising such a nucleic acid molecule. Also provided is a recombinant cell comprising such a recombinant molecule. Also provided are uses of such nucleic acid molecules, recombinant molecules, and recombinant cells to produce any of such antibody proteins. Also provided is a composition that can include any of the antibody proteins. Also provided is a method to protect a patient from RSV infection; such method comprises administering to the patient any of such immunogens or vaccines, wherein such administration protects the patient from RSV infection.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1. Structural basis of motavizumab binding to its F glycoprotein epitope. (a) RSV neutralization curves for palivizumab and motavizumab IgG as determined by a flow cytometric assay. (b) Surface representation of the Fab and ribbon representation of the peptide, viewed looking down at the CDRs. (c) Interactions between the peptide and six Fab CDRs. Side chains are shown for those residues making intermolecular interactions. Dashed lines represent hydrogen bonds. Residues in the hydrophobic patch on the heavy chain are shown with transparent surfaces. These include Trp52 and Trp53 on the right, and Ile97, Phe98 and Phe100 on the left (d) Motavizumab mutations that alter affinity to the F glycoprotein. Side chains are shown for those Fab residues that increase affinity by directly contacting the peptide (red), by altering the position of residues that directly contact the peptide (cyan), and by binding to the F glycoprotein outside the primary epitope or enhancing long-range electrostatic interactions (magenta). Fab residues in yellow decrease the affinity for the F glycoprotein but increase in vivo potency compared to an earlier version of motavizumab.



FIG. 2. Motavizumab binding to the RSV F glycoprotein. (a) Superposition of the motavizumab-bound peptide (grey) and residues 229-252 of the PIV5 F glycoprotein structure (red). (b) Ribbon representation of the model of motavizumab Fab (green and blue) bound to the PIV5 F glycoprotein monomer (tan) via the superposition shown in a. (c) Same as b, except the entire PIV5 F glycoprotein trimer is shown (tan, green, pink). (d) Magnification of the boxed area shown in c. (e) Gel filtration elution profile and corresponding Coomassie blue-stained SDS-PAGE gel of the RSV F0 Fd glycoprotein. (f) Gel filtration elution profile and corresponding Coomassie blue-stained SDS-PAGE gel of a mixture of RSV Fo Fd glycoprotein and excess palivizumab Fab. Densitometric analysis of the gel yields a ratio of 2.97 Fabs per trimer.



FIG. 3. Peptide electron density. Stereo image of 2Fo—Fc density contoured at 1σ around the 24-amino acid peptide, viewed from the bound antibody. The electrostatic potential of the peptide in this orientation is shown in FIG. 4 (Right).



FIG. 4. Shape and electrostatic potential complementarity. (Left) Cartoon and stick representation of the peptide bound to a surface of the motavizumab Fab colored according to electrostatic potentials (negative potentials are colored red, positive potentials are colored blue). (Right) Cartoon and stick representation of the six motavizumab CDRs bound to a surface of the peptide colored according to electrostatic potentials. Heavy chain CDRs are green, and light chain CDRs are blue. The scale is from −5 to 5 kT/e. The images are related by a 180° rotation.



FIG. 5. RSV/PIV5 F glycoprotein alignments. FIG. 5-1 (also labeled “a”) Sequence alignment. Sequence of the crystallized PIV5 F glycoprotein fragment aligned with the corresponding residues from the RSV F glycoprotein A2 strain. The sequences were first aligned using CLUSTALW2 (Larkin M A et al., 2007, Bioinformatics 23, 2947-2948) and then manually adjusted to align the furin cleavage site and disulfide bonds. Secondary structure of the PIV5 F glycoprotein as determined from the crystal structure and the PSIPRED-predicted (Bryson K et al., 2005, Nucl. Acids Res. 33, W36-38) RSV F and PIV5 F glycoprotein secondary structure is shown. The motavizumab epitope is highlighted in grey. Arrows indicate β-strands and coils indicate α-helices. Numbering corresponds to the RSV F glycoprotein. FIG. 5-2 (also labeled “b”) Structure alignment. The structure of the motavizumab-bound RSV F peptide was used to refine the alignment with the PIV5 F glycoprotein. Superposition of RSV F residues 254-277 with PIV5 F residues 229-252 provided an rmsd of 2.1 Å for all 24 Cα atoms. This optimal structure alignment requires a shift of 3 amino acids relative to the sequence alignment in a, which also results in a better sequence alignment for this region.



FIG. 6. RSV F0 Fd cross-linking Coomassie-stained SDS-PAGE gel of glutaraldehyde cross-linked RSV F0 Fd proteins immunoprecipitated with motavizumab IgG.



FIG. 7. Comparison of the motavizumab epitope to Protein A and the 1lp1b scaffold. The 24-amino acid sequence of the motavizumab epitope on RSV F is shown on top, with the two helical regions identified in the crystal structure indicated by blue and orange cylinders. The sequence of a single domain of Protein A from S. aureus is shown (labeled Original), as well as the sequence of the 1lp1b003 scaffold (labeled Final). Bold residues were mutated to preserve motavizumab contact residues, while underlined residues were mutated to stabilize the conformation of the two helices. Structures of the motavizumab/peptide complex and the scaffold are shown below the sequences.



FIG. 8. Recombinant immunogen structure. Expression of immunogen 1lp1b003 in HEK293 and bacterial cells yields proteins that are alpha-helical in solution and have a melting temperature that confirms the proteins are folded in solution.





DETAILED DESCRIPTION

Previous attempts at making an RSV vaccine using the RSV F protein have been unsuccessful. This was due to poor immunogenicity or a concern about eliciting T cells that could worsen disease. The inventors have now solved the crystal structures of two, antibody-binding domains of the F protein when they are bound to their respective monoclonal antibodies: motavizumab, which binds to the F protein at a domain spanning amino acids 254 to 277 (see, e.g., Wu H et al., 2007, J. Mol. Biol. 368, 652-665); and chimeric 101F antibody (also referred to as 101F or CH101 (Centocor)) that binds to the F protein at a domain spanning from amino acids 422-436 (see, e.g., Wu, S-J et al., 2007, J. Gen Virol 88, 2719-2723). Analysis of this structure has led to the identification of the contact residues in the F protein when bound to motavizumab or 101F antibody. This information allows the identification of non-RSV proteins that have a similar three-dimensional structure to the respective antibody-binding domains (referred to as scaffold proteins), which can then be modified to contain the appropriate residues that enable the modified protein to bind motavizumab or 101F. Since such a modified protein is unrelated to the RSV F protein, except for the contact residues, it can be used as an immunogen to elicit antibodies against the RSV F protein. Preferably the immunogens do not elicit a significant cellular response against the F protein. The information gained from the three-dimensional model can also be used to design antibodies that have a high affinity for the RSV F protein, and that can be used to protect individuals from RSV infection.


Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.


It should be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative” limitation.


It should be understood that as used herein, the term “a” entity or “an” entity refers to one or more of that entity. For example, a peptide or protein refers to one or more peptides or proteins. As such, the terms “a”, “an”, “one or more” and “at least one” can be used interchangeably. Similarly the terms “comprising”, “including” and “having” can be used interchangeably. Moreover, as used herein, the terms about and substantially refer to a variation of less than 5% from the object of the term, and preferably less than 2%.


The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates, which may need to be independently confirmed.


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.


Except as otherwise noted, the methods and techniques of the present embodiments are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification.


It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination. All combinations of the embodiments are specifically embraced by the present invention and are disclosed herein just as if each and every combination was individually and explicitly disclosed. In addition, all sub-combinations are also specifically embraced by the present invention and are disclosed herein just as if each and every such sub-combination was individually and explicitly disclosed herein.


One embodiment of the present invention is an immunogen comprising an antibody-binding domain, wherein the three-dimensional structure of the antibody-binding domain of the immunogen spatially corresponds to the three-dimensional structure of an antibody-binding domain of a peptide derived from RSV F protein, when such peptide is in a complex with an RSV neutralizing antibody that specifically binds the F protein. In one embodiment, the neutralizing antibody is palivizumab (SYNAGIS®). In another embodiment, the neutralizing antibody is motavizumab. In yet another embodiment, the neutralizing antibody is 101F. In one embodiment, the neutralizing antibody can be any antibody that binds an antibody-binding domain recognized by motavizumab (i.e., an antibody-binding domain to which motavizumab binds). In one embodiment, the neutralizing antibody can be any antibody that binds an antibody-binding domain recognized by 101F antibody (also referred to herein as 101F).


As used herein, a peptide derived from the RSV F protein is any peptide comprising at least a portion of SEQ ID NO:1, wherein said portion comprises an antibody-binding domain that binds palivizumab, motavizumab or 101F. Such a peptide can also be referred to as an RSV F peptide. As used herein, an antibody-binding domain is a group, or cluster, of amino acids within a protein or peptide, wherein at least one of the amino acid residues in the sequence interacts directly, or indirectly (e.g., forms a bond, such as an ionic bond or salt-bridge) with at least one amino acid residue in an antibody such as palivizumab, motavizumab or 101F, such that the antibody specifically binds the peptide. As used herein, the terms selectively, selective, specific, and the like, indicate the antibody has a greater affinity for the RSV protein or peptide, or the immunogen, than it does for proteins unrelated to the RSV F protein or peptide. More specifically, the terms selectively, selective, specific, and the like indicate that the affinity of the antibody the RSV protein or peptide, or the immunogen, is statistically significantly higher than its affinity for a negative control (e.g., an unrelated protein) as measured using a standard assay (e.g., ELISA). Suitable techniques for assaying the ability of an antibody to selectively interact with the RSV protein or peptide, or the immunogen, are known to those skilled in the art. Amino acid residues that act directly or indirectly to form bonds at the interface of two molecules, such as a peptide and antibody, are referred to as contact residues. Contact residues within a molecule can be contiguous, non-contiguous, or partly contiguous in the two-dimensional (linear) structure (i.e., linearly contiguous, linearly non-contiguous, or the like), but are sufficiently contiguous, or close together, in the three-dimensional structure to form an epitope (i.e., structurally contiguous).


In one embodiment, the peptide comprises a palivizumab-binding domain. In another embodiment, the peptide comprises a motavizumab antibody-binding domain (also referred to herein as a motavizumab antibody-binding site, motavizumab-binding domain, or motavizumab-binding site). In a preferred embodiment, the motavizumab antibody-binding site corresponds to amino acids 254-277 of SEQ ID NO:1. The amino acid sequence spanning residues 254-277 is NSELLSLIND MPITNDQKKL MSNN, also denoted herein as SEQ ID NO:2.


In yet another embodiment the peptide comprises a 101F antibody-binding site (also referred to herein as a 101F antibody-binding domain). In another preferred embodiment, the 101F antibody-binding site corresponds to amino acids 422-436 of SEQ ID NO:1. The amino acid sequence spanning residues 422-436 is STASNKNRGI IKTFS, also denoted herein as SEQ ID NO:3, except that the S at amino acid position 422 is replaced by a C in RSV F protein. In yet another preferred embodiment, the 101F antibody-binding site corresponds to amino acids 427-436 of SEQ ID NO:1. The amino acid sequence spanning residues 427-436 is KNRGIIKTFS, also denoted herein as SEQ ID NO:4. In yet another preferred embodiment, the 101F antibody-binding site corresponds to amino acids 422-438 of SEQ ID NO:1. The amino acid sequence spanning residues 422-438 is STASNKNRGI IKTFSNG, also denoted herein as SEQ ID NO:9, except that the S at amino acid position 422 is replaced by a C in RSV F protein.


A preferred embodiment of the present invention is an immunogen comprising an antibody-binding domain, wherein the three-dimensional structure of such antibody-binding domain spatially corresponds to the three-dimensional structure of a peptide consisting of amino acids 254-277 from the RSV F protein, when such peptide is complexed with motavizumab. Another preferred embodiment of the present invention is an immunogen comprising an antibody-binding domain, wherein the three-dimensional structure of such antibody-binding domain spatially corresponds to the three-dimensional structure of a peptide consisting of amino acids 427-436 from the RSV F protein, when such peptide is complexed with 101F. Another preferred embodiment of the present invention is an immunogen comprising an antibody-binding domain, wherein the three-dimensional structure of such antibody-binding domain spatially corresponds to the three-dimensional structure of a peptide consisting of amino acids 427-438 from the RSV F protein, when such peptide is complexed with 101F.


As used herein, the terms spatially corresponds, spatially corresponding, and the like, are used to indicate that when a three-dimensional model of a protein is superimposed on a three dimensional model of a RSV F peptide comprising a motavizumab or 101F binding domain when such peptide is in a complex with motavizumab or 101F, respectively, coordinates defining the spatial position of backbone atoms in the protein vary from coordinates defining the spatial location of analogous backbone atoms in the antibody-binding domain of the RSV F peptide, when such peptide is in a complex with motavizumab, by less than about 10 angstroms. Backbone atoms are those atoms in an amino acid that form the peptide backbone, or 3-dimensional folding pattern, of the 3-dimensional model. As such, backbone atoms are those atoms that make up the base, but not the side chain, of amino acid residues in s protein (i.e., nitrogen, carbon, alpha carbon, and oxygen). Analogous backbone atoms are atoms, that are in the same position within an amino acid. The term spatial position refers to an object's location in three-dimensional space, as defined by X, Y and Z coordinates. One system for determining the three-dimensional structure of a protein is X-ray crystallography. It is understood by those skilled in the relevant art that three-dimensional structures of proteins are defined using atomic coordinates. Thus, in one embodiment of the present invention the three-dimensional structure of the complex between the peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab is defined by the atomic coordinates recited, or specified, in Protein Data Bank (of the Research Collaboratory for Structural Bioinformatics (RCSB) accession code 3IXT (i.e., the atomic coordinates deposited at the Protein Data Bank under accession code 3IXT; also referred to as PDB acc code 3IXT). These coordinates were recited in Table 1 of U.S. Provisional Patent Application No. 61/253,826, filed Oct. 21, 2009 (U.S. 61/253,826). In another embodiment of the present invention, the three-dimensional structure of a complex between a peptide consisting of amino acid sequence SEQ ID NO:3 and 101F is defined by the atomic coordinates recited, or specified, in Protein Data Bank (of the Research Collaboratory for Structural Bioinformatics (RCSB) accession code 3O41 (also referred to as PDB acc code 3O41). These coordinates are more highly refined atomic coordinates corresponding to the atomic coordinates recited in Table 2; such refinement led to a three-dimensional structure that when superimposed on the three-dimensional structure of the complex defined by the atomic coordinates recited in Table 2 could not be distinguished visually from the latter structure; any differences were less than 0.1 angstroms. In another embodiment of the present invention, the three-dimensional structure of a complex between a peptide consisting of amino acid sequence SEQ ID NO:9 and 101F is defined by the atomic coordinates specified in Protein Data Bank (of the Research Collaboratory for Structural Bioinformatics (RCSB), accession code 3O45 (also referred to as PDB acc code 3O45).


While an immunogen of the present invention comprises an antibody binding domain spatially corresponding to an antibody-binding domain from an RSV F peptide present in a complex defined by the coordinates in PDB acc code 3IXT or in PDB acc code 3O41, it should be understood that some small variance in the spatial orientation of the immunogen antibody binding domain is permissible, as long as the immunogen binds palivizumab, motavizumab, or 101F. Thus one embodiment of the present invention is an immunogen comprising an antibody binding domain that has a three-dimensional structure defined by atomic coordinates having less than 10%, less than 5%, less than 3%, less than 2% or less than 1% variation from the atomic coordinates recited in PDB acc code 3IXT or in PDB acc code 3O41. Another embodiment of the presenting invention is an immunogen comprising an antibody-binding domain having a three-dimensional structure represented by atomic coordinates defining backbone atoms that have a root mean square deviation of less than 10 angstroms, less than 5 angstroms, less than 3 angstroms, less than 2 angstroms, or less than 1 angstrom from the backbone atoms of the antibody-binding domain of an RSV F peptide in a complex defined by the coordinates recited in PDB acc code 3IXT or in PDB acc code 3O41. Yet another embodiment of the present invention is an immunogen comprising an antibody-binding domain having a three-dimensional structure represented by atomic coordinates defining backbone atoms, wherein each atom has a root mean square deviation of less than 0.4 angstroms, less than 0.3 angstroms, less than 0.2 angstroms, or less than 0.1 angstrom from the corresponding backbone atom in the antibody-binding domain of an RSV F peptide in a complex defined by the coordinates recited in PDB acc code 3IXT or in PDB acc code 3O41.


As disclosed herein, an immunogen of the present invention comprises an antibody-binding domain that spatially corresponds to the antibody-binding domain in the RSV F peptide when bound to motavizumab or 101F. It is preferable that an immunogen of the present invention contain little or no homology to RSV F protein sequences from outside of an antibody-binding domain. It is also preferable that the immunogen not include any contiguous sequence from RSV F protein that is of sufficient length to generate a cellular immune response. As used herein, a cellular immune response, or cell-mediated immunity, refers to a T lymphocyte immune response and the release of related cytokines and other immunomodulatory molecules in response to an antigen that contains an antigenic peptide fragment consisting of a specific sequence of about 10 amino acids. In contrast, a humoral immune response, or humoral immunity, refers to the production by B-lymphocytes of antibodies (e.g., IgG, IgM or IgA antibodies) in response to an antigen. Such antibodies preferably neutralize RSV. One embodiment of the present invention is an immunogen that elicits a humoral immune response, but not a significant cellular immune response against RSV. One embodiment of the present invention is an immunogen that elicits a humoral immune response, but not a cellular immune response against RSV.


In one embodiment, an immunogen of the present invention comprises less than 12 consecutive (also referred to herein as contiguous or adjacent) amino acids from the sequence of the RSV F protein. In one embodiment, an immunogen of the present invention comprises less than 11 consecutive amino acids from the sequence of the RSV F protein. In one embodiment, an immunogen of the present invention comprises less than 10 consecutive amino acids from the sequence of the RSV F protein. In one embodiment, an immunogen of the present invention comprises less than 9 consecutive amino acids from the sequence of the RSV F protein. In one embodiment, an immunogen of the present invention comprises less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, or less than 3 contiguous amino acids from the sequence of the RSV F protein. In one embodiment, an immunogen of the present invention does not comprise an amino acid sequence from the RSV F protein that lies outside of the antibody-binding domain and that could elicit a cellular immune response to the RSV F protein. In a preferred embodiment, an immunogen of the present invention comprises less than 7, less than 6, less than 5, less than 4, or less than 3 contiguous amino acids from sequences outside of amino acids 254-277 from SEQ ID NO:1. In another preferred embodiment, an immunogen of the present invention comprises less than 7, less than 6, less than 5, less than 4, or less than 3 contiguous amino acids from outside of amino acids 427-436 from SEQ ID NO:1.


As described above, antibody-binding domains contain contact residues. As used herein, a contact residue is any amino acid present in a molecule (e.g., a peptide or antibody) that interacts directly or indirectly (e.g., forms an ionic bond either directly, or indirectly through a salt bridge), with an amino acid in a second molecule (e.g., a peptide or antibody), thereby resulting in formation of a complex between the two molecules. Preferably, immunogens of the present invention have contact residues capable of binding to the contact residues in motavizumab, or 101F, that are responsible for the binding of the antibody to the RSV F protein peptide. This disclosure provides immunogens of the embodiments that have contact residues capable of binding to the contact residues in motavizumab that are responsible for the binding of the antibody to the RSV F protein peptide. The disclosure also provides immunogens of the embodiment that have contact residues capable of binding to the contact residues in 101F that are responsible for the binding of the antibody to the RSV F protein peptide.


One embodiment of the present invention is an immunogen comprising a motavizumab-binding domain, wherein the contact residues within such motavizumab-binding domain have a spatial orientation represented by atomic coordinates defining backbone atoms, wherein each atom has a root mean square deviation of less than 10 angstroms, less than 5 angstroms, less than 2 angstroms, less than 1 angstrom, less than 0.4 angstroms, less than 0.3 angstroms, less than 0.2 angstroms, less or than 0.1 angstrom from the corresponding backbone atom in the antibody-binding domain of an RSV F peptide in a complex defined by the coordinates defined by the coordinates specified in PDB acc code 3IXT. In one embodiment, an immunogen comprises a motavizumab-binding domain that comprises contact residues that have a spatial orientation represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms from the corresponding backbone atoms of contact residues in an RSV F peptide that contacts motavizumab in a complex defined by the coordinates specified in PDB acc code 3IXT.


The disclosure includes an immunogen of the embodiments in which the motavizumab-binding domain from such immunogen comprises less than 15 amino acids of the motavizumab-binding domain from the RSV F peptide and in which such amino acids are in clusters of no more than 8 consecutive amino acids per cluster. The disclosure also includes an immunogen of the embodiments in which the motavizumab-binding domain from such immunogen comprises less than 15 amino acids of the motavizumab-binding domain from the RSV F peptide, and in which such amino acids are in clusters of no more than 3 consecutive amino acids per cluster. Also included are immunogens in which such amino acids are in clusters of no more than 7, 6, 5, 4, 2 or 1 consecutive amino acids per cluster.


Another embodiment of the present invention is an immunogen comprising an 101F-binding domain, wherein the contact residues within such 101F-binding domain have a spatial orientation represented by atomic coordinates defining backbone atoms, wherein each atom has a root mean square deviation of less than 10 angstroms, less than 5 angstroms, less than 2 angstroms, less than 1 angstrom, less than 0.4 angstroms, less than 0.3 angstroms, less than 0.2 angstroms, or less than 0.1 angstrom from the corresponding backbone atom in the antibody-binding domain of an RSV F peptide in a complex defined by the coordinates recited in PDB acc code 3O41. In one embodiment, an immunogen comprises a 101F antibody-binding domain that comprises contact residues that have a spatial orientation represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms from the corresponding backbone atoms of contact residues in an RSV F peptide that contacts 101F antibody in a complex defined by the coordinates recited in PDB acc code 3O41.


The disclosure includes an immunogen of the embodiments in which the 101F-binding domain from such immunogen comprises no more than 10 amino acids of the 101F-binding domain from the RSV F peptide, and in which such amino acids are in clusters of no more than 8 consecutive amino acids per cluster. Also included are immunogens in which such amino acids are in clusters of no more than 7, 6, 5, 4, 3, 2 or 1 consecutive amino acids per cluster.


In order to produce immunogens described herein, the inventors have developed novel methods of identifying proteins that comprise regions, referred to as superpositions, that spatially correspond to an antibody-binding domain from an RSV F peptide present in a complex defined by the coordinates in PDB acc code 3IXT or in PDB acc code 3O41. Such proteins are referred to as scaffolds, protein scaffolds, scaffold proteins, scaffold protein sequences, and the like. Scaffold proteins are useful for creating immunogens of the present invention in that they hold contact residues in the immunogen in the proper spatial orientation to facilitate interaction between such residues and contact residues of motavizumab, or 101F. Moreover, the selection criteria select only those proteins that substantially or wholly lack immunodominant RSV epitopes that would elicit a cellular immune response. This method, which is referred to as superpositioning, comprises determining the three-dimensional structure of an epitope of interest and then computationally searching a database of known protein structures to identify those proteins that can be structurally superimposed onto the epitope of interest with minimal root mean square deviation of their coordinates. Such a method can be accomplished using software such as, for example, ROSETTA. Superpositioning has been described in PCT International Publication No. WO 2008/025015 A2, published Feb. 28, 2008, which is hereby incorporated by reference in its entirety. Once suitable scaffold proteins have been identified, they can be altered according to the methods disclosed herein. A related method that can be used for the analysis of complex epitopes is referred to as double superpositioning. In this method, which is similar to superpositioning, scaffolds are scanned for structural similarity to each of the two epitope segments individually, and whenever a match is found to one of the segments, that scaffold is searched a second time for structural similarity to the other epitope segment, with the rigid body position of the second epitope segment relative to the scaffold pre-determined by the superposition of the first segment to the scaffold. “Double” matches are identified if (a) one epitope segment matches a scaffold with backbone rmsd/nsup<threshold 1 and the other segment matches with backbone rmsd/nsup<threshold 2, where threshold 1 is typically 0.15 and threshold 2 is typically 0.2, or (b) if both segments are superimposed simultaneously onto the scaffold and the backbone rmsd/nsup is <threshold 3, where threshold 3 is typically 0.2. In this instance, the term “backbone rmsd” is defined as the root-mean square deviation of a structural alignment computed over the backbone atoms N, CA, C, O; and “backbone rmsd/nsup” is defined as the “backbone rmsd” divided by the number of aligned residues.


One embodiment of the present invention is an immunogen in which contact residues of the motavizumab, or 101F, binding domain, are embedded in a protein scaffold that spatially corresponds to an antibody-binding domain from an RSV F peptide present in a complex defined by the coordinates in PDB acc code 3IXT or in PDB acc code 3O41, respectively. Such a protein scaffold can be identified using the three-dimensional structure of a complex described by the coordinates in PDB acc code 3IXT or in PDB acc code 3O41. As used herein, embedding of contact residues in a protein scaffold refers to positioning contact residues within the scaffold such that such contact residues form an antibody-binding domain and such that the protein scaffold retains its proper three-dimensional structure.


Analysis of the three-dimensional structure of the complex of the F peptide bound to motavizumab described by the coordinates in PDB acc cod 3IXT shows that the contact residues are embedded in a three-dimensional structure comprising two alpha helices. Thus, one embodiment of the present invention is an immunogen comprising an antibody-binding domain that binds motavizumab, wherein the contact residues in such antibody-binding domain are embedded in a protein scaffold comprising a three-dimensional structure that has two alpha helices, wherein the helices are defined by atomic coordinates defining backbone atoms, wherein each atom has a root mean square deviation of less than 10 angstroms, less than 5 angstroms, less than 2 angstroms, less than 1 angstrom, less than 0.4 angstroms, less than 0.3 angstroms, less than 0.2 angstroms, or less than 0.1 angstrom from the corresponding backbone atom in the antibody-binding domain of an RSV F peptide in a complex defined by the coordinates recited in PDB acc cod 3IXT. In one embodiment, the alpha helices consist of amino acids 2-10 and amino acids 15-23 of the F peptide.


The disclosure provides an immunogen of the embodiments in which contact residues of the motavizumab-binding domain are embedded in a protein scaffold comprising a three-dimensional structure having two alpha helices defined by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the two alpha helices of the peptide consisting of amino acid sequence SEQ ID NO:2 when complexed with motavizumab, the three-dimensional model of such complex being defined by the coordinates specified in PDB acc cod 3IXT. The disclosure also provides an immunogen of the embodiments in which contact residues of the 101F antibody-binding domain are embedded in a protein scaffold comprising a three-dimensional structure with atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the peptide consisting of amino acid sequence SEQ ID NO:4 when complexed with 101F antibody, the 3-dimensional model of such complex being defined by the coordinates specified in PDB acc code 3O41. The disclosure also provides an immunogen of the embodiments in which contact residues of the 101F antibody-binding domain are embedded in a protein scaffold comprising a three-dimensional structure with atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the peptide consisting of amino acid sequence SEQ ID NO:9 when complexed with 101F antibody, the 3-dimensional model of such complex being defined by the coordinates specified in PDB acc code 3O45.


As has been described, scaffold proteins are identified by their spatial similarity to the three-dimensional structure of a motavizumab or 101F binding domain. Moreover, preferable scaffold sequences do not share significant homology with the RSV F protein (i.e., they do not elicit a cellular immune response against RSV). Thus, using the techniques described herein, the inventors have now identified several proteins that can serve as scaffolds for creating motavizumab-binding and 101F-binding immunogens. Examples of such proteins include, but are not limited to, Staphylococcus aureus protein, Helicobacter pylori CagZ protein, and equine infectious anemia virus p26 protein. One embodiment is a scaffold protein having PDB (Protein Data Bank) accession code 1LP1, preferably scaffold 1LP1_b (SEQ ID NO:11). Another embodiment is a scaffold protein having PDB accession code 1S2X, preferably scaffold 1S2X_a (SEQ ID NO:12). One embodiment is a truncated 1 S2X_a scaffold that has SEQ ID NO:13; this scaffold is truncated at the carboxyl terminus. Another embodiment is a scaffold protein having PDB accession code 2EIA, preferably scaffold 2EIA_a (SEQ ID NO:14).


It will be understood by those in the relevant art that while a protein scaffold may comprise a three-dimensional structure capable of holding contact residues in the correct spatial position, and since such a scaffold protein may be unrelated to the RSV F protein, the scaffold protein itself may not contain amino acids that spatially correspond to contact residues in the F protein. Consequently, an unmodified, scaffold protein may not be able to bind to motavizumab or 101F. As used herein the term unmodified scaffold protein is a scaffold protein represented by a three-dimensional model, a portion of which spatially corresponds to the antibody-binding domain of an RSV F protein in a complex defined by the atomic coordinates in PDB acc cod 3IXT or in PDB acc cod 3O41, but which has not been altered to contain any of the contact residues present in the RSV F protein. Amino acids in the RSV F protein identified as interacting with the contact residues in motavizumab are the amino acids at positions 255, 258, 259, 261, 262, 263, 267, 268, 269, 271, 272, 273, 275 and 276 of SEQ ID NO:1. Amino acids in the RSV F protein identified as interacting with the contact residues in 101F are the amino acids at positions 427, 429, 431, 432, 433, 434, 435 and 436 of SEQ ID NO:1. Thus one embodiment of the present invention is an immunogen that comprises sequence from a scaffold protein, wherein at least one amino acid in such scaffold protein sequence spatially corresponding to a contact residue in the RSV F protein is substituted with the amino acid residue present at such spatially corresponding contact residue in the F peptide. Such an immunogen can be produced by recombinant methods and/or synthesizing a nucleic acid molecule that encodes such immunogen and expressing it to make a recombinant immunogen. Such an immunogen can be tested for efficacy by measuring the immunogen's ability to bind to its respective antibody, or to neutralize RSV, using techniques known to those skilled in the art.


One preferred embodiment of the present invention is a scaffold protein comprising at least one substitution selected from the group consisting of:


(a) substituting a serine for the amino acid spatially corresponding to the amino acid at position 2 of SEQ ID NO:2;


(b) substituting a leucine for the amino acid spatially corresponding to the amino acid at position 5 of SEQ ID NO:2;


(c) substituting a serine for the amino acid spatially corresponding to the amino acid at position 6 of SEQ ID NO:2;


(d) substituting a isoleucine for the amino acid spatially corresponding to the amino acid at position 8 of SEQ ID NO:2;


(e) substituting a asparagine for the amino acid spatially corresponding to the amino acid at position 9 of SEQ ID NO:2;


(f) substituting a aspartic acid for the amino acid spatially corresponding to the amino acid at position 10 of SEQ ID NO:2;


(g) substituting a threonine for the amino acid spatially corresponding to the amino acid at position 14 of SEQ ID NO:2;


(h) substituting a asparagine for the amino acid spatially corresponding to the amino acid at position 15 of SEQ ID NO:2;


(i) substituting a aspartic acid for the amino acid spatially corresponding to the amino acid at position 16 of SEQ ID NO:2;


(j) substituting a lysine for the amino acid spatially corresponding to the amino acid at position 18 of SEQ ID NO:2;


(k) substituting a lysine for the amino acid spatially corresponding to the amino acid at position 19 of SEQ ID NO:2;


(l) substituting a leucine for the amino acid spatially corresponding to the amino acid at position 20 of SEQ ID NO:2;


(m) substituting a serine for the amino acid spatially corresponding to the amino acid at position 22 of SEQ ID NO:2; and


(n) substituting a asparagine for the amino acid spatially corresponding to the amino acid at position 23 of SEQ ID NO:2.


One embodiment is a scaffold protein comprising all of the afore-mentioned substitutions.


Another preferred embodiment of the present invention is a scaffold protein comprising at least one substitution selected from the group consisting of:


(a) substituting a lysine for the amino acid spatially corresponding to the amino acid at position 1 of SEQ ID NO:4;


(b) substituting a arginine for the amino acid spatially corresponding to the amino acid at position 3 of SEQ ID NO:4;


(c) substituting a isoleucine for the amino acid spatially corresponding to the amino acid at position 5 of SEQ ID NO:4;


(d) substituting a isoleucine for the amino acid spatially corresponding to the amino acid at position 6 of SEQ ID NO:4;


(e) substituting a lysine for the amino acid spatially corresponding to the amino acid at position 7 of SEQ ID NO:4;


(f) substituting a threonine for the amino acid spatially corresponding to the amino acid at position 8 of SEQ ID NO:4;


(g) substituting a phenylalanine for the amino acid spatially corresponding to the amino acid at position 9 of SEQ ID NO:4; and,


(h) substituting a serine for the amino acid spatially corresponding to the amino acid at position 10 of SEQ ID NO:4.


One embodiment is a scaffold protein comprising all of the afore-mentioned substitutions.


In one embodiment, an immunogen comprises amino acids 2, 5, 6, 8, 9, 16, 18, 19, 20, 22 and 23 of SEQ ID NO:2 substituted at the spatially corresponding positions of a scaffold protein. In one embodiment, an immunogen comprises amino acids 2, 5, 6, 9, 16, 18, 19, 20, 22 and 23 of SEQ ID NO:2 substituted at the spatially corresponding positions of a scaffold protein.


One embodiment of the present invention is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b001 (SEQ ID NO:18), 1lp1b002 (SEQ ID NO:21), 1lp1b003 (SEQ ID NO:24), 1lp1b004 (SEQ ID NO:149), 1s2xa001 (SEQ ID NO:152), 1s2xa002 (SEQ ID NO:155), 1s2xa003 (SEQ ID NO:158), 1s2xa004 (SEQ ID NO:164), 2eiaa001 (SEQ ID NO:167), and 2eiaa002 (SEQ ID NO:170), the amino acid sequences of which are disclosed in the Examples. One embodiment is an immunogen comprising 1lp1b001 (SEQ ID NO:18). One embodiment is an immunogen comprising 1lp1b002 (SEQ ID NO:21). One embodiment is an immunogen comprising 1lp1b003 (SEQ ID NO:24). One embodiment is an immunogen comprising 1lp1b004 (SEQ ID NO:149). One embodiment is an immunogen comprising 1s2xa001 (SEQ ID NO:152). One embodiment is an immunogen comprising 1s2xa002 (SEQ ID NO:155). One embodiment is an immunogen comprising 1s2xa003 (SEQ ID NO:158). One embodiment is an immunogen comprising 1s2xa004 (SEQ ID NO:164). One embodiment is an immunogen comprising 2eiaa001 (SEQ ID NO:167). One embodiment is an immunogen comprising 2eiaa002 (SEQ ID NO:170). One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


One embodiment is an immunogen comprising an amino acid sequence of protein 1s2xa003_PADRE (SEQ ID NO:161). One embodiment is a variant of such immunogen, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


One embodiment is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1s2xa001_N_His (SEQ ID NO:177), 1s2xa002_N_His (SEQ ID NO:178), 1s2xa003_N_His (SEQ ID NO:179), 1s2xa004_N_His (SEQ ID NO:180), and 2eiaa002_N_His (SEQ ID NO:181), the amino acid sequences of which are disclosed in the Examples. One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


In addition to substituting amino acids in the portion of the scaffold protein spatially corresponding to the antibody-binding domain of RSV F peptide, other changes can be made to the immunogen as well, as long as such changes do not reduce the affinity of motavizumab or 101F for the immunogen. For example, amino acid residues outside of the antibody-binding domain may be altered in order to reduce steric interference between the backbone atoms of the antibody and the backbone atoms of the scaffold protein portion of the immunogen. In addition, amino acids that are outside of the antibody-binding domain can be substituted with an amino acid that allows the formation of a new ionic bond, thereby strengthening the interaction between the immunogen and the antibody. Such alteration of the scaffold protein is herein referred to as epitope conformation stabilization.


One embodiment of the invention is an immunogen comprising a motavizumab-binding domain spatially corresponding to a motavizumab-binding domain from an RSV F peptide, wherein the scaffold protein sequence of such immunogen has been subject to epitope stabilization conformation to reduce steric hindrance and/or increase the affinity of the immunogen for motavizumab. Another embodiment of the invention is an immunogen comprising a 101F-binding domain spatially corresponding to a 101F-binding domain from an RSV F peptide, wherein the scaffold protein sequence of such immunogen has been subject to epitope stabilization conformation to reduce steric hindrance and/or increase the affinity of the immunogen for 101F.


In addition to alterations of the immunogen sequence that result in reduced steric hindrance and/or increase the affinity of the immunogen for motavizumab of 101F, the immunogen can be subject to a process referred to as resurfacing, addition of N-linked glycosylation sites, or PEGylation. As used herein, the term resurfacing refers to a process whereby amino acid substitutions are introduced into scaffold sequences that are outside of the antibody-binding domain in order to eliminate or hide immunodominant epitopes. For example, amino acids within an immunodominant epitope can be substituted with neutral amino acids (i.e., having an uncharged R group) so that the epitope is no longer bound by an antibody. In some embodiments, N-linked glycosylation sites can be introduced into the protein, resulting in glycosylation of the immunogen such that immunodominant epitopes are hidden from the immune system and thus do not elicit a strong humoral or cell mediated immune response. In some embodiments, a scaffold can be PEGylated (i.e., treated with polyethylene glycol), or otherwise treated, to mask immunodominant epitopes. Such processes can also be referred to as cloaking. Methods of producing resurfaced proteins have been previously described in, for example, PCT International Publication No. WO/2009/100376 entitled, “Antigenic Cloaking and Its Use”, published Aug. 13, 2009, which is hereby incorporated by reference in its entirety. As used herein, the phrase immunodominant epitope refers to an epitope within a protein or peptide that is most easily recognized by the immune system and thus has the greatest influence on the specificity of an antibody elicited by a protein or peptide containing the immunodominant epitope.


One embodiment of the present invention is an immunogen comprising sequences from a scaffold protein, wherein such immunogen binds motavizumab or 101F, and wherein scaffold protein sequences outside of the antibody-binding domain of such immunogen have been subject to resurfacing. In one embodiment, amino acids in the scaffold protein sequences of the immunogen are substituted with neutral amino acids. In another embodiment, glycosylation sites are introduced into scaffold protein sequences of the immunogen, or the immunogen is submitted to PEGylation methodology such that immunodominant epitopes present in the immunogen are hidden from the immune system by glycosylation or PEGylation of the immunogen. It should be appreciated that immunogens of the present invention can comprise combinations of the amino acid alterations discussed above. Whether scaffold protein sequences will require the introduction of neutral amino acids, glycosylation or PEGylation or combinations of such types of alterations depends on the nature of the sequences present in the scaffold protein. It is within the ability of those skilled in the art to determine which alterations will best eliminate or hide immunodominant epitopes outside of the antibody-binding domain. Moreover, methods of substituting amino acids into a protein or peptide, introducing glycosylation sites into a protein or peptide or PEGylating such protein or peptide are known to those skilled in the art.


A preferred embodiment of the present invention is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of mota1lp1b.m1.c1.d1_glyc1 (SEQ ID NO:55), mota1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56), mota1lp1b.m1.c1.d1_des11 (SEQ ID NO:66), mota1lp1b.m1.c1.d1_des12 (SEQ ID NO:67), mota1lp1b.m1.c1.d1_des13 (SEQ ID NO:68), mota1lp1b.m1.c1.d1_des15 (SEQ ID NO:69), mota1lp1b.m1.c1.d1_des16 (SEQ ID NO:70), mota1lp1b.m1.c1.d1_des17 (SEQ ID NO:71), mota1lp1b.m1.c1.d1_des18 (SEQ ID NO:72), mota1lp1b.m1.c1.d1_des19 (SEQ ID NO:73), mota1lp1b.m1.c1.d1_des110 (SEQ ID NO:74), and mota1lp1b.m1.c1.d1_des111 (SEQ ID NO:75), the amino acid sequences of which are disclosed in the Examples.


One embodiment is an immunogen that has one or more N-linked glycosylation sites, such as, but not limited to, mota1lp1b.m1.c1.d1 glyc1 (SEQ ID NO:55), mota1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56), 1lp1b003_Glyc1 (SEQ ID NO:39), 1lp1b003_Glyc2 (SEQ ID NO:42), 1lp1b003_Glyc3 (SEQ ID NO:45), 1lp1b003_Glyc4 (SEQ ID NO:48), 1lp1b003_Glyc5 (SEQ ID NO:51), or 1lp1b003_Glyc6 (SEQ ID NO:54). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_glyc1 (SEQ ID NO:55). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56). One embodiment is an immunogen comprising 1lp1b003_Glyc1 (SEQ ID NO:39). One embodiment is an immunogen comprising 1lp1b003_Glyc2 (SEQ ID NO:42). One embodiment is an immunogen comprising 1lp1b003_Glyc3 (SEQ ID NO:45). One embodiment is an immunogen comprising 1lp1b003_Glyc4 (SEQ ID NO:48). One embodiment is an immunogen comprising 1lp1b003_Glyc5 (SEQ ID NO:51). One embodiment is an immunogen comprising 1lp1b003_Glyc6 (SEQ ID NO:54). One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


One embodiment is an immunogen that has been resurfaced, such as, but not limited to, mota1lp1b.m1.c1.d1_des11 (SEQ ID NO:66), mota1lp1b.m1.c1.d1_des12 (SEQ ID NO:67), mota1lp1b.m1.c1.d1_des13 (SEQ ID NO:68), mota1lp1b.m1.c1.d1_des15 (SEQ ID NO:69), mota1lp1b.m1.c1.d1_des16 (SEQ ID NO:70), mota1lp1b.m1.c1.d1_des17 (SEQ ID NO:71), mota1lp1b.m1.c1.d1_des18 (SEQ ID NO:72), mota1lp1b.m1.c1.d1_des19 (SEQ ID NO:73), mota1lp1b.m1.c1.d1_des110 (SEQ ID NO:74), or mota1lp1b.m1.c1.d1_des111 (SEQ ID NO:75). Additional examples are 1lp1b003_Surf1 (SEQ ID NO:59), 1lp1b003_Surf6 (SEQ ID NO:62), and 1lp1b003_Surf8 (SEQ ID NO:65). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des11 (SEQ ID NO:66). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des12 (SEQ ID NO:67). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des13 (SEQ ID NO:68). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des15 (SEQ ID NO:69). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des16 (SEQ ID NO:70). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des17 (SEQ ID NO:71). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des18 (SEQ ID NO:72). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des19 (SEQ ID NO:73). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des110 (SEQ ID NO:74). One embodiment is an immunogen comprising mota1lp1b.m1.c1.d1_des111 (SEQ ID NO:75). One embodiment is an immunogen comprising 1lp1b003_Surf1 or 1lp1b003_Surf8. One embodiment is an immunogen comprising 1lp1b003_Surf1 (SEQ ID NO:59). One embodiment is an immunogen comprising 1lp1b003_Surf6 (SEQ ID NO:62). One embodiment is an immunogen comprising 1lp1b003_Surf8 (SEQ ID NO:65). One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


The disclosure provides for immunogens of the embodiments that are multivalent. Without being bound by theory, it is believed that multivalent immunogens can elicit enhanced neutralizing antibody responses. A multivalent immunogen of the embodiments is an immunogen of the disclosure that includes a particle enabling attachment of one or more immunogens. Such a particle can be of a material known to those skilled in the art. Examples of particles include, but are not limited to, ferritin, viral capsid proteins, virus-like particles, and other proteins that assemble into high-copy, large particles. Such attachment is accomplished so as to not significantly reduce the ability of an immunogen of the embodiments to elicit a neutralizing humoral response against RSV. Such attachment can be accomplished by covalently binding an immunogen to such a particle or can be accomplished by designing a nucleic acid molecule than encodes an immunogen of the embodiments and a particle, or subunit thereof. In one embodiment, a multivalent immunogen can be administered as a prime and/or boost. In one embodiment, a multivalent immunogen can be administered as a prime. In one embodiment, a multivalent immunogen can be administered as a boost.


One embodiment is an immunogen of the embodiments that is attached to ferritin. Ferritin, a globular protein complex consisting of 24 protein subunits, is a ubiquitous intracellular protein that stores iron and releases it in a controlled manner. The use of ferritin fusion proteins as vaccines has been described, for example, by Carter D C, et al., U.S. Pat. No. 7,097,841 B2, issued Aug. 29, 2006. One embodiment is a multivalent immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003 ferritin (SEQ ID NO:138), 1lp1b003_eumS (SEQ ID NO:140), 1lp1b003_eumSP (SEQ ID NO:142), 1lp1b003_eumL (SEQ ID NO:144), and 1lp1b003_eumLP (SEQ ID NO:146). One embodiment is an immunogen comprising 1lp1b003 ferritin (SEQ ID NO:138). One embodiment is an immunogen comprising 1lp1b003_eumS (SEQ ID NO:140). One embodiment is an immunogen comprising 1lp1b003_eumSP (SEQ ID NO:142). One embodiment is an immunogen comprising 1lp1b003_eumL (SEQ ID NO:144). One embodiment is an immunogen comprising 1lp1b3_eumLP (SEQ ID NO:146). One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


The disclosure also provides an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003_K46A (SEQ ID NO:78), 1lp1b003_Q52A (SEQ ID NO:81), 1lp1b003_I13L_F27A (SEQ ID NO:87), 1lp1b003_L41I_L42V (SEQ ID NO:90), 1lp1b-003_L41I_L42A (SEQ ID NO:93), 1lp1b003_I13A (SEQ ID NO:105), 1lp1b003_L16A (SEQ ID NO:108), 1lp1b003_F27A (SEQ ID NO:111), 1lp1b003_L41A (SEQ ID NO:114), 1lp1b003_L42A (SEQ ID NO:117), and 1lp1b003_Neg1 (SEQ ID NO:132). One embodiment is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003_C43 (SEQ ID NO:33), 1lp1b003_C47 (SEQ ID NO:36), 1lp1b003_Glyc3 (SEQ ID NO:45), 1lp1b003_Glyc4 (SEQ ID NO:48), 1lp1b003_Glyc5 (SEQ ID NO:51), 1lp1b003_Glyc6 (SEQ ID NO:54), 1lp1b003_K46A_Q52A (SEQ ID NO:84), lp1b003_Glycine1 (SEQ ID NO:120), 1lp1b003_Glycine2 (SEQ ID NO:123), 1lp1b003_Pos1 (SEQ ID NO:126), 1lp1b003 Pos2 (SEQ ID NO:129), and 1lp1b003_Neg2 (SEQ ID NO:135). One embodiment is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003_I13A_L42A (SEQ ID NO:96), 1lp1b003 L19A (SEQ ID NO:99), and 1lp1b003_L19A_L41I (SEQ ID NO:102). Such immunogens differ from immunogen 1lp1b003 with respect to surface charge, glycosylation pattern and intrinsic flexibility. One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


One embodiment is an immunogen comprising an amino acid sequence of protein 1lp1b003_K272E (SEQ ID NO:30). This immunogen lacks a key contact residue of the motavizumab-binding domain.


One embodiment is an immunogen comprising an amino acid sequence of protein 1lp1b003 F2Y H15N (SEQ ID NO:27). This immunogen was expressed in HEK293 cells and complexed with motavizumab, crystallized, and a three-dimensional model defined therefrom.


The disclosure provides an RSV immunogen that comprises an amino acid sequence of 1LP1_b (SEQ ID NO: 11) having from one to twenty amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of:


(a) substitution of a serine at amino acid position N-25 in SEQ ID NO:11;


(b) substitution of a leucine at amino acid position 1-28 in SEQ ID NO:11;


(c) substitution of a serine at amino acid position Q-29 in SEQ ID NO: 11;


(d) substitution of an isoleucine at amino acid position L-31 in SEQ ID NO:11;


(e) substitution of an asparagine at amino acid position K-32 in SEQ ID NO:11;


(f) substitution of an aspartic acid at amino acid position K-4 in SEQ ID NO:11;


(g) substitution of a lysine at amino acid position Q-6 in SEQ ID NO:11;


(h) substitution of a lysine at amino acid position Q-7 in SEQ ID NO:11;


(i) substitution of a leucine at amino acid position N-8 in SEQ ID NO:11;


(j) substitution of a serine at amino acid position F-10 in SEQ ID NO:11; and


(k) substitution of an asparagine at amino acid position Y-11 in SEQ ID NO:11. Such an RSV immunogen can comprise all of the substitutions (a) through (k). An RSV immunogen can elicit a humoral immune response against RSV. The phrase “from one to twenty amino acid substitutions” allows for substitutions beyond those that are specified in (a) through (k). In one embodiment, the three-dimension structure of the antibody-binding domain is maintained or improved when such substitution(s) are made. One embodiment is an RSV immunogen that comprises an amino acid sequence that has up to nine substitutions (i.e., any number ranging from 0 through 9 substitutions) in an amino acid sequence of a protein selected from the group consisting of 1lp1b001 (SEQ ID NO:18), 1lp1b002 (SEQ ID NO:21), 1lp1b003 (SEQ ID NO:24), and 1lp1b004 (SEQ ID NO:149). One embodiment is an RSV immunogen that comprises an amino acid sequence that has up to twelve substitutions in the amino acid sequence of protein 1lp1b003 (SEQ ID NO:24). One embodiment is an RSV immunogen that comprises an amino acid sequence that is at least 95% identical to an amino acid sequence of a protein selected from the group consisting of 1lp1b001 (SEQ ID NO:18), 1lp1b002 (SEQ ID NO:21), 1lp1b003 (SEQ ID NO:24), and 1lp1b004 (SEQ ID NO:149). One embodiment is an RSV immunogen that comprises an amino acid sequence that is at least 95% identical to the amino acid sequence of 1lp1b003 (SEQ ID NO:24). One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of 1lp1b001 (SEQ ID NO:18), 1lp1b002 (SEQ ID NO:21), 1lp1b003 (SEQ ID NO:24), and 1lp1b004 (SEQ ID NO:149). One embodiment is an RSV immunogen comprising 1lp1b003; such an immunogen comprises amino acid sequence SEQ ID NO:24. One embodiment is an RSV immunogen that is N-linked glycosylated. One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of mota1lp1b.m1.c1.d1_glyc1 (SEQ ID NO:55), mota1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56), 1lp1b003_Glyc1 (SEQ ID NO:39), 1lp1b003_Glyc2 (SEQ ID NO:42), 1lp1b003_Glyc3 (SEQ ID NO:45), 1lp1b003_Glyc4 (SEQ ID NO:48), 1lp1b003_Glyc5 (SEQ ID NO:51), and 1lp1b003_Glyc6 (SEQ ID NO:54). One embodiment is an RSV immunogen that is resurfaced. One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of mota1lp 1b.m1.c1.d1_des11 (SEQ ID NO:66), mota1lp1b.m1.c1.d1_des12 (SEQ ID NO:67), mota1lp1b.m1.c1.d1_des13 (SEQ ID NO:68), mota1lp1b.m1.c1.d1_des15 (SEQ ID NO:69), mota1lp1b.m1.c1.d1_des16 (SEQ ID NO:70), mota1lp1b.m1.c1.d1_des17 (SEQ ID NO:71), mota1lp1b.m1.c1.d1_des18 (SEQ ID NO:72), mota1lp1b.m1.c1.d1_des19 (SEQ ID NO:73), mota1lp1b.m1.c1.d1_des110 (SEQ ID NO:74), mota1lp1b.m1.c1.d1_des111 (SEQ ID NO:75), 1lp1b003_Surf1 (SEQ ID NO:59), 1lp1b003_Surf6 (SEQ ID NO:62), and 1lp1b003_Surf8 (SEQ ID NO:65). One embodiment is an RSV immunogen that is a multivalent immunogen. One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of 1lp1b003 ferritin (SEQ ID NO:138), 1lp1b003_eumS (SEQ ID NO:140), 1lp1b003_eumSP (SEQ ID NO:142), 1lp1b003_eumL (SEQ ID NO:144), and 1lp1b003_eumLP (SEQ ID NO:146). One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of 1lp1b003_K46A (SEQ ID NO:78), 1lp1b003_Q52A (SEQ ID NO:81), 1lp1b003_I13L_F27A (SEQ ID NO:87), 1lp1b003_L41I_L42V (SEQ ID NO:90), 1lp1b-003_L41I_L42A (SEQ ID NO:93), lp1b003_I13A (SEQ ID NO:105), 1lp1b003_L16A (SEQ ID NO:108), 1lp1b003_F27A (SEQ ID NO:111), 1lp1b003_L41A (SEQ ID NO:114), 1lp1b003_L42A (SEQ ID NO:117), and 1lp1b003_Neg1 (SEQ ID NO:132). One embodiment is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003_C43 (SEQ ID NO:33), 1lp1b003_C47 (SEQ ID NO:36), 1lp1b003_Glyc3 (SEQ ID NO:45), 1lp1b003_Glyc4 (SEQ ID NO:48), 1lp1b003_Glyc5 (SEQ ID NO:51), 1lp1b003_Glyc6 (SEQ ID NO:54), 1lp1b003_K46A_Q52A (SEQ ID NO:84), lp1b003_Glycine1 (SEQ ID NO:120), 1lp1b003_Glycine2 (SEQ ID NO:123), 1lp1b003_Pos1 (SEQ ID NO:126), 1lp1b003_Pos2 (SEQ ID NO:129), and 1lp1b003_Neg2 (SEQ ID NO:135). One embodiment is an immunogen comprising an amino acid sequence of a protein selected from the group consisting of 1lp1b003_I13A_L42A (SEQ ID NO:96), 1lp1b003_L19A (SEQ ID NO:99), and 1lp1b003_L19A_L41I (SEQ ID NO:102). An RSV immunogen disclosed herein can elicit a humoral immune response against RSV. In one embodiment, such an RSV immunogen can elicit a humoral immune response against RSV, but does not elicit a cellular immune response. An RSV immunogen as disclosed herein can have a motavizumab antibody binding domain comprising less than 12 consecutive amino acids from a motavizumab antibody binding domain of RSV fusion protein. In one embodiment, an RSV immunogen has a motavizumab antibody-binding domain comprising less than 9 consecutive amino acids from a motavizumab antibody-binding domain of RSV fusion protein.


The disclosure provides an RSV immunogen comprising an amino acid sequence of truncated 1S2X_a (SEQ ID NO:13) having from one to twenty-five amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of:


(a) substitution of a serine at amino acid position 92 in SEQ ID NO:13;


(b) substitution of a leucine at amino acid position 95 in SEQ ID NO:13;


(c) substitution of a serine at amino acid position 96 in SEQ ID NO:13;


(d) substitution of an isoleucine at amino acid position 98 in SEQ ID NO:13;


(e) substitution of an asparagine at amino acid position 99 in SEQ ID NO:13;


(f) substitution of an aspartic acid at amino acid position 100 in SEQ ID NO:13;


(g) substitution of an asparagine at amino acid position 105 in SEQ ID NO:13;


(h) substitution of an aspartic acid at amino acid position 106 in SEQ ID NO:13;


(i) substitution of a lysine at amino acid position 108 in SEQ ID NO:13;


(j) substitution of a lysine at amino acid position 109 in SEQ ID NO:13;


(k) substitution of a leucine at amino acid position 110 in SEQ ID NO:13;


(l) substitution of a serine at amino acid position 112 in SEQ ID NO:13; and


(m) substitution of an asparagine at amino acid position 113 in SEQ ID NO:13. Such an RSV immunogen can comprise all of the substitutions (a) through (m). An RSV immunogen can elicit a humoral immune response against RSV. The phrase “from one to twenty-five amino acid substitutions” allows for substitutions beyond those that are specified in (a) through (m). In one embodiment, the three-dimension structure of the antibody-binding domain is maintained or improved when such substitution(s) are made. One embodiment is an RSV immunogen that comprises an amino acid sequence that has up to twelve substitutions (i.e., any number ranging from 0 through 12 substitutions) in an amino acid sequence of a protein selected from the group consisting of 1s2xa001 (SEQ ID NO:152), 1s2xa002 (SEQ ID NO:155), 1s2xa003 (SEQ ID NO:158), and 1s2xa004 (SEQ ID NO:164). One embodiment is an RSV immunogen that comprises an amino acid sequence that has up to twelve substitutions in the amino acid sequence of protein 1s2xa003 (SEQ ID NO:158). One embodiment is an RSV immunogen that comprises an amino acid sequence that is at least 95% identical to an amino acid sequence of a protein selected from the group consisting of 1s2xa001 (SEQ ID NO:152), 1s2xa002 (SEQ ID NO:155), 1s2xa003 (SEQ ID NO:158), and 1s2xa004 (SEQ ID NO:164). One embodiment is an RSV immunogen that comprises an amino acid sequence that is at least 95% identical to an amino acid sequence of protein 1s2xa003 (SEQ ID NO:158). One embodiment is an RSV immunogen that comprises an amino acid sequence of a protein selected from the group consisting of 1s2xa001 (SEQ ID NO:152), 1s2xa002 (SEQ ID NO:155), 1s2xa003 (SEQ ID NO:158), and 1s2xa004 (SEQ ID NO:164). One embodiment is an RSV immunogen comprising 1s2xa003; such an immunogen comprises amino acid sequence SEQ ID NO:158. One embodiment is an RSV immunogen that is N-linked glycosylated. One embodiment is an RSV immunogen that is resurfaced. One embodiment is an RSV immunogen that is a multivalent immunogen. An RSV immunogen disclosed herein can elicit a humoral immune response against RSV. In one embodiment, such an RSV immunogen can elicit a humoral immune response against RSV, but does not elicit a cellular immune response. An RSV immunogen as disclosed herein can have a motavizumab antibody binding domain comprising less than 12 consecutive amino acids from a motavizumab antibody binding domain of RSV fusion protein. In one embodiment, an RSV immunogen has a motavizumab antibody-binding domain comprising less than 9 consecutive amino acids from a motavizumab antibody-binding domain of RSV fusion protein.


An immunogen of the embodiments does not have an amino acid sequence that consists of SEQ ID NO:1. An immunogen of the embodiments does not have an amino acid sequence that consists of SEQ ID NO:2. An immunogen of the embodiments does not have an amino acid sequence that consists of SEQ ID NO:3. An immunogen of the embodiments does not have an amino acid sequence that consists of SEQ ID NO:9. An immunogen of the embodiments does not have an amino acid sequence that consists of SEQ ID NO:10.


The present invention also discloses an immunogen comprising an RSV F protein that is stabilized in its pre-fusion, trimeric state. Such an immunogen comprises an RSV F protein in which the furin cleavage sites can (but need not be) mutated to reduce or prevent cleavage and a trimerization motif (such as a fibritin T4 trimerization motif) preferably appended to a truncated C terminus lacking the F protein transmembrane and cellular domain so that the resultant RSV F protein remains in a trimeric, pre-fusion conformation.


One embodiment of the present invention is an immunogen comprising an amino acid sequence of a protein selected from RSV F0 Fd (also referred to as RSV F0 Fd) (SEQ ID NO:174), RSV F Fd (SEQ ID NO:175), and RSV F0 Fd GAG (SEQ ID NO:176), the amino acid sequences of which are disclosed in the Examples. One embodiment is an immunogen comprising RSV F0 Fd (SEQ ID NO:174). One embodiment is an immunogen comprising RSV F Fd (SEQ ID NO:175). One embodiment is an immunogen comprising RSV F0 Fd GAG (SEQ ID NO:176). One embodiment is a variant of any of such immunogens, wherein the variant shares at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% amino acid sequence identity with such immunogen. Preferably the variant retains contact residues of such immunogen.


Immunogens of the instant invention can be produced recombinantly or they can be produced synthetically. Also encompassed are immunogens that are combinations of recombinant and synthetic molecules. General methods for producing and isolating recombinant or synthetic proteins or peptides are known to those skilled in the art. It should be noted that, as used herein, an isolated, or biologically pure, molecule, is one that has been removed from its natural milieu. As such the terms isolated, biologically pure, and the like, do not necessarily reflect the extent to which the immunogen has been purified.


An immunogen of the embodiments can also comprise one or more motifs that can aid in purification of the immunogen, processing of the immunogen, and/or the immunogenicity of the immunogen. Examples include, but are not limited to, an HRV3C site, a caspase 3 site, a His tag, a Strep tag, MBP (maltose binding protein) or a functional fragment thereof, a factor Xa site, a TEV site, and a PADRE motif. A PADRE (Pan HLA DR-binding epitope peptide) motif has been shown to elicit T-cell help to stimulate a good antibody response; see, e.g., Alexander J, et al., 1994, Immunity 1, 751-761.


One embodiment is a protein comprising an amino acid sequence of an immunogen of the embodiments. Such a protein can be produced recombinantly or synthetically.


One embodiment of the present invention is a nucleic acid molecule that encodes an immunogen of the present disclosure. Such a nucleic acid molecule comprises a nucleic acid sequence that encodes an amino acid sequence of an immunogen of the embodiments. A nucleic acid molecule of the embodiments can include DNA, RNA, or derivatives of either DNA or RNA. A nucleic acid molecule can encode one or more immunogens of the embodiments. Nucleic acid molecules of the disclosure have been subjected to human manipulation. Such a nucleic acid molecule can be produced using recombinant DNA technology (e.g., polymerase chain reaction (PCR) amplification, cloning), chemical synthesis, or a combination of recombinant DNA technology and chemical synthesis. In one embodiment, a nucleic acid molecule, such as a nucleic acid molecule encoding a scaffold protein, can be modified by inserting, deleting, substituting, and/or inverting one or more nucleotides to yield a nucleic acid molecule that encodes an immunogen of the present invention. A nucleic acid molecule can also be modified to introduce codons that are better recognized by the system used to produce protein from a nucleic acid molecule of the disclosure.


One embodiment is a nucleic acid molecule encoding an immunogen comprising a scaffold protein with one or more contact residues, as described herein, embedded in it. Such embedding can be accomplished using techniques described herein as well as techniques known by one skilled in the art.


Nucleic acid molecules of the present invention can be produced using a number of methods known to those skilled in the art; see, for example, Sambrook J et al., 2001, Molecular Cloning: a Laboratory Manual, 3rd edition, Cold Spring Harbor Laboratory Press, and Ausubel F et al., 1994, Current Protocols in Molecular Biology, John Wiley & Sons. For example, nucleic acid molecules can be modified using a variety of techniques including, but not limited to, classic mutagenesis techniques and recombinant DNA techniques, such as site-directed mutagenesis, chemical treatment of a nucleic acid molecule to induce mutations, restriction enzyme cleavage of a nucleic acid fragment, ligation of nucleic acid fragments, polymerase chain reaction (PCR) amplification and/or mutagenesis of selected regions of a nucleic acid sequence, synthesis of oligonucleotide mixtures and ligation of mixture groups to “build” a mixture of nucleic acid molecules and combinations thereof. Nucleic acid molecules of the embodiments can be selected from a mixture of modified nucleic acids by screening for the function of the protein encoded by the nucleic acid (e.g., the ability of such a nucleic acid molecule to encode an immunogen that binds to motavizumab or 101F).


The disclosure provides a recombinant molecule that comprises a nucleic acid encoding an immunogen of the embodiments operatively linked to at least one transcription control sequence capable of effecting expression of the nucleic acid molecule in a recombinant cell. A recombinant cell is a host cell that is transformed with a recombinant molecule of the embodiments; i.e., a recombinant cell comprises a recombinant molecule. A recombinant molecule can comprise one or more nucleic acid molecules encoding an immunogen of the embodiments operatively linked to one or more transcription control sequences. A recombinant cell can comprise one or more recombinant molecules. In one embodiment, a nucleic acid molecule is operatively linked to a recombinant vector that includes a transcription control sequence to produce a recombinant molecule. Such a vector can be a plasmid vector, a viral vector, or other vector. Such a vector can be DNA, RNA, or a derivative of DNA or RNA. Host cells to transform can be selected based on their ability to effect expression of a nucleic acid molecule of the embodiments. Host cells can also be selected that effect post-translational modifications. Methods to select, produce and use recombinant vectors, recombinant molecules, and recombinant cells of the embodiments are known to those skilled in the art. Proteins and immunogens of the embodiments can be produced by culturing recombinant cells of the embodiments. Methods to effect such production and recovery of such proteins and immunogens are known to those skilled in the art, see for example Sambrook J et al., ibid, and Ausubel, F et al., ibid.


The disclosure also provides a recombinant molecule that is a nucleic acid immunogen or vaccine. That is, such a recombinant molecule can be administered to a subject to elicit a humoral immune response against RSV. Such a response can be a neutralizing humoral immune response. Such a response can be protective. Such a vaccine comprises a recombinant molecule comprising a nucleic acid molecule that encodes an immunogen of the embodiments. In one embodiment, the recombinant molecule is a nucleic acid molecule of the embodiments operatively linked to a recombinant vector. Suitable vectors can be selected by one skilled in the art. Examples include, but are not limited, to adenovirus, adeno-associated virus, cytomegalovirus (CMV), herpes virus, poliovirus, retrovirus, sindbis virus, vaccinia virus, or any other DNA or RNA virus vector.


The present invention also discloses methods of making an immunogen of the present invention. One embodiment is a method that involves using the three-dimensional structure of the antibody-binding domain of a RSV F peptide, when such peptide is bound to an RSV neutralizing antibody, to identify a protein comprising a similar three-dimensional structure, and then substituting the contact residues from the RSV F peptide into the spatially corresponding positions in the native scaffold protein to create an immunogen. In one embodiment, the RSV-neutralizing antibody is palivizumab (SYNAGIS®). In another embodiment, the RSV-neutralizing antibody is motavizumab. In yet another embodiment, the RSV-neutralizing antibody is 101F. In one embodiment the three-dimensional structure of complex between the RSV F peptide and the antibody is represented by atomic coordinates defining backbone atoms that have a root mean square deviation of less than 10 angstroms from the backbone atoms of the complex defined by the coordinates recited in PDB acc code 3IXT or in PDB acc code 3O41. A preferred embodiment of the present invention is a method to produce an immunogen that elicits a potent neutralizing humoral response against RSV, the method comprising:


(a) obtaining a three-dimensional model substantially corresponding to the three-dimensional model represented by the coordinates set forth in PDB acc code 3IXT or in PDB acc code 3O41;


(b) using the model obtained in (a) model to identify a native protein scaffold having a three-dimensional structure represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the three-dimensional model of the RSV F peptide defined by atomic coordinates in PDB acc code 3IXT or in PDB acc code 3O41;


(c) substituting amino acids in the native protein scaffold that spatially correspond to contact residues in the RSV F protein in such three-dimensional model, with the spatially corresponding contact residues in the F protein in such three-dimensional model to create an immunogen containing a transplanted epitope; and


(d) producing said immunogen comprising such transplanted epitope. As has previously been discussed, immunogens produced using the disclosed methods can also be modified to remove sequences related to the RSV F protein, reduce steric hindrance and/or to increase the affinity of the immunogen for motavizumab or 101F. Thus, in one embodiment, the method further comprises modifying the immunogen created in step (c) by substituting amino acids outside of the antibody-binding domain to (a) reduce steric hindrance, (b) introduce new ionic bonds between the immunogen and the antibody, (c) stabilize the protein in a conformation that maintains the transplanted epitope in the spatial conformation found in the three-dimensional model represented by the coordinates in PDB acc code 3IXT or in PDB acc code 3O41. Such a method can also include, but not be limited to, introducing flexibility, N-linked glycosylation sites, positively or negatively charged amino acids, shielding against immunodominant epitopes, or other beneficial features.


The disclosure provides a method to produce an immunogen that elicits a potent neutralizing humoral immune response against RSV. The method comprises:


(a) obtaining a three-dimensional model substantially corresponding to the three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, wherein the model substantially represents the atomic coordinates specified in Protein Data Bank accession code 3IXT;


(b) using the model to identify a candidate protein scaffold having a three-dimensional structure represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the three-dimensional model of the RSV F peptide defined by atomic coordinates in Protein Data Bank accession code 3IXT;


(c) substituting amino acids in the candidate protein scaffold that spatially correspond to contact residues in the RSV F protein in the three-dimensional model, with the spatially corresponding contact residues in the F protein in the three-dimensional model to create a protein containing a transplanted epitope; and


(d) producing the protein comprising the transplanted epitope. The method can further include the step of modifying the protein of step (c) to stabilize the protein in a conformation that maintains the transplanted epitope in the antibody-bound conformation of the three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, wherein the model substantially represents the atomic coordinates specified in Protein Data Bank accession code 3IXT.


The disclosure also provides a method to produce an immunogen that elicits a potent neutralizing humoral immune response against RSV. The method comprises:


(a) obtaining a three-dimensional model substantially corresponding to the three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, wherein the model substantially represents the atomic coordinates specified in Protein Data Bank accession code 3O41;


(b) using the model to identify a candidate protein scaffold having a three-dimensional structure represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the three-dimensional model of the RSV F peptide defined by atomic coordinates in Protein Data Bank accession code 3O41;


(c) substituting amino acids in the candidate protein scaffold that spatially correspond to contact residues in the RSV F protein in the three-dimensional model, with the spatially corresponding contact residues in the F protein in the three-dimensional model to create a protein containing transplanted epitope; and


(d) producing the protein comprising the transplanted epitope. The method can further include the step of modifying the protein of step (c) to stabilize the protein in a conformation that maintains the transplanted epitope in the antibody-bound conformation of the three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, wherein the model substantially represents the atomic coordinates specified in Protein Data Bank accession code 3O41.


The three-dimensional model of a complex between the RSV F peptide and motavizumab, or 101F, disclosed herein provides an understanding of how residues in each molecule interact to form a complex. As disclosed herein, such information is useful in producing immunogens that stimulate a humoral immune response against the RSV F protein. Such information can also be used to produce an antibody (also referred to herein as an antibody protein) that has a higher, or lower, affinity for the RSV F peptide. More specifically, by knowing how the peptide and the antibody align in three-dimensional space, the sequence of the antibody can be altered to introduce new amino acids capable of forming bonds with amino acids in the peptide. Thus, one embodiment of the present invention is a modified RSV neutralizing antibody that is more potent than motavizumab, or 101F; such modified antibody comprises a peptide-binding site for the RSV F peptide, wherein such modified antibody contains amino acid substitutions when compared to the amino acid sequence of motavizumab or 101F, wherein such substitutions result in the formation of new ionic bonds between the modified antibody and the RSV F peptide, and wherein such new ionic bonds result in the modified antibody having a higher affinity for the RSV F protein. In one embodiment, the modified antibody is created by introducing sequence alterations into the amino acid sequence of motavizumab. In another embodiment, the modified antibody is created by introducing sequence alterations into the amino acid sequence of 101F. An antibody protein of the embodiments can be of any size that exhibits more potent neutralization of RSV than does motavizumab or 101F antibody. For example, an antibody protein can comprise an entire heavy chain and an entire light chain or can comprise a portion thereof that retains more potent neutralization activity. In one embodiment an antibody protein is an antigen-binding fragment. In one embodiment, an antibody protein is a single polypeptide chain.


A preferred embodiment of the present invention is a modified neutralizing antibody that exhibits more potent neutralization of RSV than does motavizumab, wherein such modified antibody is produced by:


(a) obtaining a three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, such complex being set forth in the three-dimensional model defined by the atomic coordinates in PDB acc code 3IXT;


(b) identifying at least one amino acid change in the interface between motavizumab and the RSV F protein, wherein such at least one change, if incorporated into motavizumab, would yield an antibody protein with a higher affinity for RSV F protein; and


(c) producing such antibody protein comprising such at least one change. Another embodiment is a modified neutralizing antibody that exhibits more potent neutralization of RSV than does 101F antibody, wherein such modified antibody is produced by:


(a) obtaining a three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, the complex being set forth in the three-dimensional model defined by the atomic coordinates in PDB acc code 3O41;


(b) identifying at least one amino acid change in the interface between 101F antibody and the RSV F protein, wherein such at least one change, if incorporated into 101F antibody, would yield an antibody protein with a higher affinity for RSV F protein; and


(c) producing such antibody protein comprising such at least one change.


As used herein, an antibody protein that exhibits more potent neutralization of RSV than does motavizumab means that a lower titer of such antibody protein is required to neutralize a given amount of RSV, as compared to the titer of motavizumab required to neutralize the same amount of RSV. As used herein, an antibody that exhibits more potent neutralization of RSV than does 101F antibody means that a lower titer of such antibody is required to neutralize a given amount of RSV, as compared to the titer of 101F required to neutralize the same amount of RSV. Suitable amino acid changes to the sequence of motavizumab that result in an antibody protein having a higher affinity for the RSV F protein are disclosed herein. This technique can also be used to modify other antibodies that bind to the motavizumab-binding site or the 101F antibody-binding site of RSV F protein.


One embodiment of the present invention is a modified antibody, wherein the heavy chain of such antibody comprises SEQ ID NO:5, except that such heavy chain comprises at least one amino acid substitution selected from the group consisting of:


(a) the amino acid at position 32 of SEQ ID NO:5 is substituted with a histidine or a glutamic acid;


(b) the amino acid at position 35 of SEQ ID NO:5 is substituted with an alanine;


(c) the amino acid at position 52 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of lysine, histidine, threonine, serine and arginine;


(d) the amino acid at position 53 of SEQ ID NO:5 is substituted with a histidine or a serine;


(e) the amino acid at position 54 of SEQ ID NO:5 is substituted with a phenylalanine or an arginine;


(f) the amino acid at position 56 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of isoleucine, serine, glutamic acid, and aspartic acid;


(g) the amino acid at position 58 of SEQ ID NO:5 is substituted with a tyrosine or a tryptophan;


(h) the amino acid at position 97 of SEQ ID NO:5 is substituted with an aspartic acid, a histidine or an arginine;


(i) the amino acid at position 99 of SEQ ID NO:5 is substituted with aspartic acid;


(j) the amino acid at position 100 of SEQ ID NO:5 is substituted with a tyrosine, a tryptophan or a histidine; and


(k) the amino acid at position 100A of SEQ ID NO:5 is substituted with a serine or a threonine.


Another embodiment of the present invention is a modified antibody, wherein the light chain of such antibody comprises SEQ ID NO:6, except that such light chain comprises at least one amino acid substitution selected from the group consisting of:


(a) the amino acid at position 32 of SEQ ID NO:6 is substituted with a phenylalanine;


(b) the amino acid at position 49 of SEQ ID NO:6 is substituted with an histidine or an arginine;


(c) the amino acid at position 92 of SEQ ID NO:6 is substituted with lysine;


(d) the amino acid at position 94 of SEQ ID NO:6 is substituted with a histidine; and,


(e) the amino acid at position 96 of SEQ ID NO:6 is substituted with a histidine.


In addition to the substitutions described above, analysis of the three-dimensional model of the complex between the RSV F peptide and motavizumab, or 101F, disclosed herein, indicates that additional contacts between the antibody and the peptide can be made by increasing the length of the CDRH2 loop in the antibody (which spans amino acids 50 through 58 of the heavy chain) by 2 residues. One embodiment of the present invention is a modified antibody, wherein the heavy chain of such antibody comprises SEQ ID NO:5, except that amino acids 50 through 58 of SEQ ID NO:5 have been replaced with an 11 amino acid sequence defined as follows:


(a) position one of said 11 amino acid sequence is a glutamic acid, a serine, or a methionine;


(b) position two of said 11 amino acid sequence is an isoleucine;


(c) position three of said 11 amino acid sequence is a histidine, an arginine, or a phenylalanine;


(d) position four of said 11 amino acid sequence is a serine;


(e) position five of said 11 amino acid sequence is a glycine;


(f) position six of said 11 amino acid sequence is an amino acid selected from the group consisting of glycine, histidine, lysine, leucine, asparagine, glutamine, serine, aspartic acid, threonine, and arginine;


(g) position seven of said 11 amino acid sequence is an amino acid selected from the group consisting of phenylalanine, lysine, serine, threonine, aspartic acid, and arginine;


(h) position eight of said 11 amino acid sequence is a glutamic acid, an asparagine, or an aspartic acid;


(i) position nine of said 11 amino acid sequence is an amino acid selected from the group consisting of aspartic acid, histidine, leucine, serine, arginine, and threonine;


(j) position ten of said 11 amino acid sequence is a tyrosine; and


(k) position eleven of said 11 amino acid sequence is a tyrosine, a phenylalanine or a histidine.


It should be understood that any combination of the above-described substitutions can be made. That is, in addition to substituting the eleven amino acid sequence described above, other substitutions can be made outside of amino acids 50-58 of SEQ ID NO:5, (e.g., substitutions into positions 32, 35, 97, 99, 100 or 100A of SEQ ID NO:5, and/or substitutions in to the light chain), so long as the resultant antibody exhibits more potent neutralization of RSV than does motavizumab or 101F.


The disclosure provides a protein comprising an amino acid sequence of any of the antibody proteins of the embodiments. The disclosure also provides a nucleic acid molecule encoding any of the antibody proteins of the embodiments. Such a nucleic acid molecule can encode one or more antibody proteins. The disclosure further provides a recombinant molecule that comprises a nucleic acid molecule encoding an antibody protein of the embodiments operatively linked to at least one transcription control sequence capable of effecting expression of the nucleic acid molecule in a recombinant cell. A recombinant molecule can comprise one or more nucleic acid molecules encoding an antibody protein of the embodiments operatively linked to one or more transcription control sequences. The disclosure also provides a recombinant cell transformed with a recombinant molecule of the embodiments; i.e., a recombinant cell comprises a recombinant molecule. A recombinant cell can comprise one or more recombinant molecules.


The disclosure provides methods to produce antibody proteins of the embodiments. An antibody protein can be produced synthetically, recombinantly, or by a combination of synthetic and recombinant methods. Methods such as those taught herein for production of immunogens can be used. In addition, methods are known to those skilled in the art.


The disclosure provides a method to produce a composition comprising a neutralizing antibody protein that exhibits more potent neutralization of RSV than does motavizumab. The method comprises:


(a) obtaining a three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, the complex being set forth in the 3-dimensional model defined by the atomic coordinates in Protein Data Bank accession code 3IXT;


(b) identifying at least one amino acid change in the interface between motavizumab and the RSV F protein, wherein the at least one change, if incorporated into motavizumab, would yield an antibody protein with a higher affinity for RSV F protein; and


(c) producing the antibody protein comprising the at least one change.


The disclosure also provides a method to produce a composition comprising a neutralizing antibody protein that exhibits more potent neutralization of RSV than does 101F antibody. The method comprises:


(a) obtaining a three-dimensional model of a complex between an RSV F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, the complex being set forth in the 3-dimensional model defined by the atomic coordinates in Protein Data Bank accession code 3O41;


(b) identifying at least one amino acid change in the interface between the 101F antibody and the RSV F protein, wherein the at least one change, if incorporated into the 101F antibody, would yield an antibody protein with a higher affinity for RSV F protein; and


(c) producing the antibody protein comprising the at least one change.


One embodiment of the present invention is an isolated crystal of a complex between an RSV F peptide and motavizumab. Another embodiment of the present invention is an isolated crystal of a complex between an RSV F peptide and 101F. As used herein, an isolated crystal is a crystal of a protein, or complex of proteins, that has been produced in a laboratory; that is, an isolated crystal is produced by an individual and is not an object found in situ in nature. It is appreciated by those skilled in the art that there are a variety of techniques to produce crystals including, but not limited to, vapor diffusion using a hanging or sitting drop methodology, vapor diffusion under oil, and batch methods; see, for example, Ducruix et al., eds., 1991, Crystallization of nucleic acids and proteins; A practical approach, Oxford University Press, and Wyckoff et al., eds., 1985, Methods in Enzymology 11, 49-185; each reference is incorporated by reference herein in its entirety. It is also to be appreciated that crystallization conditions can be adjusted depending on a protein's inherent characteristics as well as on a protein's concentration in a solution and that a variety of precipitants can be added to a protein solution in order to effect crystallization; such precipitants are known to those skilled in the art. In a preferred embodiment, a crystal of a complex between an RSV F peptide and motavizumab or 101F is produced in a solution by adding a precipitant such as polyethylene glycol (PEG) or PEG monomethylether.


One embodiment of the present invention is an isolated crystal of a complex between an RSV F peptide and motavizumab, or 101F, such crystal being produced by the vapor diffusion method using a reservoir solution comprising about 17.5% (w/v) PEG 8000, 0.2 M zinc acetate, and 0.1 M cacodylate pH 6.5. Another embodiment of the present invention is an isolated crystal of a complex between an RSV F peptide and motavizumab, or 101F, wherein obtained by a method comprising:

    • (a) producing an initial crystal using the vapor diffusion method at a temperature of about 20° C., with a reservoir solution comprising about 17.5% (w/v) PEG 8000, 0.2 M zinc acetate, and 0.1 M cacodylate pH 6.5; and
    • (b) streak-seeding the initial crystal obtained in (a) into hanging drops consisting of 1 μl of protein complex and 1 μl of 30% (w/v) PEG 1500.


Isolated crystals of the present invention can include heavy atom derivatives, such as, but not limited to, gold, platinum, mercury, selenium, copper, and lead. Such heavy atoms can be introduced randomly or introduced in a manner based on knowledge of three-dimensional models of the present invention. Additional crystals of the present invention are not derivatized.


A preferred crystal of the present invention diffracts X-rays to a resolution of about 4.5 angstroms or higher (i.e., lower number meaning higher resolution), with resolutions of about 4.0 angstroms or higher, about 3.5 angstroms or higher, about 3.25 angstroms or higher, about 3 angstroms or higher, about 2.5 angstroms or higher, about 2.3 angstroms or higher, about 2 angstroms or higher, about 1.5 angstroms or higher, and about 1 angstrom or higher being increasingly more preferred. It is appreciated, however, that additional crystals of lower resolutions can have utility in discerning overall topology of the structures, e.g., location of a contact residues between an F peptide and its respective antibody. Preferred are crystals are those described in Table 3 and Table 4.









TABLE 3







Data collection and refinement statistics (molecular replacement)









Motavizumab/peptide














Data collection




Space group
P43212



Cell dimensions



a = b, c (Å)
90.75, 232.06



Resolution (Å)
50-2.75



Rmerge
11.3 (52.9)



I/σI
12.2 (1.8) 



Completeness (%)
93.9 (88.3)



Redundancy
4.6 (3.5)



Refinement



Resolution (Å)
2.75



No. reflections
23,502



Rwork/Rfree (%)
21.3/27.4



No. atoms



Protein
6464



Ligand/ion
396



Water
150



B-factors



Protein
57.1



Ligand/ion
89.2



Water
47.0



R.m.s. deviations



Bond lengths (Å)
0.004



Bond angles (°)
0.742







Values in parentheses are for highest-resolution shell.













TABLE 4







Data collection and refinement statistics (molecular replacement)









101F/peptide














Data collection




Space group
P212121



Cell dimensions



a, b, c (Å)
79.90, 92.98, 141.22



Resolution (Å)
50-1.95



Rmerge
10.7 (47.9)



I/σI
15.5 (2.0) 



Completeness (%)
95.7 (77.4)



Redundancy
5.9 (4.3)



Refinement



Resolution (Å)
1.95



No. reflections
69,877



Rwork/Rfree (%)
17.7/22.0



No. atoms



Protein
6695



Ligand/ion
223



Water
727



B-factors



Protein
43.1



Ligand/ion
61.2



Water
50.7



R.m.s. deviations



Bond lengths (Å)
0.006



Bond angles (°)
1.042







Values in parentheses are for highest-resolution shell.






One embodiment of the present invention is a three-dimensional model of a complex between an RSV F peptide consisting of SEQ ID NO:2 and motavizumab, wherein said model is substantially represented by the atomic coordinates specified in PDB acc code 3IXT. Another embodiment of the present invention is a three-dimensional model of a complex between an RSV F peptide consisting of SEQ ID NO:4 and 101F, wherein said model is substantially represented by the atomic coordinates specified in PDB acc code 3O41. Another embodiment of the present invention is a three-dimensional model of a complex between an RSV F peptide consisting of SEQ ID NO:9 and 101F, wherein the model is substantially represented by the atomic coordinates specified in PDB acc code 3O45. As used herein, a model that is substantially represented by atomic coordinates listed herein includes not only those models literally represented by the coordinates but also models representing a coordinate transformation of atomic coordinates disclosed herein, for example, by changing the relative spatial orientation of the coordinates. A three-dimensional model of a complex between an RSV F peptide and motavizumab, or 101F, is a representation, a mathematical model, or image that predicts the actual structure of the corresponding complex. As such, a three-dimensional model is a tool that can be used to probe the relationship between the region's structure and function at the atomic level and to design immunogens and modified. It is well known to those skilled in the art, however, that a three-dimensional model of a protein derived by analysis of protein crystals is not identical to the inherent structure of the protein. See, for example, Branden et al., Introduction to Protein Structure, Garland Publishing Inc., New York and London, 1991, especially on page 277, which states “not surprisingly the model never corresponds precisely to the actual crystal.” Furthermore, the model can be subjected to further refinements to more closely correspond to the actual structure of a complex between an RSV F peptide and motavizumab or 101F. Such a refined model, which is an example of a modification of the present invention, is a better predictor of the actual structure and mechanism of action of the protein that the model represents. Refinements can include models determined to more preferred degrees of resolution, preferably to about 4.5 angstroms, more preferably to about 4 angstroms, more preferably to about 3.5 angstroms, more preferably to about 3.25 angstroms, more preferably to about 3 angstroms, more preferably to about 2.5 angstroms, more preferably to about 2.3 angstroms, more preferably to about 2 angstroms, more preferably to about 1.5 angstroms, and even more preferably to about 1 angstrom. Preferred refinements are obtained using the three-dimensional model as a basis for such improvements.


One embodiment of the present invention is a composition comprising an immunogen or an antibody protein of the present invention. Another embodiment is a composition comprising a nucleic acid molecule, protein, recombinant molecule or recombinant cell of the embodiments. One type of composition is a vaccine. A composition of the present invention can be formulated in an excipient that a patient to be treated can tolerate. Examples of such excipients include water, saline, Ringer's solution, dextrose solution, Hank's solution, and other aqueous physiologically balanced salt solutions. Excipients can also contain minor amounts of additives, such as substances that enhance isotonicity and chemical or biological stability. Examples of buffers include phosphate buffer, bicarbonate buffer, and Tris buffer. Standard formulations can either be liquids or solids that can be taken up in a suitable liquid as a suspension or solution for administration to a patient. In one embodiment, a non-liquid formulation may comprise the excipient salts, buffers, stabilizers, etc., to which sterile water or saline can be added prior to administration.


A composition of the present invention may also include one or more adjuvants or carriers. Adjuvants are typically substances that enhance the immune response of a patient to a specific antigen, and carriers include those compounds that increase the half-life of a composition in the treated patient.


Immunogens and antibodies of the present invention are intended for use in protection against infection by RSV. The immunogens disclosed herein protect against RSV infection by eliciting a humoral immune response against the F protein of RSV. This humoral response results in neutralization of the virus. Antibodies of the present invention protect against infection with RSV by binding and neutralizing the virus. Thus one embodiment of the present invention is a method to protect a patient from RSV infection, the method comprising administering to the patient an immunogen or an antibody produced using the methods disclosed herein. One embodiment is a method to elicit a neutralizing humoral immune response against RSV, the method comprising administering an immunogen of the embodiments, wherein such administration elicits a neutralizing humoral immune response against RSV. In one embodiment, the immunogen is administered to a patient. One embodiment is a method to protect a patient from RSV infection comprising administering to the patient an immunogen, wherein such administration protects the patient from RSV infection. One embodiment is a method to elicit a neutralizing humoral immune response against RSV, the method comprising administering a nucleic acid vaccine of the embodiments, wherein such administration elicits a neutralizing humoral immune response against RSV. In one embodiment, the nucleic acid vaccine is administered to a patient. One embodiment is a method to protect a patient from RSV infection comprising administering to the patient a nucleic acid vaccine, wherein such administration protects the patient from RSV infection. One embodiment is a method to protect a patient from RSV infection comprising administering to the patient an antibody protein, wherein such administration protects the patient from RSV infection.


As used herein the phrase protect a patient from RSV infection includes preventing a patient from being infected by RSV, as well as treating a patient already infected with RSV. As used herein the term patient refers to any animal in need of such prevention or treatment. The animal can be a human or a non-human animal. A preferred animal to treat is a mammal. A patient can be of any age. In one embodiment, an immunogen or antibody can be administered to an infant. In one embodiment, an immunogen or antibody can be administered to a patient that is older than an infant. An immunogen or antibody can be administered or applied per se, or as a composition. An immunogen or antibody of the present invention, or a composition thereof, can be administered to a patient by a variety of routes, including, but limited to, by injection (e.g., intravenous, intramuscular, subcutaneous, intrathecal, intraperitoneal), by inhalation, by oral (e.g., in a pill, tablet, capsule, powder, syrup, solution, suspension, thin film, dispersion or emulsion.), transdermal, transmucosal, pulmonary, buccal, intranasal, sublingual, intracerebral, intravaginal rectal or topical administration or by any other convenient method known to those of skill in the art.


The amount of an immunogen or antibody of the present invention, and/or a composition thereof that will be effective can be determined by standard clinical techniques known in the art. Such an amount is dependent on, among other factors, the patient being treated, including, but not limited to the weight, age, and condition of the patient, the intended effect of the composition, the manner of administration and the judgment of the prescribing physician.


An immunogen or antibody of the present invention, or a composition thereof, can be administered alone or in combination with one or more other pharmaceutical agents, including other immunogens or antibodies of the present invention. The specific composition depends on the desired mode of administration, as is well known to the skilled artisan. One composition can include an immunogen of the present invention comprising motavizumab-binding contact residues. Another composition can include an immunogen of the present invention comprising 101F-binding contact residues. One composition comprises a combination of both immunogens. Another composition is an antibody of the present invention. Yet another composition comprises a nucleic acid vaccine comprising at least one nucleic acid molecule encoding an immunogen of the present invention. The disclosure also provides for a combination comprising one or more immunogens and/or antibodies (i.e., antibody proteins) of the embodiments with one or more other RSV immunogens and/or antibodies. The disclosure also provides for a combination comprising one or more immunogens and/or antibodies (i.e., antibody proteins) of the embodiments with one or more protective agents, such as, but not limited to, an agent that protects from infection by a virus, bacterium, parasite, or other infectious agents.


In one embodiment, administration can comprise a prime followed by one or more boosts. A prime can comprise a composition comprising at least one of the immunogens disclosed herein, or a nucleic acid encoding such an immunogen. A boost can comprise at least one of the immunogen disclosed herein, or a nucleic acid encoding such an immunogen. In one embodiment the boost comprises an immunogen that has been resurfaced (compared to the first immunogen) to further boost the humoral immune response against RSV contact residues in the motavizumab or 101F binding domains. In one embodiment the boost comprises a multivalent immunogen.


EXAMPLES

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the embodiments, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, and temperature is in degrees Celsius. Standard abbreviations may be used. For example, amino acids can be denoted by either the standard 3-letter or 1-letter code.


Example 1
Three-Dimensional Structure of Rsv F Protein and Motavizumab

This Example describes the crystallization and determination of the 3-dimensional structure of a complex between motavizumab and the 24-residue RSV fusion (F) peptide spanning amino acids 254-277 of the F protein (i.e., NSELLSLIND MPITNDQKKL MSNN, also denoted herein as SEQ ID NO:2) that includes the binding domain of motavizumab. The amino acid sequence of the F protein used in these studies is as follows: MELLILKANA ITTILTAVTF CFASGQNITE EFYQSTCSAV SKGYLSALRT GWYTSVITIE LSNIKENKCN GTDAKVKLIK QELDKYKNAV TELQLLMQST PATNNRARRE LPRFMNYTLN NAKKTNVTLS KKRKRRFLGF LLGVGSAIAS GVAVSKVLHL EGEVNKIKSA LLSTNKAVVS LSNGVSVLTS KVLDLKNYID KQLLPIVNKQ SCSISNIETV IEFQQKNNRL LEITREFSVN AGVTTPVSTY MLTNSELLSL INDMPITNDQ KKLMSNNVQI VRQQSYSIMS IIKEEVLAYV VQLPLYGVID TPCWKLHTSP LCTTNTKEGS NICLTRTDRG WYCDNAGSVS FFPQAETCKV QSNRVFCDTM NSLTLPSEVN LCNVDIFNPK YDCKIMTSKT DVSSSVITSL GAIVSCYGKT KCTASNKNRG IIKTFSNGCD YVSNKGVDTV SVGNTLYYVN KQEGKSLYVK GEPIINFYDP LVFPSDEFDA SISQVNEKIN QSLAFIRKSD ELL, also denoted herein as SEQ ID NO:1.


To enhance the potency of palivizumab, each residue in the six complementarity-determining regions (CDRs) was individually substituted with the other 19 amino acids (a total number of 1,121 unique single variants were assayed), and combinations of beneficial substitutions assessed (Wu H et al., 2007, J. Mol. Biol. 368, 652-665; Wu H et al., 2005, J. Mol. Biol. 350, 126-144). This led to the development of a second-generation antibody, motavizumab, which is ˜10 times more potent than palivizumab (Wu H et al., 2007, ibid.). Only 13 amino acids differ between motavizumab and palivizumab. Of these, seven individually increase the affinity of the antibody to the F glycoprotein, resulting in a 0.035 nM Kd (versus 1.4 nM for palivizumab) (Johnson S et al., 1997, J. Infect. Dis. 176, 1215-1224; Wu H et al., 2007, ibid.; Wu H et al., 2005, ibid.). This disclosure includes a characterization of the structural basis of motavizumab affinity, model mutations with enhanced affinity, and structural implications for motavizumab binding in the trimeric F glycoprotein context.


Motavizumab is ˜10-fold more potent than its predecessor, palivizumab (SYNAGIS®), the FDA-approved monoclonal antibody used to prevent respiratory syncytial virus (RSV) infection. The structure of motavizumab in complex with a 24-residue peptide corresponding to its epitope on the RSV-fusion (F) glycoprotein reveals the structural basis for its increased potency. Modeling suggests that motavizumab recognizes a different quaternary configuration of the F glycoprotein than observed in a homologous structure.


Recombinant motavizumab IgG molecules that were shown to neutralize RSV potently (FIG. 1a) were used to create antigen-binding fragments (Fabs) for crystallographic analysis. Crystals of the Fab were obtained in complex with a 24-residue peptide, which corresponds to residues 254-277 of the RSV F glycoprotein A2 strain (NSELLSLIND MPITNDQKKL MSNN) and represents the known epitope for palivizumab/motavizumab (Arbiza J et al., 1992, J. Gen. Virol. 74, 2225-2234). The crystals diffracted X-rays to 2.75 Å, and a molecular replacement solution was obtained containing two molecules of the previously-determined unliganded palivizumab structure (Johnson L S et al., U.S. Pat. No. 7,229,618, issued Jun. 12, 2007) per asymmetric unit. Initial maps showed two regions of well-defined helical density near the CDRs of each Fab. These regions were modeled as the peptide, and the structure refined to an Rcrys/Rfree=21.3/27.4%; see Table 3.


The peptide forms a helix-loop-helix (FIG. 1b), in agreement with secondary structure predictions of the RSV F glycoprotein (Smith B J et al., 2002, Protein Eng. 15, 365-371). The main-chain electron density for the peptide was good for all residues and the side chain density was good for residues 262-276, but weak or non-existent for residues N- and C-terminal to this region (FIG. 3). The variable domains of the peptide-bound motavizumab structure and the unbound palivizumab structure are similar (rmsd 1.8 Å for variable domain Ca), with the largest differences occurring in the three heavy chain CDRs.


To understand the structural basis for the high affinity interaction between motavizumab and the RSV F protein, the structure of the peptide/Fab complex was analyzed. The interface between the peptide and Fab buries a total of 1,304 Å2 of surface area (680 Å2 on the peptide and 624 Å2 on the Fab, as calculated by PISA, Krissinel E et al., 2007, J. Mol. Biol. 372, 774-797) and has a shape complementarity (Sc) value of 0.76, which is substantially higher than the typical range of 0.64-0.68 for antibody/antigen complexes (Lawrence M C et al., 1993, J. Mol. Biol. 234, 946-950). The electrostatic potentials on the surface of the peptide and Fab are also complementary, with several acidic patches on the Fab interacting with positively charged regions on the peptide (FIG. 4). Approximately 73% of the surface area buried on the Fab is located on the heavy chain, which possesses a large hydrophobic region consisting of residues from the second and third CDRs (FIG. 1c). This region contacts peptide residues located along the length of both helices. The four peptide residues between the two helices do not contribute significantly to motavizumab binding to RSV F, having only 8 Å2 buried at the interface. Interactions between the peptide and heavy chain include hydrogen bonds formed between the peptide side chain of Asn262 and the Fab side chains of Asp54 and Lys56, as well as a hydrogen bond between the peptide side chain of Ser275 and the carbonyl oxygen of Fab residue Ile97. There are also several interactions between the peptide and light chain. These include a hydrogen bond between the side chain of peptide residue Asn268 and the carbonyl oxygen of Gly90, as well as a salt-bridge between the peptide side chain of Lys272 and the side chain of Asp49 in the second CDR (FIG. 1c).


The interactions between the peptide and motavizumab Fab are consistent with RSV F glycoprotein mutations known to disrupt antibody binding to this epitope. It has been demonstrated that mutations N262Y, N268I and K272E decrease the binding of several antibodies that recognize this region of the F glycoprotein (Arbiza J et al., ibid.). The mutations K272M and K272Q have also been found in RSV F glycoprotein escape mutants that are resistant to palivizumab (Zhao X et al., 2004, J. Infect. Dis. 190, 1941-1946). The side chains of these three peptide residues all form hydrogen bonds or salt bridges with residues in the Fab (FIG. 1c), interactions that would be lost by the mutations listed above.


To investigate the structural basis for motavizumab's enhanced potency over palivizumab, the positions of the 7 altered residues that increase the affinity to the F glycoprotein were analyzed in the peptide-bound crystal structure (FIG. 1d). Three of the seven altered residues (S32A, T98F and W100F in the heavy chain) directly contact the peptide and are located in the large hydrophobic patch described earlier. Both the S32A and T98F substitutions increase the hydrophobicity of this patch, favoring interactions with the peptide. As for the W100F mutation, the smaller Phe side chain is able to pack tightly against peptide residues N268 and K272. The larger Trp side chain found in palivizumab would likely alter the conformation of these residues, which make hydrogen bond and salt bridge interactions with the Fab, respectively. When the three palivizumab residues were modeled into the complex, the Sc value decreased from 0.76 to 0.70, reflecting the poorer fit between the peptide and Fab.


The other four substitutions that increase the potency of motavizumab do not contact the peptide directly. Two of the mutations (D58H and S95D in the heavy chain) are located near the interface with the peptide, and their side chains interact with other residues in the CDRs. Thus, they likely exert their effects indirectly by altering the position of other amino acids that do contact the peptide. The side chains of the two remaining substitutions, S65D in the heavy chain and S29R in the light chain, have weak electron density and do not contact any residues in the peptide or Fab. However, both substitutions increase the on-rate of motavizumab for the F glycoprotein, and the S29R mutation alone results in a 4.4-fold increase in RSV neutralization in vitro9. Collectively, these data suggest that the S65D and S29R side chains either bind to residues in the F glycoprotein located outside the primary epitope or increase favorable long-range electrostatic interactions. Relevant to this, motavizumab binds to the peptide 6.000-fold weaker than the full-length F protein (230 nM vs 0.035 nM) (Wu H et al., 2007, ibid.; Tous G I et al., 2006, U.S. patent application Ser. No. 11/230,593), though some fraction of the decrease in peptide affinity is likely due to the peptide not adopting the helix-loop-helix conformation in solution (Lopez J A et al., 1993, J. Gen. Virol. 74, 2567-2577).


An earlier version of motavizumab contained residues Phe52, Phe53, and Asp55 in the light chain CDR2, which increased in vitro RSV neutralization ˜2-fold (Wu H et al., 2007, ibid.). However, these residues also increased non-specific tissue binding and decreased the in vivo potency (Wu H et al., 2007, ibid.), perhaps due to the two solvent-exposed Phe residues (FIG. 1d). Thus, they were ultimately returned to the amino acids found in palivizumab (Ser52, Lys53, and Ala55).


To visualize the binding of motavizumab to the full-length F glycoprotein, a model was generated based on the pre-fusion parainfluenza virus 5 (PIV5) structure (Yin H S et al., 2006, Nature 439, 38-44) (12.4% sequence identity to RSV F (Smith B J et al., ibid.)). A sequence alignment (FIG. 5a) identified a similar helix-loop-helix, and structural analysis provided a precise alignment (FIG. 5b), shifting the PIV5 sequence by three amino acids to provide a motavizumab epitope/PIV5 superposition of 2.1 Å rmsd for all 24 peptide Ca atoms (FIG. 2a). A model was generated by orienting the Fab via superposition of the bound peptide onto the corresponding epitope in the PIV5 F glycoprotein structure. The resulting model shows no clashes between the Fab and F glycoprotein monomer to which it is bound (FIG. 2b).


In the pre-fusion trimeric context, however, both the heavy and light chains of the Fab clash with an adjacent RSV F monomer that packs against the same face of the helix-loop-helix that motavizumab binds (FIG. 2c,d). The location of the epitope at a subunit interface may explain why this neutralizing epitope is so highly conserved in RSV strains. Since motavizumab neutralizes RSV by preventing the fusion of the viral and cellular membranes (Johnson S et al., 1997, ibid.), motavizumab must bind to the F glycoprotein before or during the transition to the post-fusion state. The extensive clashes in the trimer model, however, suggest that motavizumab would be unable to bind the pre-fusion trimeric F glycoprotein as it exists in the PIV5 structure. To address this issue experimentally, a soluble RSV F glycoprotein was expressed and purified in a form similar to the PIV5 F glycoprotein used in the modeling. Specifically, the known furin cleavage sites were mutated and a fibritin trimerization motif (Tao Y et al., 1997, Structure 5, 789-798) was appended to the truncated C terminus to keep the protein in a trimeric, pre-fusion conformation. This stabilized RSV F glycoprotein, referred to as RSV F0 Fd, eluted from a gel filtration column with a retention volume consistent with that of a glycosylated trimer (FIG. 2e). To determine whether motavizumab or palivizumab is able to bind the pre-fusion trimeric RSV F glycoprotein, palivizumab Fab was added in excess to a solution of RSV F0 Fd, and the mixture was passed over a gel filtration column. The elution profile contained two peaks, corresponding to excess Fab and a complex of the Fab and F glycoprotein (FIG. 2f). The elution volume of the complex peak was consistent with a trimeric F glycoprotein bound by three Fabs, in agreement with the ratio (1:2.97) of F glycoprotein and Fab bands observed on a Coomassie stained SDS-PAGE gel containing fractions from the complex peak (FIG. 2f).


Collectively, these data suggest that motavizumab binds to or induces a conformation of the trimeric F glycoprotein that is different from that observed in the PIV5 F pre-fusion structure. One possibility is that the structure of the RSV F glycoprotein differs significantly from that of PIV5, although the predicted RSV F glycoprotein secondary structure appears similar to that observed in the PIV5 F pre-fusion crystal structure (FIG. 5). Another possibility is that motavizumab traps an intermediate between pre- and post-fusion forms. It has been suggested that during this transition, which is one of the largest structural rearrangements known, the F glycoprotein monomers transiently dissociate prior to forming the trimeric post-fusion conformation (Yin H S et al., ibid). It is to be noted in this regard that glutaraldehyde crosslinking of the soluble F glycoprotein trimer does not inhibit motavizumab binding (FIG. 6). Alternatively, the RSV F glycoprotein in its pre-fusion conformation may contain sufficient flexibility to bind three motavizumab Fabs. Modeling studies indicate that a ˜30° rotation of domain III parallel to the 3-fold axis would allow clash-free binding of three Fabs. A similar degree of rotation has been observed in cryo-EM tomograms after the binding of neutralizing antibodies to Dengue virus (Lok S-M, et al., 2008, Nat Struct Mol Biol 15, 312-317) and HIV-1 (Liu J et al., 2008, Nature 455, 109-113). Such flexibility may be a more common feature of viral fusion proteins than previously thought.


Materials and Methods

a. Cloning, expression and purification of motavizumab IgG. Two DNA fragments encoding the variable heavy and light chains of motavizumab (Wu H et al., 2007, J. Mol. Biol. 368, 652-665) with appropriate signal sequences were synthesized by GeneArt (Regensburg, Germany) and cloned in-frame into mammalian expression vectors containing human IgG1 heavy and light constant domains, respectively. The amino acid sequence of the variable heavy chain of motavizumab is as follows: QVTLRESGPA LVKPTQTLTL TCTFSGFSLS TAGMSVGWIR QPPGKALEWL ADIWWDDKKH YNPSLKDRLT ISKDTSKNQV VLKVTNMDPA DTATYYCARD MIFNFYFDVW GQGTTVTVSS, also denoted herein as SEQ ID NO:5. The amino acid sequence of the variable light chain of motavizumab is as follows: DIQMTQSPST LSASVGDRVT ITCSASSRVG YMHWYQQKPG KAPKLLIYDT SKLASGVPSR FSGSGSGTEF TLTISSLQPD DFATYYCFQG SGYPFTFGGG TKVEIK, also denoted herein as SEQ ID NO:6.


Both vectors were co-transfected at a 1:1 ratio into HEK293F cells (Invitrogen, Life Technologies, Carlsbad, Calif.) in serum-free 293Freestyle medium (Invitrogen). After 3 hours, valproic acid (Sigma, St. Louis, Mo.) was added to 4 mM final concentration. Expression lasted for four days at 37° C. with 10% CO2 and shaking at 125 rpm in disposable flasks. The supernatant was collected, filtered, and passed over 5 ml of Protein A agarose resin (Pierce). After washing with several column volumes of phosphate-buffered saline (PBS), the resin was eluted with 25 ml of IgG Elution Buffer (Pierce, Thermo Fisher Scientific, Rockford, Ill.) and immediately neutralized with 1 M Tris pH 8.0. The eluted protein was dialyzed against PBS and stored at 4° C.


b. Measurement of antibody-mediated neutralization. RSV expressing green fluorescent protein (GFP) was provided by Mark Peeples and Peter Collins and constructed as previously reported (Hallak L K, et al., 2000, Virology 271, 264-275). Antibody-mediated neutralization was measured using HEp-2 cells. GFP-RSV was added to serial four-fold dilutions of serum and/or antibody in 96-well plates and incubated at 37° C. for one hour. Serum concentrations ranged from 1:10 to 1:40,960. After one hour, 100 μl of virus/serum mixture was added to 5×104 cells/100 μl per well in 96-well plates. Infection was monitored as a function of GFP expression (encoded by the viral genome) at 18 hours post-infection by flow cytometry (LSR II, BD Bioscience, CA, USA). Prior to assessment by flow cytometry, cells were treated with trypsin to ensure a single-cell suspension optimal for analysis and fixed with 0.5% paraformaldehyde, Data were analyzed by curve fitting and non-linear regression (GraphPad Prism, GraphPad Software Inc., San Diego Calif.) in order to demonstrate the percent neutralization at a given antibody concentration, and the neutralization activity was compared based on the EC50.


c. Digestion and purification of motavizumab Fab fragments. The purified motavizumab IgG protein was reduced with 100 mM dithiothreitol at 37° C. for 1 hour and then alkylated with 2 mM iodoacetamide for 48 hours at 4° C. 10 ml of reduced and alkylated IgG in PBS at 3.5 mg/ml was combined with 15 μg of endoproteinase Lys-C (Roche) and incubated at 37° C. for 6 hours. The reaction was quenched by the addition of TLCK and leupeptin to 50 μg/ml and 2 μg/ml, respectively. To remove the Fc fragments from the Fab fragments, the quenched reaction was passed over 5 ml of Protein A agarose. The Fab-containing flow through was further purified over an S200 gel filtration column and concentrated aliquots were stored frozen at −80° C.


d. Protein crystallization and data collection. A peptide with the sequence NSELLSLIND MPITNDQKKL MSNN, corresponding to residues 254-277 of the RSV F protein and also denoted herein as SEQ ID NO:2) was synthesized by American Peptide (Sunnyvale, Calif.) with an acetylated N-terminus and an amidated C-terminus. A five-fold molar excess of peptide was incubated with motavizumab Fab at 22° C. for 1.5 hours and then concentrated to give a 13.1 mg/ml solution of Fab/peptide complex in 2 mM Tris pH 7.5, 150 mM NaCl. Initial crystals were grown by the vapor diffusion method in sitting drops at 20° C. by mixing 0.1 μl of protein complex with 0.1 μl of reservoir solution (17.5% (w/v) PEG 8000, 0.2 M zinc acetate, 0.1 M cacodylate pH 6.5) using a Cartesian Honeybee crystallization robot (Genomic Solutions). These initial crystals were streak-seeded into hanging drops consisting of 1 μl protein complex and 1 μl 30% (w/v) PEG 1500. After several days rectangular crystals appeared in a single drop with dimensions 40×40×10 μm. These crystals were flash frozen in liquid nitrogen in 40% (w/v) PEG 1500, 30% (v/v) ethylene glycol and loaded into a cryopuck. Data were collected at a wavelength of 0.82656 Å at the SER-CAT beamline ID-22 using the robot automounter (Advanced Photon Source, Argonne National Laboratory).


e. Structure determination, model building and refinement. Diffraction data were processed with the HKL2000 suite (Otwinowski Z et al., 1997, Methods Enzymol. 276, 307-326, Academic Press) and a molecular replacement solution was found by PHASER (McCoy A J et al, 2007, J. Appl. Crystallog. 40, 658-674) using the palivizumab Fab structure (PDB ID: 2hwz) as a search model. Two Fab molecules were placed in the asymmetric unit. After rigid body and TLS refinement using PHENIX (Adams, P D et al., 2002, Acta Crystallogr., Section D 58, 1948-1954), helical peptide density was evident near the CDRs of each Fab. Model building was carried out using COOT (Emsley, P et al., 2004, Acta Crystallogr., Section D 60, 2126-2132) and refinement was performed with PHENIX using NCS restraints. Final data collection and refinement statistics are presented in Table 3. The atomic coordinates for the motavizumab/peptide complex have been deposited in the Protein Data Bank under PDB accession code 3IXT. The atomic coordinates for the peptide portion of the complex are indicated below in Table 6.









TABLE 6





Atomic coordinates for the motavizumab binding peptide



























ATOM
3243
N
ASN
P
254
7.154
8.372
−9.660
1.00
124.11

N


ANISOU
3243
N
ASN
P
254
13767
16195
17192
−3482
2530
2430
N


ATOM
3244
CA
ASN
P
254
7.612
9.736
−9.412
1.00
121.53

C


ANISOU
3244
CA
ASN
P
254
13313
15765
17097
−3858
2355
2424
C


ATOM
3245
C
ASN
P
254
6.768
10.583
−8.462
1.00
111.67

C


ANISOU
3245
C
ASN
P
254
12385
14386
15658
−4047
1969
2168
C


ATOM
3246
O
ASN
P
254
5.706
11.098
−8.827
1.00
107.07

O


ANISOU
3246
O
ASN
P
254
12235
13626
14819
−4027
2035
2025
O


ATOM
3247
CB
ASN
P
254
7.853
10.480
−10.730
1.00
123.56

C


ANISOU
3247
CB
ASN
P
254
13592
15864
17490
−3953
2745
2570
C


ATOM
3248
CG
ASN
P
254
9.265
10.333
−11.225
1.00
130.78

C


ANISOU
3248
CG
ASN
P
254
13976
16914
18799
−4017
2965
2837
C


ATOM
3249
OD1
ASN
P
254
10.213
10.485
−10.461
1.00
132.18

O


ANISOU
3249
OD1
ASN
P
254
13727
17233
19260
−4212
2717
2883
O


ATOM
3250
ND2
ASN
P
254
9.416
10.009
−12.511
1.00
136.19

N


ANISOU
3250
ND2
ASN
P
254
14673
17575
19498
−3837
3434
3010
N


ATOM
3251
N
SER
P
255
7.270
10.702
−7.237
1.00
104.70

N


ANISOU
3251
N
SER
P
255
11278
13608
14896
−4206
1562
2109
N


ATOM
3252
CA
SER
P
255
6.624
11.477
−6.192
1.00
96.74

C


ANISOU
3252
CA
SER
P
255
10531
12499
13728
−4386
1160
1872
C


ATOM
3253
C
SER
P
255
6.465
12.942
−6.584
1.00
85.08

C


ANISOU
3253
C
SER
P
255
9239
10769
12318
−4677
1173
1822
C


ATOM
3254
O
SER
P
255
5.469
13.589
−6.245
1.00
75.10

O


ANISOU
3254
O
SER
P
255
8390
9349
10797
−4716
1012
1614
O


ATOM
3255
CB
SER
P
255
7.430
11.374
−4.900
1.00
100.06

C


ANISOU
3255
CB
SER
P
255
10618
13086
14314
−4514
733
1850
C


ATOM
3256
OG
SER
P
255
8.819
11.450
−5.170
1.00
101.92

O


ANISOU
3256
OG
SER
P
255
10318
13442
14965
−4658
796
2057
O


ATOM
3257
N
GLU
P
256
7.456
13.465
−7.294
1.00
90.87

N


ANISOU
3257
N
GLU
P
256
9673
11460
13395
−4878
1375
2016
N


ATOM
3258
CA
GLU
P
256
7.446
14.869
−7.683
1.00
100.04

C


ANISOU
3258
CA
GLU
P
256
11006
12355
14651
−5184
1410
1998
C


ATOM
3259
C
GLU
P
256
6.561
15.110
−8.898
1.00
95.96

C


ANISOU
3259
C
GLU
P
256
10919
11641
13901
−5014
1792
2007
C


ATOM
3260
O
GLU
P
256
5.860
16.117
−8.970
1.00
93.69

O


ANISOU
3260
O
GLU
P
256
11032
11111
13454
−5117
1734
1875
O


ATOM
3261
CB
GLU
P
256
8.867
15.360
−7.952
1.00
117.93

C


ANISOU
3261
CB
GLU
P
256
12779
14646
17381
−5502
1494
2201
C


ATOM
3262
CG
GLU
P
256
9.623
14.514
−8.951
1.00
137.85

C


ANISOU
3262
CG
GLU
P
256
14946
17336
20096
−5326
1918
2463
C


ATOM
3263
CD
GLU
P
256
10.877
15.200
−9.446
1.00
155.42

C


ANISOU
3263
CD
GLU
P
256
16748
19544
22759
−5665
2092
2662
C


ATOM
3264
OE1
GLU
P
256
11.414
16.058
−8.713
1.00
159.74

O


ANISOU
3264
OE1
GLU
P
256
17121
20045
23527
−6047
1784
2594
O


ATOM
3265
OE2
GLU
P
256
11.323
14.883
−10.568
1.00
161.28

O


ANISOU
3265
OE2
GLU
P
256
17337
20320
23624
−5559
2543
2879
O


ATOM
3266
N
LEU
P
257
6.597
14.186
−9.853
1.00
101.08

N


ANISOU
3266
N
LEU
P
257
11497
12391
14515
−4733
2173
2157
N


ATOM
3267
CA
LEU
P
257
5.751
14.295
−11.035
1.00
105.52

C


ANISOU
3267
CA
LEU
P
257
12464
12797
14833
−4526
2529
2159
C


ATOM
3268
C
LEU
P
257
4.279
14.282
−10.639
1.00
92.20

C


ANISOU
3268
C
LEU
P
257
11263
11055
12715
−4343
2347
1883
C


ATOM
3269
O
LEU
P
257
3.455
14.971
−11.242
1.00
80.48

O


ANISOU
3269
O
LEU
P
257
10189
9377
11014
−4288
2459
1793
O


ATOM
3270
CB
LEU
P
257
6.036
13.160
−12.018
1.00
115.65

C


ANISOU
3270
CB
LEU
P
257
13585
14225
16131
−4230
2932
2345
C


ATOM
3271
CG
LEU
P
257
7.431
13.112
−12.643
1.00
125.63

C


ANISOU
3271
CG
LEU
P
257
14378
15562
17795
−4348
3209
2635
C


ATOM
3272
CD1
LEU
P
257
7.455
12.109
−13.788
1.00
125.83

C


ANISOU
3272
CD1
LEU
P
257
14393
15674
17744
−4003
3651
2792
C


ATOM
3273
CD2
LEU
P
257
7.861
14.487
−13.125
1.00
131.86

C


ANISOU
3273
CD2
LEU
P
257
15206
16112
18783
−4685
3323
2722
C


ATOM
3274
N
LEU
P
258
3.961
13.490
−9.619
1.00
87.87

N


ANISOU
3274
N
LEU
P
258
10664
10685
12036
−4239
2072
1749
N


ATOM
3275
CA
LEU
P
258
2.600
13.395
−9.107
1.00
83.75

C


ANISOU
3275
CA
LEU
P
258
10548
10153
11119
−4083
1890
1479
C


ATOM
3276
C
LEU
P
258
2.187
14.657
−8.352
1.00
89.10

C


ANISOU
3276
C
LEU
P
258
11470
10660
11724
−4309
1559
1292
C


ATOM
3277
O
LEU
P
258
1.104
15.199
−8.579
1.00
87.97

O


ANISOU
3277
O
LEU
P
258
11742
10389
11292
−4214
1564
1118
O


ATOM
3278
CB
LEU
P
258
2.460
12.175
−8.196
1.00
74.89

C


ANISOU
3278
CB
LEU
P
258
9305
9260
9891
−3929
1714
1410
C


ATOM
3279
CG
LEU
P
258
2.444
10.811
−8.889
1.00
75.87

C


ANISOU
3279
CG
LEU
P
258
9350
9528
9951
−3633
2030
1520
C


ATOM
3280
CD1
LEU
P
258
2.583
9.674
−7.882
1.00
52.50

C


ANISOU
3280
CD1
LEU
P
258
6237
6763
6946
−3528
1837
1495
C


ATOM
3281
CD2
LEU
P
258
1.177
10.648
−9.714
1.00
71.13

C


ANISOU
3281
CD2
LEU
P
258
9153
8864
9008
−3407
2256
1381
C


ATOM
3282
N
SER
P
259
3.050
15.118
−7.452
1.00
84.33

N


ANISOU
3282
N
SER
P
259
10608
10060
11373
−4593
1263
1317
N


ATOM
3283
CA
SER
P
259
2.753
16.304
−6.662
1.00
78.21

C


ANISOU
3283
CA
SER
P
259
10063
9112
10542
−4822
925
1137
C


ATOM
3284
C
SER
P
259
2.572
17.520
−7.562
1.00
81.91

C


ANISOU
3284
C
SER
P
259
10820
9288
11015
−4936
1112
1161
C


ATOM
3285
O
SER
P
259
1.932
18.496
−7.175
1.00
92.12

O


ANISOU
3285
O
SER
P
259
12470
10393
12137
−5016
910
979
O


ATOM
3286
CB
SER
P
259
3.845
16.550
−5.623
1.00
86.96

C


ANISOU
3286
CB
SER
P
259
10806
10283
11954
−5123
583
1172
C


ATOM
3287
OG
SER
P
259
5.128
16.495
−6.216
1.00
111.78

O


ANISOU
3287
OG
SER
P
259
13511
13467
15494
−5284
782
1427
O


ATOM
3288
N
LEU
P
260
3.135
17.452
−8.765
1.00
80.16

N


ANISOU
3288
N
LEU
P
260
10467
9019
10971
−4923
1507
1389
N


ATOM
3289
CA
LEU
P
260
2.966
18.509
−9.754
1.00
81.64

C


ANISOU
3289
CA
LEU
P
260
10963
8919
11138
−4989
1747
1442
C


ATOM
3290
C
LEU
P
260
1.590
18.417
−10.392
1.00
82.16

C


ANISOU
3290
C
LEU
P
260
11501
8937
10780
−4636
1897
1295
C


ATOM
3291
O
LEU
P
260
0.872
19.411
−10.499
1.00
82.48

O


ANISOU
3291
O
LEU
P
260
11967
8754
10617
−4633
1836
1160
O


ATOM
3292
CB
LEU
P
260
4.035
18.407
−10.840
1.00
88.21

C


ANISOU
3292
CB
LEU
P
260
11497
9732
12286
−5076
2148
1745
C


ATOM
3293
CG
LEU
P
260
5.418
18.965
−10.516
1.00
96.65

C


ANISOU
3293
CG
LEU
P
260
12153
10763
13805
−5501
2072
1901
C


ATOM
3294
CD1
LEU
P
260
6.324
18.821
−11.725
1.00
101.88

C


ANISOU
3294
CD1
LEU
P
260
12556
11421
14733
−5537
2540
2197
C


ATOM
3295
CD2
LEU
P
260
5.316
20.420
−10.084
1.00
95.37

C


ANISOU
3295
CD2
LEU
P
260
12284
10293
13658
−5824
1844
1782
C


ATOM
3296
N
ILE
P
261
1.234
17.212
−10.823
1.00
88.03

N


ANISOU
3296
N
ILE
P
261
12167
9892
11387
−4332
2090
1316
N


ATOM
3297
CA
ILE
P
261
−0.060
16.970
−11.445
1.00
90.12

C


ANISOU
3297
CA
ILE
P
261
12819
10164
11258
−3990
2230
1161
C


ATOM
3298
C
ILE
P
261
−1.191
17.335
−10.489
1.00
88.77

C


ANISOU
3298
C
ILE
P
261
12956
9997
10776
−3935
1882
852
C


ATOM
3299
O
ILE
P
261
−2.217
17.877
−10.901
1.00
91.98

O


ANISOU
3299
O
ILE
P
261
13765
10301
10884
−3760
1914
694
O


ATOM
3300
CB
ILE
P
261
−0.206
15.502
−11.893
1.00
84.69

C


ANISOU
3300
CB
ILE
P
261
11965
9720
10492
−3712
2451
1212
C


ATOM
3301
CG1
ILE
P
261
0.855
15.159
−12.941
1.00
83.55

C


ANISOU
3301
CG1
ILE
P
261
11551
9571
10622
−3717
2830
1515
C


ATOM
3302
CG2
ILE
P
261
−1.598
15.246
−12.447
1.00
77.49

C


ANISOU
3302
CG2
ILE
P
261
11434
8840
9169
−3386
2552
1011
C


ATOM
3303
CD1
ILE
P
261
0.798
13.729
−13.426
1.00
52.78

C


ANISOU
3303
CD1
ILE
P
261
7511
5886
6658
−3439
3060
1577
C


ATOM
3304
N
ASN
P
262
−0.990
17.044
−9.209
1.00
83.99

N


ANISOU
3304
N
ASN
P
262
12161
9519
10233
−4069
1551
766
N


ATOM
3305
CA
ASN
P
262
−1.986
17.356
−8.193
1.00
82.83

C


ANISOU
3305
CA
ASN
P
262
12280
9392
9801
−4027
1218
479
C


ATOM
3306
C
ASN
P
262
−2.127
18.861
−7.981
1.00
87.67

C


ANISOU
3306
C
ASN
P
262
13199
9728
10385
−4198
1038
385
C


ATOM
3307
O
ASN
P
262
−3.234
19.372
−7.812
1.00
83.35

O


ANISOU
3307
O
ASN
P
262
13029
9126
9514
−4047
926
157
O


ATOM
3308
CB
ASN
P
262
−1.644
16.661
−6.874
1.00
78.11

C


ANISOU
3308
CB
ASN
P
262
11416
8988
9275
−4124
919
432
C


ATOM
3309
CG
ASN
P
262
−2.826
16.596
−5.930
1.00
77.84

C


ANISOU
3309
CG
ASN
P
262
11645
9043
8886
−3998
662
140
C


ATOM
3310
OD1
ASN
P
262
−3.953
16.917
−6.307
1.00
76.30

O


ANISOU
3310
OD1
ASN
P
262
11788
8814
8388
−3808
726
−36
O


ATOM
3311
ND2
ASN
P
262
−2.576
16.177
−4.698
1.00
81.46

N


ANISOU
3311
ND2
ASN
P
262
11948
9632
9373
−4089
373
85
N


ATOM
3312
N
ASP
P
263
−0.999
19.566
−8.004
1.00
97.04

N


ANISOU
3312
N
ASP
P
263
14221
10740
11909
−4514
1020
558
N


ATOM
3313
CA
ASP
P
263
−0.983
21.013
−7.804
1.00
100.49

C


ANISOU
3313
CA
ASP
P
263
14953
10871
12357
−4727
858
490
C


ATOM
3314
C
ASP
P
263
−1.686
21.770
−8.934
1.00
102.79

C


ANISOU
3314
C
ASP
P
263
15690
10934
12433
−4545
1113
477
C


ATOM
3315
O
ASP
P
263
−2.186
22.876
−8.727
1.00
106.47

O


ANISOU
3315
O
ASP
P
263
16552
11164
12737
−4576
960
335
O


ATOM
3316
CB
ASP
P
263
0.455
21.520
−7.650
1.00
102.30

C


ANISOU
3316
CB
ASP
P
263
14866
10975
13029
−5142
815
689
C


ATOM
3317
CG
ASP
P
263
1.092
21.093
−6.337
1.00
108.70

C


ANISOU
3317
CG
ASP
P
263
15314
11971
14017
−5337
452
646
C


ATOM
3318
OD1
ASP
P
263
0.402
20.464
−5.506
1.00
114.86

O


ANISOU
3318
OD1
ASP
P
263
16137
12945
14560
−5155
235
467
O


ATOM
3319
OD2
ASP
P
263
2.291
21.390
−6.141
1.00
105.41

O


ANISOU
3319
OD2
ASP
P
263
14566
11512
13972
−5673
387
789
O


ATOM
3320
N
MET
P
264
−1.719
21.172
−10.122
1.00
99.31

N


ANISOU
3320
N
MET
P
264
15204
10558
11973
−4337
1494
624
N


ATOM
3321
CA
MET
P
264
−2.353
21.793
−11.286
1.00
101.70

C


ANISOU
3321
CA
MET
P
264
15924
10663
12055
−4121
1756
628
C


ATOM
3322
C
MET
P
264
−3.853
21.998
−11.096
1.00
101.82

C


ANISOU
3322
C
MET
P
264
16354
10719
11614
−3798
1606
325
C


ATOM
3323
O
MET
P
264
−4.487
21.285
−10.318
1.00
107.68

O


ANISOU
3323
O
MET
P
264
17006
11714
12195
−3678
1411
137
O


ATOM
3324
CB
MET
P
264
−2.113
20.952
−12.540
1.00
102.27

C


ANISOU
3324
CB
MET
P
264
15845
10841
12173
−3929
2178
829
C


ATOM
3325
CG
MET
P
264
−0.678
20.946
−13.026
1.00
108.04

C


ANISOU
3325
CG
MET
P
264
16229
11494
13326
−4205
2416
1147
C


ATOM
3326
SD
MET
P
264
−0.528
20.158
−14.640
1.00
100.61

S


ANISOU
3326
SD
MET
P
264
15237
10623
12367
−3926
2946
1371
S


ATOM
3327
CE
MET
P
264
−1.176
18.532
−14.285
1.00
169.29

C


ANISOU
3327
CE
MET
P
264
23718
19718
20885
−3627
2880
1227
C


ATOM
3328
N
PRO
P
265
−4.429
22.971
−11.821
1.00
98.01

N


ANISOU
3328
N
PRO
P
265
16332
9993
10916
−3647
1708
279
N


ATOM
3329
CA
PRO
P
265
−5.855
23.274
−11.736
1.00
96.48

C


ANISOU
3329
CA
PRO
P
265
16538
9840
10281
−3308
1576
−11
C


ATOM
3330
C
PRO
P
265
−6.600
22.538
−12.832
1.00
99.88

C


ANISOU
3330
C
PRO
P
265
17031
10449
10470
−2920
1854
−35
C


ATOM
3331
O
PRO
P
265
−6.903
23.135
−13.862
1.00
107.99

O


ANISOU
3331
O
PRO
P
265
18396
11298
11338
−2724
2054
4
O


ATOM
3332
CB
PRO
P
265
−5.906
24.778
−12.034
1.00
96.43

C


ANISOU
3332
CB
PRO
P
265
17008
9434
10197
−3353
1558
−9
C


ATOM
3333
CG
PRO
P
265
−4.471
25.182
−12.423
1.00
101.02

C


ANISOU
3333
CG
PRO
P
265
17421
9763
11198
−3739
1743
308
C


ATOM
3334
CD
PRO
P
265
−3.756
23.909
−12.728
1.00
99.21

C


ANISOU
3334
CD
PRO
P
265
16674
9802
11219
−3787
1950
497
C


ATOM
3335
N
ILE
P
266
−6.871
21.257
−12.630
1.00
92.24

N


ANISOU
3335
N
ILE
P
266
15761
9817
9470
−2812
1869
−98
N


ATOM
3336
CA
ILE
P
266
−7.583
20.481
−13.634
1.00
85.03

C


ANISOU
3336
CA
ILE
P
266
14890
9088
8331
−2466
2115
−145
C


ATOM
3337
C
ILE
P
266
−8.590
19.538
−12.993
1.00
86.91

C


ANISOU
3337
C
ILE
P
266
15019
9666
8336
−2297
1962
−412
C


ATOM
3338
O
ILE
P
266
−8.549
19.293
−11.785
1.00
82.19

O


ANISOU
3338
O
ILE
P
266
14252
9177
7798
−2465
1709
−505
O


ATOM
3339
CB
ILE
P
266
−6.617
19.675
−14.518
1.00
78.06

C


ANISOU
3339
CB
ILE
P
266
13719
8232
7709
−2518
2455
147
C


ATOM
3340
CG1
ILE
P
266
−5.546
19.003
−13.661
1.00
80.88

C


ANISOU
3340
CG1
ILE
P
266
13614
8689
8428
−2834
2367
295
C


ATOM
3341
CG2
ILE
P
266
−5.970
20.572
−15.555
1.00
79.39

C


ANISOU
3341
CG2
ILE
P
266
14092
8085
7986
−2562
2711
381
C


ATOM
3342
CD1
ILE
P
266
−4.627
18.100
−14.447
1.00
81.89

C


ANISOU
3342
CD1
ILE
P
266
13428
8887
8799
−2850
2692
565
C


ATOM
3343
N
THR
P
267
−9.499
19.019
−13.812
1.00
80.38

N


ANISOU
3343
N
THR
P
267
14300
9006
7236
−1969
2120
−540
N


ATOM
3344
CA
THR
P
267
−10.486
18.064
−13.339
1.00
74.40

C


ANISOU
3344
CA
THR
P
267
13432
8579
6258
−1823
2026
−796
C


ATOM
3345
C
THR
P
267
−9.782
16.825
−12.802
1.00
74.01

C


ANISOU
3345
C
THR
P
267
12977
8689
6456
−2022
2059
−671
C


ATOM
3346
O
THR
P
267
−8.683
16.485
−13.251
1.00
67.48

O


ANISOU
3346
O
THR
P
267
11949
7775
5917
−2152
2249
−391
O


ATOM
3347
CB
THR
P
267
−11.443
17.644
−14.465
1.00
73.99

C


ANISOU
3347
CB
THR
P
267
13523
8679
5910
−1462
2221
−933
C


ATOM
3348
OG1
THR
P
267
−10.701
17.004
−15.514
1.00
60.56

O


ANISOU
3348
OG1
THR
P
267
11695
6938
4375
−1444
2539
−686
O


ATOM
3349
CG2
THR
P
267
−12.174
18.855
−15.024
1.00
77.11

C


ANISOU
3349
CG2
THR
P
267
14340
8931
6028
−1209
2179
−1063
C


ATOM
3350
N
ASN
P
268
−10.417
16.159
−11.840
1.00
59.47

N


ANISOU
3350
N
ASN
P
268
11025
7080
4491
−2033
1884
−876
N


ATOM
3351
CA
ASN
P
268
−9.888
14.920
−11.286
1.00
48.08

C


ANISOU
3351
CA
ASN
P
268
9252
5793
3225
−2179
1909
−783
C


ATOM
3352
C
ASN
P
268
−9.596
13.891
−12.375
1.00
55.47

C


ANISOU
3352
C
ASN
P
268
10056
6797
4225
−2062
2237
−637
C


ATOM
3353
O
ASN
P
268
−8.630
13.133
−12.273
1.00
54.86

O


ANISOU
3353
O
ASN
P
268
9713
6728
4405
−2194
2331
−421
O


ATOM
3354
CB
ASN
P
268
−10.855
14.328
−10.257
1.00
53.90

C


ANISOU
3354
CB
ASN
P
268
9967
6772
3740
−2156
1726
−1056
C


ATOM
3355
CG
ASN
P
268
−10.927
15.144
−8.978
1.00
58.38

C


ANISOU
3355
CG
ASN
P
268
10612
7284
4285
−2303
1394
−1166
C


ATOM
3356
OD1
ASN
P
268
−10.158
16.083
−8.779
1.00
75.62

O


ANISOU
3356
OD1
ASN
P
268
12839
9243
6652
−2460
1280
−1032
O


ATOM
3357
ND2
ASN
P
268
−11.852
14.783
−8.101
1.00
47.61

N


ANISOU
3357
ND2
ASN
P
268
9272
6124
2693
−2264
1246
−1417
N


ATOM
3358
N
ASP
P
269
−10.431
13.863
−13.413
1.00
50.59

N


ANISOU
3358
N
ASP
P
269
9628
6233
3359
−1795
2401
−762
N


ATOM
3359
CA
ASP
P
269
−10.226
12.943
−14.528
1.00
56.37

C


ANISOU
3359
CA
ASP
P
269
10286
7018
4115
−1657
2711
−646
C


ATOM
3360
C
ASP
P
269
−8.905
13.221
−15.234
1.00
59.80

C


ANISOU
3360
C
ASP
P
269
10635
7235
4851
−1743
2918
−294
C


ATOM
3361
O
ASP
P
269
−8.203
12.299
−15.639
1.00
62.89

O


ANISOU
3361
O
ASP
P
269
10824
7662
5411
−1754
3128
−108
O


ATOM
3362
CB
ASP
P
269
−11.370
13.039
−15.538
1.00
71.42

C


ANISOU
3362
CB
ASP
P
269
12391
8940
5804
−1348
2747
−844
C


ATOM
3363
CG
ASP
P
269
−12.648
12.395
−15.041
1.00
84.67

C


ANISOU
3363
CG
ASP
P
269
13937
10746
7487
−1286
2495
−1096
C


ATOM
3364
OD1
ASP
P
269
−12.604
11.690
−14.010
1.00
92.63

O


ANISOU
3364
OD1
ASP
P
269
14755
11867
8574
−1464
2388
−1136
O


ATOM
3365
OD2
ASP
P
269
−13.698
12.591
−15.688
1.00
83.85

O


ANISOU
3365
OD2
ASP
P
269
13907
10648
7302
−1064
2423
−1224
O


ATOM
3366
N
GLN
P
270
−8.573
14.497
−15.390
1.00
64.57

N


ANISOU
3366
N
GLN
P
270
11405
7611
5517
−1803
2873
−206
N


ATOM
3367
CA
GLN
P
270
−7.335
14.872
−16.063
1.00
71.87

C


ANISOU
3367
CA
GLN
P
270
12255
8322
6732
−1916
3088
124
C


ATOM
3368
C
GLN
P
270
−6.111
14.487
−15.232
1.00
66.25

C


ANISOU
3368
C
GLN
P
270
11165
7611
6395
−2220
3022
333
C


ATOM
3369
O
GLN
P
270
−5.112
14.014
−15.771
1.00
63.28

O


ANISOU
3369
O
GLN
P
270
10569
7210
6264
−2269
3258
593
O


ATOM
3370
CB
GLN
P
270
−7.334
16.364
−16.420
1.00
77.84

C


ANISOU
3370
CB
GLN
P
270
13324
8807
7444
−1924
3063
156
C


ATOM
3371
CG
GLN
P
270
−8.148
16.691
−17.673
1.00
80.33

C


ANISOU
3371
CG
GLN
P
270
13996
9079
7446
−1579
3248
70
C


ATOM
3372
CD
GLN
P
270
−8.616
18.136
−17.723
1.00
83.13

C


ANISOU
3372
CD
GLN
P
270
14748
9212
7627
−1516
3120
−21
C


ATOM
3373
OE1
GLN
P
270
−8.413
18.901
−16.780
1.00
84.81

O


ANISOU
3373
OE1
GLN
P
270
14984
9302
7938
−1737
2880
−48
O


ATOM
3374
NE2
GLN
P
270
−9.256
18.512
−18.824
1.00
79.56

N


ANISOU
3374
NE2
GLN
P
270
14633
8701
6894
−1195
3272
−78
N


ATOM
3375
N
LYS
P
271
−6.196
14.678
−13.918
1.00
60.48

N


ANISOU
3375
N
LYS
P
271
10357
6924
5697
−2403
2697
213
N


ATOM
3376
CA
LYS
P
271
−5.116
14.283
−13.018
1.00
55.55

C


ANISOU
3376
CA
LYS
P
271
9378
6334
5394
−2666
2580
374
C


ATOM
3377
C
LYS
P
271
−4.934
12.773
−13.079
1.00
57.38

C


ANISOU
3377
C
LYS
P
271
9365
6774
5663
−2571
2725
433
C


ATOM
3378
O
LYS
P
271
−3.825
12.271
−13.244
1.00
62.74

O


ANISOU
3378
O
LYS
P
271
9755
7458
6624
−2654
2863
681
O


ATOM
3379
CB
LYS
P
271
−5.428
14.700
−11.578
1.00
63.38

C


ANISOU
3379
CB
LYS
P
271
10389
7354
6338
−2828
2186
190
C


ATOM
3380
CG
LYS
P
271
−5.798
16.165
−11.399
1.00
71.40

C


ANISOU
3380
CG
LYS
P
271
11709
8164
7255
−2891
2007
77
C


ATOM
3381
CD
LYS
P
271
−5.976
16.508
−9.923
1.00
73.41

C


ANISOU
3381
CD
LYS
P
271
11962
8448
7481
−3058
1618
−89
C


ATOM
3382
CE
LYS
P
271
−6.348
17.972
−9.729
1.00
79.73

C


ANISOU
3382
CE
LYS
P
271
13100
9024
8170
−3106
1436
−210
C


ATOM
3383
NZ
LYS
P
271
−6.484
18.328
−8.289
1.00
81.22

N


ANISOU
3383
NZ
LYS
P
271
13306
9231
8324
−3263
1055
−373
N


ATOM
3384
N
LYS
P
272
−6.045
12.061
−12.936
1.00
59.97

N


ANISOU
3384
N
LYS
P
272
9815
7272
5698
−2396
2694
196
N


ATOM
3385
CA
LYS
P
272
−6.067
10.609
−13.005
1.00
64.48

C


ANISOU
3385
CA
LYS
P
272
10236
8018
6247
−2295
2835
211
C


ATOM
3386
C
LYS
P
272
−5.540
10.125
−14.347
1.00
65.65

C


ANISOU
3386
C
LYS
P
272
10337
8126
6481
−2144
3203
414
C


ATOM
3387
O
LYS
P
272
−4.759
9.179
−14.420
1.00
73.32

O


ANISOU
3387
O
LYS
P
272
11076
9153
7629
−2143
3344
594
O


ATOM
3388
CB
LYS
P
272
−7.498
10.111
−12.791
1.00
71.71

C


ANISOU
3388
CB
LYS
P
272
11343
9099
6804
−2148
2769
−110
C


ATOM
3389
CG
LYS
P
272
−7.654
8.607
−12.842
1.00
69.02

C


ANISOU
3389
CG
LYS
P
272
10904
8914
6405
−2062
2916
−129
C


ATOM
3390
CD
LYS
P
272
−9.070
8.192
−12.486
1.00
63.72

C


ANISOU
3390
CD
LYS
P
272
10380
8374
5458
−1978
2784
−475
C


ATOM
3391
CE
LYS
P
272
−9.260
6.706
−12.712
1.00
72.38

C


ANISOU
3391
CE
LYS
P
272
11382
9479
6638
−1889
2842
−522
C


ATOM
3392
NZ
LYS
P
272
−10.652
6.276
−12.439
1.00
78.98

N


ANISOU
3392
NZ
LYS
P
272
12280
10361
7368
−1861
2666
−820
N


ATOM
3393
N
LEU
P
273
−5.982
10.782
−15.412
1.00
72.34

N


ANISOU
3393
N
LEU
P
273
11427
8876
7183
−1992
3357
382
N


ATOM
3394
CA
LEU
P
273
−5.528
10.465
−16.757
1.00
79.75

C


ANISOU
3394
CA
LEU
P
273
12374
9757
8168
−1828
3715
570
C


ATOM
3395
C
LEU
P
273
−4.010
10.595
−16.852
1.00
79.24

C


ANISOU
3395
C
LEU
P
273
12026
9589
8493
−1996
3846
913
C


ATOM
3396
O
LEU
P
273
−3.330
9.710
−17.371
1.00
75.30

O


ANISOU
3396
O
LEU
P
273
11345
9139
8126
−1917
4088
1097
O


ATOM
3397
CB
LEU
P
273
−6.200
11.395
−17.767
1.00
94.04

C


ANISOU
3397
CB
LEU
P
273
14528
11450
9753
−1649
3815
487
C


ATOM
3398
CG
LEU
P
273
−5.770
11.277
−19.227
1.00
113.16

C


ANISOU
3398
CG
LEU
P
273
17028
13782
12187
−1462
4190
682
C


ATOM
3399
CD1
LEU
P
273
−6.239
9.956
−19.811
1.00
112.29

C


ANISOU
3399
CD1
LEU
P
273
16900
13764
12001
−1203
4252
540
C


ATOM
3400
CD2
LEU
P
273
−6.307
12.451
−20.037
1.00
120.74

C


ANISOU
3400
CD2
LEU
P
273
18353
14581
12941
−1316
4239
626
C


ATOM
3401
N
MET
P
274
−3.485
11.700
−16.334
1.00
79.04

N


ANISOU
3401
N
MET
P
274
11957
9425
8650
−2231
3683
989
N


ATOM
3402
CA
MET
P
274
−2.059
11.987
−16.418
1.00
74.78

C


ANISOU
3402
CA
MET
P
274
11127
8792
8495
−2431
3795
1296
C


ATOM
3403
C
MET
P
274
−1.203
11.155
−15.457
1.00
65.60

C


ANISOU
3403
C
MET
P
274
9560
7778
7586
−2574
3665
1398
C


ATOM
3404
O
MET
P
274
−0.024
10.928
−15.710
1.00
74.49

O


ANISOU
3404
O
MET
P
274
10378
8910
9013
−2651
3830
1656
O


ATOM
3405
CB
MET
P
274
−1.801
13.479
−16.198
1.00
74.94

C


ANISOU
3405
CB
MET
P
274
11256
8595
8623
−2663
3660
1326
C


ATOM
3406
CG
MET
P
274
−2.416
14.374
−17.260
1.00
74.28

C


ANISOU
3406
CG
MET
P
274
11579
8324
8320
−2509
3832
1289
C


ATOM
3407
SD
MET
P
274
−2.167
16.114
−16.892
1.00
88.02

S


ANISOU
3407
SD
MET
P
274
13510
9770
10163
−2789
3656
1307
S


ATOM
3408
CE
MET
P
274
−0.385
16.157
−16.707
1.00
90.50

C


ANISOU
3408
CE
MET
P
274
13341
10039
11004
−3149
3763
1647
C


ATOM
3409
N
SER
P
275
−1.786
10.698
−14.357
1.00
58.19

N


ANISOU
3409
N
SER
P
275
8622
6967
6520
−2596
3377
1198
N


ATOM
3410
CA
SER
P
275
−1.010
9.947
−13.374
1.00
64.47

C


ANISOU
3410
CA
SER
P
275
9078
7895
7523
−2708
3223
1286
C


ATOM
3411
C
SER
P
275
−0.910
8.458
−13.721
1.00
74.91

C


ANISOU
3411
C
SER
P
275
10295
9363
8806
−2489
3436
1353
C


ATOM
3412
O
SER
P
275
0.101
7.817
−13.440
1.00
72.80

O


ANISOU
3412
O
SER
P
275
9708
9176
8775
−2514
3463
1541
O


ATOM
3413
CB
SER
P
275
−1.561
10.161
−11.957
1.00
60.08

C


ANISOU
3413
CB
SER
P
275
8580
7392
6856
−2841
2817
1068
C


ATOM
3414
OG
SER
P
275
−2.883
9.673
−11.829
1.00
70.03

O


ANISOU
3414
OG
SER
P
275
10112
8736
7759
−2675
2778
807
O


ATOM
3415
N
ASN
P
276
−1.963
7.919
−14.334
1.00
83.21

N


ANISOU
3415
N
ASN
P
276
11620
10445
9551
−2269
3580
1189
N


ATOM
3416
CA
ASN
P
276
−1.973
6.536
−14.813
1.00
85.57

C


ANISOU
3416
CA
ASN
P
276
11892
10842
9777
−2054
3812
1230
C


ATOM
3417
C
ASN
P
276
−1.096
6.359
−16.042
1.00
96.38

C


ANISOU
3417
C
ASN
P
276
13146
12159
11314
−1929
4178
1491
C


ATOM
3418
O
ASN
P
276
−0.144
5.567
−16.049
1.00
92.15

O


ANISOU
3418
O
ASN
P
276
12348
11689
10978
−1880
4301
1694
O


ATOM
3419
CB
ASN
P
276
−3.381
6.118
−15.237
1.00
75.58

C


ANISOU
3419
CB
ASN
P
276
10961
9605
8152
−1863
3840
940
C


ATOM
3420
CG
ASN
P
276
−4.332
5.986
−14.084
1.00
72.43

C


ANISOU
3420
CG
ASN
P
276
10672
9306
7544
−1958
3565
685
C


ATOM
3421
OD1
ASN
P
276
−3.988
5.453
−13.032
1.00
74.52

O


ANISOU
3421
OD1
ASN
P
276
10786
9642
7885
−2060
3406
716
O


ATOM
3422
ND2
ASN
P
276
−5.554
6.460
−14.280
1.00
76.67

N


ANISOU
3422
ND2
ASN
P
276
11472
9834
7825
−1890
3468
404
N


ATOM
3423
N
ASN
P
277
−1.460
7.108
−17.082
1.00
109.71

N


ANISOU
3423
N
ASN
P
277
15053
13728
12903
−1847
4339
1470
N


ATOM
3424
CA
ASN
P
277
−0.906
6.972
−18.430
1.00
126.53

C


ANISOU
3424
CA
ASN
P
277
17182
15771
15122
−1646
4669
1637
C


ATOM
3425
C
ASN
P
277
0.332
6.120
−18.713
1.00
157.12

C


ANISOU
3425
C
ASN
P
277
20737
19702
19260
−1569
4888
1908
C


ATOM
3426
O
ASN
P
277
0.450
5.458
−19.705
1.00
165.30

O


ANISOU
3426
O
ASN
P
277
21836
20704
20266
−1305
5122
1951
O


ATOM
3427
CB
ASN
P
277
−0.925
8.285
−19.231
1.00
126.96

C


ANISOU
3427
CB
ASN
P
277
17414
15664
15160
−1697
4809
1710
C


ATOM
3428
CG
ASN
P
277
0.019
9.368
−18.832
1.00
135.53

C


ANISOU
3428
CG
ASN
P
277
18295
16661
16538
−2011
4771
1919
C


ATOM
3429
OD1
ASN
P
277
−0.273
10.174
−17.959
1.00
144.85

O


ANISOU
3429
OD1
ASN
P
277
19523
17793
17721
−2213
4463
1791
O


ATOM
3430
ND2
ASN
P
277
0.991
9.583
−19.696
1.00
136.64

N


ANISOU
3430
ND2
ASN
P
277
18299
16728
16891
−2014
5086
2191
N


END










The Ramachandran plot shows 95.6% of all residues in favored regions and 99.3% of all residues in allowed regions. All structural images were created using PyMol (Delano Scientific, http://www.pymol.org).


f. Cloning, expression and purification of RSV F0 Fd, also referred to as RSV F0 Fd. A codon-optimized DNA fragment encoding amino acid residues 1-513 of the RSV F protein strain A2 with mutations R106Q, R109S, R135S and R136S was synthesized by GeneArt with a 3′ fragment encoding the residues SAIGGYIPEA PRDGQAYVRK DGEWVLLSTF LGGIEGRHHH HHH, also denoted herein as SEQ ID NO:15). This gene was cloned into a variant of the pHLSec mammalian expression vector (Aricescu A R, et. al., 2006, Acta Crystallogr. D Biol. Crystallogr. 62, 1243-1250) and protein was expressed using the 293Freestyle expression system as described above for the motavizumab IgG expression. Protein was purified from the supernatant using Ni-NTA resin (Qiagen, Venlo, the Netherlands) followed by gel filtration on a SUPEROSE™6 column with a running buffer of 2 mM Tris-HCl pH 7.5, 150 mM NaCl. The peak corresponding to a trimer was pooled, concentrated and stored at 4° C.


g. RSV F0 Fd cross-linking and immunoprecipitation. RSV F0 Fd (5 μg, 0.2 μM) in PBS was incubated with glutaraldehyde at concentrations of 0, 1, and 10 mM for 5 min at room temperature. Glycine was added to a final concentration of 100 mM to quench the reaction. The cross-linked and control proteins were incubated with 5 μg of motavizumab IgG for 30 min at room temperature. 20 μl of a Protein A agarose slurry (Pierce) was added and incubated for 90 min at room temperature. The resin was centrifuged, washed with PBS containing Tween 20, and then boiled in reducing SDS-PAGE loading buffer.


Example 2
Three-dimensional structure of RSV F protein and 101F antibody

This Example describes the crystallization and determination of the 3-dimensional structure of a complex between the 101F antibody and the 15-residue RSV fusion (F) peptide corresponding to amino acids 422-436 of the F protein (i.e., STASNKNRGI IKTFS, also denoted herein as SEQ ID NO:3) that includes the binding domain of 101F.


Recombinant 101F IgG and a peptide comprising the 101F binding domain were combined and the resultant complex submitted to crystallization and analysis as follows.


a. Cloning, expression and purification of 101F IgG: Two DNA fragments encoding the variable heavy and light chains of 101F with signal sequences were synthesized by GeneArt and cloned in-frame into mammalian expression vectors containing mouse IgG1 heavy and light constant domains, respectively. The amino acid sequence of the variable heavy chain of 101F is as follows: QVTLKESGPG ILQPSQTLSL TCSFSGFSLS TSGMGVSWIR QPSGKGLEWL AHIYWDDDKR YNPSLKSRLT ISKDTSRNQV FLKITSVDTA DTATYYCARL YGFTYGFAYW GQGTLVTVSA, also denoted herein as SEQ ID NO:7. The amino acid sequence of the variable light chain of 101F is as follows: DIVLTQSPAS LAVSLGQRAT IFCRASQSVD YNGISYMHWF QQKPGQPPKL LIYAASNPES GIPARFTGSG SGTDFTLNIH PVEEEDAATY YCQQIIEDPW TFGGGTKLEI K, also denoted herein as SEQ ID NO:8.


Both vectors were co-transfected at a 1:1 ratio into HEK293F cells (Invitrogen) in serum-free 293Freestyle medium (Invitrogen). After 3 hours, valproic acid (Sigma) was added to 4 mM final concentration. Expression lasted for five days at 37° C. with 10% CO2 and shaking at 125 rpm in disposable flasks. The supernatant was collected, filtered, and passed over 5 ml of Protein G agarose resin (Pierce). After washing with several column volumes of phosphate-buffered saline (PBS), the resin was eluted with 15 ml of IgG Elution Buffer (Pierce) and immediately neutralized with 1 M Tris pH 8.0. The eluted protein was dialyzed against PBS and stored at 4° C.


b. Digestion and purification of 101F Fab fragments. The purified 101F IgG protein was reduced with 100 mM dithiothreitol at 37° C. for 1 hour and then alkylated with 2 mM iodoacetamide for 48 hours at 4° C. Ten ml of reduced and alkylated IgG in PBS at 1.5 mg/ml was combined with 0.275 ODs of Ficin (Sigma), 20 mM L-cysteine, 1 mM EDTA and incubated at 37° C. for 1 hour. The reaction was quenched by the addition of iodoacetamide to 40 mM final concentration. To remove the Fc fragments from the Fab fragments, the quenched reaction was passed over 1 ml of Protein A agarose. The Fab-containing flow through was further purified over an S200 gel filtration column and concentrated aliquots were stored frozen at −80° C.


c. Protein crystallization and data collection. A peptide with the sequence STASNKNRGI IKTFS (SEQ ID NO:3), corresponding to the originally identified 101F epitope of CTASNKNRGI IKTFS (residues 422-436 of RSV F protein), also denoted herein as SEQ ID NO:10, was synthesized by American Peptide with an acetylated N-terminus and an amidated C-terminus. A five-fold molar excess of peptide was incubated with 101F Fab at 22° C. for 1.5 hours and then concentrated to give an 8.3 mg/ml solution of Fab/peptide complex. Crystals were grown by the vapor diffusion method in sitting drops at 20° C. by mixing 1 μl of protein complex with 1 μl of reservoir solution (15% (w/v) PEG 4000, 0.2 M lithium sulfate, 0.1 M Tris pH 8.5). These crystals were flash frozen in liquid nitrogen in 20% (w/v) PEG 4000, 0.2M lithium sulfate, 0.1M Tris pH 8.5 and 15% (v/v) 2R,3R-butanediol. Data were collected at a wavelength of 0.82656 Å at the SER-CAT beamline ID-22 (Advanced Photon Source, Argonne National Laboratory).


The structure of the 101F/peptide complex was determined and a model built and refined using a method similar to that described in Example 1. Final data collection and refinement statistics are presented in Table 4.


The atomic coordinates for the complex between the 15-residue F peptide and 101F antibody are indicated in PDB acc code 3O41. The atomic coordinates for the peptide portion of the complex are indicated below in Table 7.









TABLE 7





Atomic coordinates of the 101F binding peptide



























ATOM
6696
O
LYS
P
427
−17.126
3.607
14.869
1.00
82.36

O


ANISOU
6696
O
LYS
P
427
11025
9697
10570
618
188
376
O


ATOM
6697
N
LYS
P
427
−17.615
3.567
18.200
1.00
90.82

N


ANISOU
6697
N
LYS
P
427
12312
10602
11594
727
198
440
N


ATOM
6698
CA
LYS
P
427
−18.312
3.323
16.942
1.00
89.25

C


ANISOU
6698
CA
LYS
P
427
12014
10493
11404
684
241
396
C


ATOM
6699
C
LYS
P
427
−17.352
2.880
15.838
1.00
83.29

C


ANISOU
6699
C
LYS
P
427
11189
9761
10696
633
229
378
C


ATOM
6700
CB
LYS
P
427
−19.092
4.566
16.507
1.00
92.06

C


ANISOU
6700
CB
LYS
P
427
12357
10914
11709
704
213
400
C


ATOM
6701
CG
LYS
P
427
−18.301
5.861
16.568
1.00
92.91

C


ANISOU
6701
CG
LYS
P
427
12507
11004
11790
733
117
442
C


ATOM
6702
CD
LYS
P
427
−19.182
7.057
16.249
1.00
93.83

C


ANISOU
6702
CD
LYS
P
427
12615
11181
11854
756
94
446
C


ATOM
6703
CE
LYS
P
427
−18.427
8.362
16.431
1.00
95.87

C


ANISOU
6703
CE
LYS
P
427
12924
11417
12084
790
−2
490
C


ATOM
6704
NZ
LYS
P
427
−19.183
9.529
15.890
1.00
97.27

N


ANISOU
6704
NZ
LYS
P
427
13079
11662
12216
804
−30
491
N


ATOM
6705
O
ASN
P
428
−17.432
0.121
13.533
1.00
63.07

O


ANISOU
6705
O
ASN
P
428
8415
7298
8250
494
373
266
O


ATOM
6706
N
ASN
P
428
−16.796
1.681
15.999
1.00
77.41

N


ANISOU
6706
N
ASN
P
428
10437
8975
10000
607
264
364
N


ATOM
6707
CA
ASN
P
428
−15.846
1.113
15.044
1.00
72.01

C


ANISOU
6707
CA
ASN
P
428
9689
8305
9366
558
258
347
C


ATOM
6708
C
ASN
P
428
−16.459
0.867
13.665
1.00
62.88

C


ANISOU
6708
C
ASN
P
428
8429
7243
8221
511
299
301
C


ATOM
6709
CB
ASN
P
428
−15.262
−0.194
15.597
1.00
75.94

C


ANISOU
6709
CB
ASN
P
428
10204
8738
9913
541
296
340
C


ATOM
6710
CG
ASN
P
428
−14.397
−0.927
14.583
1.00
78.67

C


ANISOU
6710
CG
ASN
P
428
10478
9101
10312
488
302
315
C


ATOM
6711
OD1
ASN
P
428
−13.646
−0.310
13.824
1.00
78.53

O


ANISOU
6711
OD1
ASN
P
428
10433
9107
10298
475
246
324
O


ATOM
6712
ND2
ASN
P
428
−14.493
−2.255
14.574
1.00
79.70

N


ANISOU
6712
ND2
ASN
P
428
10577
9219
10484
456
368
283
N


ATOM
6713
O
ARG
P
429
−16.017
−0.008
9.350
1.00
42.31

O


ANISOU
6713
O
ARG
P
429
5527
4851
5699
350
340
193
O


ATOM
6714
N
ARG
P
429
−15.878
1.492
12.642
1.00
51.34

N


ANISOU
6714
N
ARG
P
429
6920
5825
6761
490
250
301
N


ATOM
6715
CA
ARG
P
429
−16.374
1.375
11.272
1.00
46.50

C


ANISOU
6715
CA
ARG
P
429
6208
5304
6157
445
280
260
C


ATOM
6716
C
ARG
P
429
−15.659
0.274
10.490
1.00
43.56

C


ANISOU
6716
C
ARG
P
429
5770
4938
5843
392
309
230
C


ATOM
6717
CB
ARG
P
429
−16.163
2.690
10.515
1.00
44.81

C


ANISOU
6717
CB
ARG
P
429
5973
5139
5912
450
211
277
C


ATOM
6718
CG
ARG
P
429
−17.099
3.817
10.872
1.00
45.89

C


ANISOU
6718
CG
ARG
P
429
6145
5300
5990
492
190
294
C


ATOM
6719
CD
ARG
P
429
−18.397
3.691
10.094
1.00
45.47

C


ANISOU
6719
CD
ARG
P
429
6021
5331
5924
469
249
254
C


ATOM
6720
NE
ARG
P
429
−19.454
3.112
10.897
1.00
43.13

N


ANISOU
6720
NE
ARG
P
429
5752
5019
5617
488
315
242
N


ATOM
6721
CZ
ARG
P
429
−20.614
2.677
10.413
1.00
45.12

C


ANISOU
6721
CZ
ARG
P
429
5947
5330
5866
467
382
204
C


ATOM
6722
NH1
ARG
P
429
−20.876
2.723
9.114
1.00
37.74

N


ANISOU
6722
NH1
ARG
P
429
4925
4475
4939
426
393
173
N


ATOM
6723
NH2
ARG
P
429
−21.514
2.173
11.239
1.00
49.71

N


ANISOU
6723
NH2
ARG
P
429
6560
5890
6437
487
439
197
N


ATOM
6724
O
GLY
P
430
−12.500
0.641
9.683
1.00
33.44

O


ANISOU
6724
O
GLY
P
430
4482
3601
4623
360
174
272
O


ATOM
6725
N
GLY
P
430
−14.632
−0.322
11.084
1.00
41.59

N


ANISOU
6725
N
GLY
P
430
5560
4614
5628
392
296
246
N


ATOM
6726
CA
GLY
P
430
−13.790
−1.264
10.361
1.00
40.69

C


ANISOU
6726
CA
GLY
P
430
5390
4501
5571
344
312
222
C


ATOM
6727
C
GLY
P
430
−12.815
−0.528
9.449
1.00
37.79

C


ANISOU
6727
C
GLY
P
430
4989
4161
5207
327
244
234
C


ATOM
6728
O
ILE
P
431
−13.217
0.553
6.316
1.00
33.85

O


ANISOU
6728
O
ILE
P
431
4303
3864
4696
252
220
183
O


ATOM
6729
N
ILE
P
431
−12.341
−1.205
8.408
1.00
36.65

N


ANISOU
6729
N
ILE
P
431
4768
4051
5106
277
263
203
N


ATOM
6730
CA
ILE
P
431
−11.431
−0.596
7.441
1.00
31.75

C


ANISOU
6730
CA
ILE
P
431
4108
3463
4494
256
204
210
C


ATOM
6731
C
ILE
P
431
−12.075
0.617
6.767
1.00
31.38

C


ANISOU
6731
C
ILE
P
431
4033
3489
4399
264
172
213
C


ATOM
6732
CB
ILE
P
431
−10.982
−1.638
6.397
1.00
32.82

C


ANISOU
6732
CB
ILE
P
431
4158
3629
4682
197
243
170
C


ATOM
6733
CG1
ILE
P
431
−10.183
−2.745
7.083
1.00
34.89

C


ANISOU
6733
CG1
ILE
P
431
4452
3813
4992
191
263
172
C


ATOM
6734
CG2
ILE
P
431
−10.131
−1.010
5.305
1.00
30.23

C


ANISOU
6734
CG2
ILE
P
431
3781
3343
4360
173
186
174
C


ATOM
6735
CD1
ILE
P
431
−8.873
−2.256
7.701
1.00
35.00

C


ANISOU
6735
CD1
ILE
P
431
4532
3755
5013
216
188
220
C


ATOM
6736
O
ILE
P
432
−11.819
4.214
4.076
1.00
32.03

O


ANISOU
6736
O
ILE
P
432
4015
3759
4396
253
−4
243
O


ATOM
6737
N
ILE
P
432
−11.352
1.736
6.711
1.00
32.07

N


ANISOU
6737
N
ILE
P
432
4149
3570
4466
283
90
251
N


ATOM
6738
CA
ILE
P
432
−11.905
2.955
6.116
1.00
34.42

C


ANISOU
6738
CA
ILE
P
432
4426
3934
4719
293
53
257
C


ATOM
6739
C
ILE
P
432
−11.284
3.355
4.773
1.00
30.32

C


ANISOU
6739
C
ILE
P
432
3834
3476
4211
255
16
247
C


ATOM
6740
CB
ILE
P
432
−11.919
4.150
7.094
1.00
37.38

C


ANISOU
6740
CB
ILE
P
432
4889
4268
5045
352
−10
306
C


ATOM
6741
CG1
ILE
P
432
−10.559
4.332
7.760
1.00
40.47

C


ANISOU
6741
CG1
ILE
P
432
5345
4580
5453
372
−74
349
C


ATOM
6742
CG2
ILE
P
432
−13.019
3.969
8.144
1.00
37.48

C


ANISOU
6742
CG2
ILE
P
432
4955
4256
5031
387
36
306
C


ATOM
6743
CD1
ILE
P
432
−10.449
5.644
8.535
1.00
44.51

C


ANISOU
6743
CD1
ILE
P
432
5936
5058
5916
426
−148
399
C


ATOM
6744
O
LYS
P
433
−8.171
0.912
3.440
1.00
32.29

O


ANISOU
6744
O
LYS
P
433
3981
3672
4616
147
29
212
O


ATOM
6745
N
LYS
P
433
−10.167
2.730
4.420
1.00
30.10

N


ANISOU
6745
N
LYS
P
433
3782
3424
4229
226
7
244
N


ATOM
6746
CA
LYS
P
433
−9.620
2.800
3.062
1.00
33.41

C


ANISOU
6746
CA
LYS
P
433
4120
3905
4670
181
−10
224
C


ATOM
6747
C
LYS
P
433
−8.993
1.454
2.691
1.00
33.94

C


ANISOU
6747
C
LYS
P
433
4143
3957
4797
137
36
194
C


ATOM
6748
CB
LYS
P
433
−8.572
3.917
2.925
1.00
32.29

C


ANISOU
6748
CB
LYS
P
433
4003
3751
4514
196
−105
267
C


ATOM
6749
CG
LYS
P
433
−9.060
5.302
3.329
1.00
29.89

C


ANISOU
6749
CG
LYS
P
433
3748
3457
4153
241
−159
301
C


ATOM
6750
CD
LYS
P
433
−7.969
6.352
3.086
1.00
33.80

C


ANISOU
6750
CD
LYS
P
433
4260
3944
4639
251
−253
342
C


ATOM
6751
CE
LYS
P
433
−8.434
7.761
3.476
1.00
31.89

C


ANISOU
6751
CE
LYS
P
433
4067
3711
4339
296
−310
376
C


ATOM
6752
NZ
LYS
P
433
−7.389
8.761
3.068
1.00
34.77

N


ANISOU
6752
NZ
LYS
P
433
4436
4078
4697
300
−400
412
N


ATOM
6753
O
THR
P
434
−7.088
0.909
0.074
1.00
30.52

O


ANISOU
6753
O
THR
P
434
3548
3608
4441
34
6
154
O


ATOM
6754
N
THR
P
434
−9.396
0.919
1.541
1.00
35.59

N


ANISOU
6754
N
THR
P
434
4261
4237
5023
90
83
148
N


ATOM
6755
CA
THR
P
434
−8.888
−0.360
1.047
1.00
35.52

C


ANISOU
6755
CA
THR
P
434
4201
4225
5071
45
130
114
C


ATOM
6756
C
THR
P
434
−7.729
−0.147
0.049
1.00
31.98

C


ANISOU
6756
C
THR
P
434
3704
3800
4647
13
82
116
C


ATOM
6757
CB
THR
P
434
−10.029
−1.234
0.462
1.00
32.20

C


ANISOU
6757
CB
THR
P
434
3712
3862
4659
12
217
60
C


ATOM
6758
OG1
THR
P
434
−10.738
−0.492
−0.539
1.00
32.65

O


ANISOU
6758
OG1
THR
P
434
3712
4008
4686
−3
211
44
O


ATOM
6759
CG2
THR
P
434
−11.021
−1.639
1.567
1.00
32.06

C


ANISOU
6759
CG2
THR
P
434
3748
3808
4627
43
267
59
C


ATOM
6760
N
PHE
P
435
−7.440
−1.137
−0.802
1.00
31.73

N


ANISOU
6760
N
PHE
P
435
3600
3795
4659
−36
125
77
N


ATOM
6761
CA
PHE
P
435
−6.234
−1.085
−1.658
1.00
31.87

C


ANISOU
6761
CA
PHE
P
435
3576
3826
4707
−66
84
79
C


ATOM
6762
C
PHE
P
435
−6.339
−0.069
−2.801
1.00
29.79

C


ANISOU
6762
C
PHE
P
435
3258
3646
4417
−82
44
77
C


ATOM
6763
O
PHE
P
435
−7.419
0.179
−3.308
1.00
30.98

O


ANISOU
6763
O
PHE
P
435
3369
3862
4540
−91
73
54
O


ATOM
6764
CB
PHE
P
435
−5.931
−2.446
−2.304
1.00
30.00

C


ANISOU
6764
CB
PHE
P
435
3274
3599
4525
−116
144
35
C


ATOM
6765
CG
PHE
P
435
−5.662
−3.565
−1.320
1.00
31.76

C


ANISOU
6765
CG
PHE
P
435
3540
3741
4784
−108
183
33
C


ATOM
6766
CD1
PHE
P
435
−6.441
−4.717
−1.337
1.00
32.77

C


ANISOU
6766
CD1
PHE
P
435
3637
3880
4934
−130
267
−9
C


ATOM
6767
CD2
PHE
P
435
−4.621
−3.473
−0.406
1.00
29.15

C


ANISOU
6767
CD2
PHE
P
435
3282
3327
4468
−81
136
75
C


ATOM
6768
CE1
PHE
P
435
−6.193
−5.770
−0.448
1.00
35.62

C


ANISOU
6768
CE1
PHE
P
435
4037
4167
5329
−124
303
−11
C


ATOM
6769
CE2
PHE
P
435
−4.360
−4.515
0.490
1.00
29.66

C


ANISOU
6769
CE2
PHE
P
435
3386
3317
4566
−75
172
73
C


ATOM
6770
CZ
PHE
P
435
−5.153
−5.665
0.468
1.00
32.59

C


ANISOU
6770
CZ
PHE
P
435
3725
3700
4958
−97
256
30
C


ATOM
6771
O
SER
P
436
−2.853
0.265
−4.625
1.00
35.91

O


ANISOU
6771
O
SER
P
436
3954
4425
5266
−146
−87
105
O


ATOM
6772
N
SER
P
436
−5.195
0.475
−3.216
1.00
28.46

N


ANISOU
6772
N
SER
P
436
3084
3474
4257
−89
−22
102
N


ATOM
6773
CA
SER
P
436
−5.063
1.195
−4.487
1.00
33.17

C


ANISOU
6773
CA
SER
P
436
3613
4149
4841
−116
−56
94
C


ATOM
6774
C
SER
P
436
−3.845
0.674
−5.250
1.00
35.26

C


ANISOU
6774
C
SER
P
436
3829
4414
5152
−155
−68
84
C


ATOM
6775
CB
SER
P
436
−4.908
2.702
−4.256
1.00
31.56

C


ANISOU
6775
CB
SER
P
436
3455
3947
4591
−79
−139
141
C


ATOM
6776
OG
SER
P
436
−6.064
3.244
−3.639
1.00
36.23

O


ANISOU
6776
OG
SER
P
436
4084
4544
5136
−44
−129
148
O


END










In the complex between the 15-residue F peptide and 101F, only the amino acids KNRGIIKTFS (SEQ ID NO:4) are modeled in the crystal structure as the remaining residues are disordered. As such, the coordinates disclosed in PDB acc code 3O41 only include those 10 amino acids of the 15-residue peptide.


Example 3
Production and Characterization of Scaffold-Based Immunogens

This Example describes the production and testing of scaffold-based immunogens designed using the atomic coordinates in PDB acc code 3IXT, i.e., the atomic coordinates of the complex between the 24-residue F peptide comprising the motavizumab binding domain and motavizumab.


Several scaffolds were designed to present the motavizumab epitope, or binding domain, using the superposition method (also referred to as side chain grafting) and the multi-segment side chain grafting method (also referred to as double superpositioning). Multi-segment side chain grafting is an extension of the superposition method described in WO 2008/025015 A2. Multi-segment side chain grafting is intended for transplantation of certain complex epitopes to scaffold proteins, in which the epitope contains two or more backbone segments in a fixed orientation relative to each other; e.g., the motavizumab epitope is composed of two helices. Although the description herein focuses on two backbone segments, it is to be appreciated that the algorithm is generalizable to any number of segments. The method works very similarly to the original superposition method, except that scaffolds are scanned for structural similarity to each of the two epitope segments individually, and whenever a match is found to one of the segments, that scaffold is searched a second time for structural similarity to the other epitope segment, with the rigid body position of the second epitope segment relative to the scaffold pre-determined by the superposition of the first segment to the scaffold. “Double” matches are identified if (a) one epitope segment matches a scaffold with backbone rmsd/nsup<threshold 1 and the other segment matches with backbone rmsd/nsup<threshold 2, where threshold 1 is typically 0.15 and threshold 2 is typically 0.2, or (b) if both segments are superimposed simultaneously onto the scaffold and the backbone rmsd/nsup is <threshold 3, where threshold 3 is typically 0.2. As used herein, backbone rmsd refers to the root-mean square deviation of a structural alignment computed over the backbone atoms N, CA, C, O; and backbone rmsd/nsup refers to backbone rmsd divided by the number of aligned residues.


An automated search of monomeric and non-monomeric proteins without co-factors within the PDB (RCSB Protein Data Bank, Brookhaven, N.Y.) was conducted in March 2009 to identify candidate scaffolds having similar three-dimensional structures to the three-dimensional structure defined by the atomic coordinates specified in PDB acc code 3IXT. Nearly all of the structural matches contained significant backbone clashes with the motavizumab antibody (multiple atomic overlaps). Several hundred structural matches were screened by eye to identify structures that could be trimmed to eliminate backbone clash. Ten variants of 3 different scaffold proteins (1lp1b (Staphylococcus aureus Protein A), 1s2xa (Helicobacter pylori CagZ protein) and 2eiaa (equine infectious anemia virus) were chosen either because no trimming was necessary (1lp1b, 2eiaa) or the necessary trimming was restricted to a terminus and could be done easily (1s2xa). It is to be appreciated that this method can also identify scaffolds requiring more advanced trimming by flexible backbone protein design.


Residues required for elicitation of a humoral immune response against RSV were then implanted into the scaffolds and epitope conformation was stabilized by genetic engineering using a method similar to that described in WO 2008/025015 A2. For example, FIG. 7 shows a final scaffold comparison for immunogen 1lp1b003. Amino acids in bold are motavizumab contact residues. Amino acids that are underlined were substituted to stabilize the structure.


The amino acid sequences of ten scaffold-based immunogens are:









11p1b_001


(SEQ ID NO: 18)


VDNSFNDEKKLASNEIAHLPNLNEEQRSAFLSSINDDPSQSANLLAEA


KKLNDAQAPK





11p1b_002


(SEQ ID NO: 21)


VDNSFNDEKKLASNEIQHLPNLNEEQRSAFISSLNDDPSQSANLLAEA


KKLNDAQAPK





11p1b_003


(SEQ ID NO: 24)


SFNDEKKLASNEIAHLPNLNEEQRSAFLSSINDDPSQSANLLAEAKKL


NDAQAPK





11p1b_004


(SEQ ID NO: 149)


SFNDEKKLASNEIQHLPNLNEEQRSAFISSLNDDPSQSANLLAEAKKL


NDAQAPK





1s2xa_001


(SEQ ID NO: 152)


GSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQNYATSLKD


SNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASANRCSQELS


FANDTIKNNDTKKLFSNEIADNFNNFTADEVARISDLVASYLPREYLP


PFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTKY





1s2xa_002


(SEQ ID NO: 155)


GSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQNYATSLKD


SNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASANRASQELS


FINDTIKNNDTKKLFSNEAADNFNNFTADEVARISDLVASYLPREYLP


PFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTKY





1s2xa_003


(SEQ ID NO: 158)


GSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQNYATSLKD


SNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASANRASQELS


FANDTIKNNDTKKLFSNEIADNFNNFTADEVARISDLVASYLPREYLP


PFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTK





1s2xa_004


(SEQ ID NO: 164)


GSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQNYATSLKD


SNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASANRASQELS


FINDTIKNNDTKKLFSNEAADNFNNFTADEVARISDLVASYLPREYLP


PFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTK





2eiaa_001


(SEQ ID NO: 167)


APRGYTTWVNTIQTNGLLNEASQNLFGILSVDATSEEMNAFLDVVPGQ


AGQKQILLDAIDKIADDWDNRHPLPNAPLVAPPQGPIPMTARFIRGLG


VPRERQMEPAFDQFRQTYRQWIIEAMSEGIKVMIGKPKAQNIRQGAKE


PYPEFVSRLLSQINDEGHPNDIKKLRSNTLTIQNANEECRNAMRHLRP


SDTGAEKMYACRDIG





2eiaa_002


(SEQ ID NO: 170)


GKPKAQNIRQGAKEPYPEFVSRLLSQINDEGHPNDIKKLRSNTLTIQN


ANEECRNAMRHLRPSDTGAEKMYACRDIG






These immunogens have been further tailored to include amino (_N) or carboxyl (_C) tags or motifs to aid in purification of immunogens of the embodiments. Examples include of production of the following immunogens:









1sx2a_001_N_His


(SEQ ID NO: 177)


HHHHHHGSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQN


YATSLKDSNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASA


NRCSQELSFANDTIKNNDTKKLFSNEIADNFNNFTADEVARISDLVA


SYLPREYLPPFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTKY*





1sx2a_002_N_His


(SEQ ID NO: 178)


HHHHHHGSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQN


YATSLKDSNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASA


NRASQELSFINDTIKNNDTKKLFSNEAADNFNNFTADEVARISDLVA


SYLPREYLPPFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTKY*





1sx2a_003_N_His


(SEQ ID NO: 179)


HHHHHHGSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQN


YATSLKDSNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASA


NRASQELSFANDTIKNNDTKKLFSNEIADNFNNFTADEVARISDLVA


SYLPREYLPPFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTK*





1sx2a_004_N_His


(SEQ ID NO: 180)


HHHHHHGSPNSRVDELGFNEAERQKILDSNSSLMRNANEVRDKFIQN


YATSLKDSNDPQDFLRRVQELRINMQKNFISFDAYYNYLNNLVLASA


NRASQELSFINDTIKNNDTKKLFSNEAADNFNNFTADEVARISDLVA


SYLPREYLPPFIDGNMMGVAFQILGIDDFGKKLNEIVQDIGTK*





2eiaa_002_N_His


(SEQ ID NO: 181)


HHHHHHGKPKAQNIRQGAKEPYPEFVSRLLSQINDEGHPNDIKKLRS


NTLTIQNANEECRNAMRHLRPSDTGAEKMYACRDIG*






Each of the four 1lp1b-based immunogens, namely 1lp1b001, 1lp1b002, 1lp1b003 and 1lp1b004 was expressed with a HRV3C site, PADRE, Caspase3 site, 6×His tag and StrepTagII in 293F mammalian cells (Invitrogen) transformed with paH (also known as p(alpha)H) vector comprising a nucleic acid sequence encoding the respective immunogen. The paH vector is a modified version of the pHLSec vector (Aricescu A R et al, ibid) that includes changes to the multi-cloning site (MCS) and removal of certain restriction enzyme sites. The resultant immunogens were purified by nickel IMAC and STREP-TACTIN® chromatography followed by gel filtration. His and Strep tags were cleaved by pro-caspase. As an example, the following sequence is 1lp1b001 with a HRV3C site (LEVLFQGP (SEQ ID NO:182)), PADRE (AKFVAAWTLKAAA (SEQ ID NO:183)), caspase3 site (DEVD (SEQ ID NO:184), 6×His tag (HHHHHH (SEQ ID NO:185)) and StrepTagII (WSHPQFEK (SEQ ID NO:186)) (each underlined) at its carboxyl terminus:









VDNSFNDEKKLASNEIAHLPNLNEEQRSAFLSSINDDPSQSANLLAE





AKKLNDAQAPKLEVLFQGPGAKFVAAWTLKAAAGDEVDGSHHHHHHS






AWSHPQFEK,



also denoted herein as_SEQ ID NO: 187.






Each of three of the four 1s2xa-based immunogens, namely 1s2xa001, 1s2xa002, and 1s2xa003, was expressed as a maltose binding protein fusion in BL21(DE3) bacteria transformed with MBP-HTSHP vector comprising a nucleic acid sequence encoding the respective immunogen. The MBP-HTSHP vector is a modified version of the pMal-c2x vector (New England Biolabs, Ipswich, Mass.) that includes a linker region with all the various tags and restriction sites. Fusion protein was recovered by nickel chromatography. The fusion protein was cleaved with pro-caspase 3 and subjected to nickel chromatography and S75 gel filtration. An anion exchange column can also be used as part of the procedure. As an example, the following sequence in bold is 1s2xa001 with maltose-binding protein, a factor Xa site, his-tag, TEV site, strep-tag, his-tag, HRV3C site and caspase 3 site (each underlined) at its amino terminus:










MKIEEGKLVIWINGDKGYNGLAEVGKKFEKDTGIKVTVEHPDKLEEK







FPQVAATGDGPDIIFWAHDRFGGYAQSGLLAEITPDKAFQDKLYPFT







WDAVRYNGKLIAYPIAVEALSLIYNKDLLPNPPKTWEEIPALDKELK







AKGKSALMFNLQEPYFTWPLIAADGGYAFKYENGKYDIKDVGVDNAG







AKAGLTFLVDLIKNKHMNADTDYSIAEAAFNKGETAMTINGPWAWSN







IDTSKVNYGVTVLPTFKGQPSKPFVGVLSAGINAASPNKELAKEFLE







NYLLTDEGLEAVNKDKPLGAVALKSYEEELAKDPRIAATMENAQKGE







IMPNIPQMSAFWYAVRTAVINAASGRQTVDEALKDAQTNSSSNNNNN






NNNNNLGIEGRISGHHHHHHHHDYDIPSSENLYFQGSASWSHPQFEK





SGHHHHHHHHDYDIPSSLEVLFQGPGSDEVDGSPNSRVDELGFNEAE






RQKILDSNSSLMRNANEVRDKFIQNYATSLKDSNDPQDFLRRVQELR







INMQKNFISFDAYYNYLNNLVLASANRCSQELSFANDTIKNNDTKKL







FSNEIADNFNNFTADEVARISDLVASYLPREYLPPFIDGNMMGVAFQ







ILGIDDFGKKLNEIVQDIGTKY,



also denoted herein as SEQ ID NO: 151.






Each immunogen of the present invention can be produced in the manner described herein.


Scaffold-based immunogens 1lp1b001, 1lp1b002, 1lp1b003, 1lp1b004, 1s2xa001, 1s2xa002 and 1s2xa003 were submitted to surface plasmon resonance binding analysis. Experiments were carried out on a Biacore 3000 instrument (GE Healthcare). Motavizumab fragment of antigen binding (Fab) was covalently coupled to a CM5 chip, and a blank surface with no antigen was created under identical coupling conditions for use as a reference. Scaffolds were serially diluted 2-fold, starting at 10 mM, into 10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA and 0.005% polysorbate 20 and injected over the immobilized motavizumab Fab and reference cell at 40 ml/min. The surface was regenerated with 10 mM glycine pH 3 at a flow rate of 40 ml/min. The data were processed with SCRUBBER-2 and double referenced by subtraction of the blank surface and a blank injection (no analyte).


Results are shown in Table 5, which indicates the Kd in nanomolar (nM) concentrations for each of the tested scaffold-based immunogens.









TABLE 5







Affinity of scaffold-based immunogens for motavizumab










Scaffold
Motavizumab affinity (Kd in nM)














1lp1b_003
66



1lp1b_001
181



1lp1b_004
296



1lp1b_002
700



1s2xa_003
3005



1s2xa_002
3638



1s2xa_001
4755










Immunogen 1lp1b003 exhibited the best affinity for motavizumab in this assay. This immunogen was also tested for binding to an irrelevant IgG and irrelevant Fab. No significant binding above baseline was detected.


Immunogen 1lp1b003 proteins, produced in both mammalian cells and bacteria as described above, were also tested for their three-dimensional structure. The data in FIG. 8 demonstrate that both mammalian- and bacterial-produced 1lp1b003 proteins are alpha helical in solution and have a melting temperature of about 60° C., which confirms that the proteins are folded in solution.


Example 4
Production of Resurfaced Scaffold-Based Immunogens

This example describes the production of scaffold-based immunogens that have been resurfaced to neutralize deleterious immunodominant epitopes.


Resurfaced variants of the 1lp1b003 scaffold were designed using protocols similar to those described in WO 2009/100376 A2. This scaffold has the highest binding affinity of the initial designs. In these resurfaced variants, a significant fraction of the non-epitope surface area on the scaffold was mutated to generate antigenic surfaces distinct from the original design. In a few cases the mutations were designed to introduce N-linked glycosylation sites, but in most cases the mutations did not. These variants are useful in heterologous prime-boost immunizations using a non-resurfaced immunogen as the prime and one or more of the resurfaced variants as a boost, with the aim of focusing the antibody response onto the antibody binding domain, or epitope, which is intended to be the only antigenic surface conserved between the non-resurfaced and resurfaced immunogens.


The sequences for several resurfaced 1lp1b immunogens are below. The first two immunogens include substitutions resulting in glycan-masked immunogens; the remaining are non-glycan resurfaced immunogens.











mota_11p1b.m1.cl.d1_glyc1



(SEQ ID NO: 55)



SFNDEKKLASNEIAHLPNLNETQRSAFLSSINDDPSQSANLLANAT



KLNDAQAP







mota_11p1b.m1.cl.d1_glyc2



(SEQ ID NO: 56)



SFNDEKKLASNEIAHLPNLNETQRSAFLSSINDDPNQSANLLANAT



KLNDAQAP







mota_11p1b.m1.c1.d1_des1_1



(SEQ ID NO: 66)



SFNDEKKLASNRIANLPNLNEEQRSAFLSKINDDPSQSANLLEEAL



KLNDAQAQK







mota_11p1b.m1.c1.d1_des1_2



(SEQ ID NO: 67)



SFNDDKKLASNRIANLPNLNEEQRSAFLSKINDDPSQSRNLLEEAL



KLNDAQAQK







mota_11p1b.m1.c1.d1_des1_3



(SEQ ID NO: 68)



SFNDKKKLASNRIANLPNLNEEQRSAFLSKINDDPSKSEELLEKAL



KLNDAQAQK







mota_11p1b.m1.c1.d1_des1_5



(SEQ ID NO: 69)



SFNDKKKLASNEIANLPNLNEEQRSAFLSKINDDPSKSEELLEEAL



KLNDAQADK







mota_11p1b.m1.c1.d1_des1_6



(SEQ ID NO: 70)



SFNDKKKLASNRIAKLPNLNEKQRSAFLSKINDDPSKSEELLKKAL



KLNKAQAKK







mota_11p1b.m1.c1.d1_des1_7



(SEQ ID NO: 71)



SFNDEKKLASNRIANLPNLNQEQRSAFLSKINDDPSQSANLLEEAL



KLNDNQAQK







mota_11p1b.m1.c1.d1_des1_8



(SEQ ID NO: 72)



SFNDDKKLASNRIANLPNLNQEQRSAFLSKINDDPSQSRNLLEEAL



KLNDNQAQK







mota_11p1b.m1.c1.d1_des1_9



(SEQ ID NO: 73)



SFNDKKKLASNRIANLPNLNQEQRSAFLSKINDDPSKSEELLEKAL



KLNDNQAQK







mota_11p1b.m1.c1.d1_des1_10



(SEQ ID NO: 74)



SFNDKKKLASNEIANLPNLNQEQRSAFLSKINDDPSKSEELLEEAL



KLNDNQADK







mota_11p1b.m1.c1.d1_des1_11



(SEQ ID NO: 75)



SFNDKKKLASNRIAKLPNLNEKQRSAFLSNINDDPSKSEELLEKAL



KLNQAQAQK






Production of these resurfaced immunogens can occur using methods similar to those described in Example 3.


Example 5
Production of Trimeric F Immunogens

The production of RSV Fo Fd immunogen was described in Example 1. Additional soluble trimeric F immunogens, stabilized in the pre-fusion conformation, namely RSV F Fd and RSV F0 Fd GAG, have been produced in a similar manner. The amino acid sequence of each of these immunogens, with the T4 fibritin trimerization domain (also referred to as Foldon, or Fd) underlined, is as follows:









RSV F0 Fd


(SEQ ID NO: 174)


MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSA


LRTGWYTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQ


LLMQSTPATNNQARSELPRFMNYTLNNAKKTNVTLSKKRKSSFLGFL


LGVGSAIASGVAVSKVLHLEGEVNKIKSALLSTNKAVVSLSNGVSVL


TSKVLDLKNYIDKQLLPIVNKQSCSISNIETVIEFQQKNNRLLEITR


EFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVR


QQSYSIMSIIKEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEG


SNICLTRTDRGWYCDNAGSVSFFPQAETCKVQSNRVFCDTMNSLTLP


SEVNLCNVDIFNPKYDCKIMTSKTDVSSSVITSLGAIVSCYGKTKCT


ASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQEGKSLYVK


GEPIINFYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELLSAIG



GYIPEAPRDGQAYVRKDGEWVLLSTFLGGIEGRHHHHHH-






RSV F Fd


(SEQ ID NO: 175)


MELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGYLSA


LRTGWYTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVTELQ


LLMQSTPATNNRARRELPRFMNYTLNNAKKTNVTLSKKRKRRFLGFL


LGVGSAIASGVAVSKVLHLEGEVNKIKSALLSTNKAVVSLSNGVSVL


TSKVLDLKNYIDKQLLPIVNKQSCSISNIETVIEFQQKNNRLLEITR


EFSVNAGVTTPVSTYMLTNSELLSLINDMPITNDQKKLMSNNVQIVR


QQSYSIMSIIKEEVLAYVVQLPLYGVIDTPCWKLHTSPLCTTNTKEG


SNICLTRTDRGWYCDNAGSVSFFPQAETCKVQSNRVFCDTMNSLTLP


SEVNLCNVDIFNPKYDCKIMTSKTDVSSSVITSLGAIVSCYGKTKCT


ASNKNRGIIKTFSNGCDYVSNKGVDTVSVGNTLYYVNKQEGKSLYVK


GEPIINFYDPLVFPSDEFDASISQVNEKINQSLAFIRKSDELLSAIG



GYIPEAPRDGQAYVRKDGEWVLLSTFLGGLVPRGSHHHHHHSAWSHP



QFEK--





RSV F0 Fd GAG


(SEQ ID NO: 176)


GAGMELLILKANAITTILTAVTFCFASGQNITEEFYQSTCSAVSKGY


LSALRTGWYTSVITIELSNIKENKCNGTDAKVKLIKQELDKYKNAVT


ELQLLMQSTPATNNRARGAGKRRFLGFLLGVGSAIASGVAVSKVLHL


EGEVNKIKSALLSTNKAVVSLSNGVSVLTSKVLDLKNYIDKQLLPIV


NKQSCSISNIETVIEFQQKNNRLLEITREFSVNAGVTTPVSTYMLTN


SELLSLINDMPITNDQKKLMSNNVQIVRQQSYSIMSIIKEEVLAYVV


QLPLYGVIDTPCWKLHTSPLCTTNTKEGSNICLTRTDRGWYCDNAGS


VSFFPQAETCKVQSNRVFCDTMNSLTLPSEVNLCNVDIFNPKYDCKI


MTSKTDVSSSVITSLGAIVSCYGKTKCTASNKNRGIIKTFSNGCDYV


SNKGVDTVSVGNTLYYVNKQEGKSLYVKGEPIINFYDPLVFPSDEFD


ASISQVNEKINQSLAFIRKSDELLSAIGGYIPEAPRDGQAYVRKDGE



WVLLSTFLGGLVPRGSHHHHHHSAWSHPQFEK--







Gel filtration analysis of RSV Fo Fd immunogen indicates that this immunogen has a calculated molecular weight of about 240 kilodaltons (kD), indicative of a trimer (FIG. 2e). Binding studies indicate that RSV Fo Fd immunogen binds motavizumab, SYNAGIS® and 101F with affinities similar to RSV F protein. RSV Fo Fd immunogen can bind motavizumab and 101F simultaneously.


Example 6
Design of Antibodies Having Higher Affinity Binding for the Motavizumab-Binding Domain

This Example describes a method to design antibodies having a higher binding affinity for the motavizumab-binding domain. Such antibodies can have utility as passive immunotherapy compositions.


Additional interactions were identified between the motavizumab antibody and its epitope using a modified Rosetta interface design protocol. The crystal structure of the complex between motavizumab and its binding domain was prepacked to remove clashes as calculated by Rosetta. The iterations of local refine docking between epitope and antibody were carried out to generate an ensemble of slightly different rigid body orientations, then the backrub algorithm was applied to both epitope and antibody to generate backbone conformational variation, and finally iterative design and minimization of sidechains in the interface was carried out. This protocol was carried out hundreds of thousands of times; 20,000 low-energy structures were generated, and the top 1% were selected based on calculated ddG. From those 200 structures, approximately 50 were selected based on the total calculated free energy. The top 50 structures were aligned and the mutations listed below were identified as ones that could enhance binding between the antibody and peptide. Several candidate mutations are listed at many of the positions. A DNA library encoding all of these mutations within a single-chain fragment variable (ScFv) construct for motavizumab will be built and screened using yeast surface display to identify the tightest binding clones.


The mutations identified that may increase binding affinity are as follows:
















Motavizumab heavy (H) chain residue
Encoded mutation









 32
H or E



 35
A



 52
K, H, T, S or R



 53
H or S



 54
F or R



 56
I, S, E or D



 58
Y or W



 97
D, H or R



 99
D



100
Y, W or H



100A
S or T





















Motavizumab light (L) chain residue
Encoded mutation







32
F


49
H or R


92
K


94
H


96
H









It was also recognized that additional contacts between the antibody and epitope could be made by increasing the length of the CDRH2 loop by 2 residues. The CDRH2 loop in the heavy chain of motavizumab spans amino acids H50 through H58. The above protocol was used for these simulations as well but it included an additional step in which the loop was explicitly rebuilt to increase the length prior to the iterations of docking, backrub and design. Following is the library of mutations for that lengthened loop. This library is merged with the library above and screened on yeast as described above.


Specifically, the 9-residue stretch between H50-H58 was removed and replaced with a variation of the 11-residue sequence below, were 1 is the first amino acid in that sequence:













Position
Encoded mutation
















1
E, S or M


2
I


3
H, R or F


4
S


5
G


6
G, H, K, L, N, Q, S, D, T or R


7
F, K, S, T, D or R


8
E, N or D


9
D, H, L, S, R or T


10
Y


11
Y, F or H.









Example 7
Binding of Immunogens to Motavizumab

This Example demonstrates the ability of immunogens of the present disclosure to bind to motavizumab.


a. Expression protocol for 1lp1b-based immunogens: Mammalian codon-optimized genes encoding 1lp1b-based immunogens were synthesized by GeneArt with an N-terminal secretion signal (MGSLQPLATLYLLGMLVASVLA (SEQ ID NO:188)) and a C-terminal HRV3C cleavage site, PADRE epitope (AKFVAAWTLKAAA (SEQ ID NO:183)), Caspase-3 cleavage site, 6×His-tag and Strep-tag II. The genes were cloned into the mammalian expression vector paH. Proteins were expressed from the plasmids by transient transfection using the FREESTYLE™ 293 expression system (Invitrogen). 1lp1b proteins were purified from the media using Ni2+-NTA resin (Qiagen) and then STREP-TACTIN® resin (IBA, Goettingen, Germany) as per manufacturer's instructions, followed by passage over a 16/60 Superdex 75 column (GE Healthcare). For SPR, ITC and CD measurements, all tags were retained. For immunization experiments, Pro-caspase 3 was added to remove the 6×His-tag and Strep-tag II. The tags and protease were removed from cleaved 1lp1b by passage over Ni2+-NTA resin.


b. Expression protocol for 1s2xa-based immunogens: E. coli codon-optimized genes encoding 1s2xa-based immunogens were synthesized by GeneArt and cloned into a custom vector based on pMAL-c2X (New England Biolabs). The expression vectors were transformed into BL21(DE3) cells, and the cells were grown in Terrific Broth (Difco, Becton Dickinson, Franklin Lakes, N.J.) at 37° C. until OD600=2.0. The temperature was then reduced to 22° C., and isopropyl f3-D-thiogalactoside (IPTG) was added to 1 mM. After overnight incubation at 22° C., the cells were harvested and lysed with BUGBUSTER™ Protein Extraction Reagent (Novagen, EMD Chemicals, Gibbstown, N.J.), and 1s2xa proteins were purified using Ni2+-NTA resin (Qiagen). Fusion tags were removed by incubation with Pro-caspase 3 and passage over Ni2+-NTA resin. 1s2xa proteins were further purified by passage over a 16/60 SUPERDEX™ 75 column (GE Healthcare), and anion exchange chromatography using a MonoQ column (GE Healthcare).


c. Production of ferritin-containing immunogens. The gene encoding 1lp1b003 fused to the N-terminus of the coding region of human ferritin was subcloned into a mammalian expression vector, such a pVRC8405 (Barourch D et al., 2005, J. Virol. 79, 8828-8834). Proteins were expressed from this plasmid by transient transfection in HEK293 GnTI−/− cells. 1lp1b003 ferritin was initially purified from the media by anion exchange chromatography using a MonoQ HR 10/10 column (GE Healthcare). The eluted protein was then passed over a column consisting of SYNAGIS® IgG covalently coupled to Protein A agarose resin (Pierce). The column was washed with phosphate-buffered saline (PBS) and eluted with Actisep Elution Medium (Sterogene, Carlsbad, Calif.). The eluted 1lp1b003 ferritin was dialyzed against PBS, concentrated, and further purified by passage over a 16/70 SUPEROSE™6 column (GE Healthcare). Additional ferritin-containing immunogens, such as 1lp1b003_eumS, 1lp1b003_eumSP, 1lp1b003eumL, and 1lp1b003_eumLP, were produced in a similar manner.


Immunogens were produced as described herein and submitted to surface plasmon resonance binding analysis as described in Example 3. Some of the immunogens were also submitted to isothermal titration calorimetry (ITC). Table 8 indicates which immunogens were tested and the results obtained. Kd refers to the Kd of motavizumab for a tested immunogen by either Biacore or ITC measurement, respectively. dH and -TdS relate to measurements of enthalpy and entropy, respectively. Table 8 also indicates the cell type (i.e., HEK293 mammalian cells or E. coli bacteria) used to produce the respective immunogen. Table 8 also provides the name of the immunogen used in Table 9. A blank square indicates that the respective immunogen was not tested in that assay. “nd” means not detectable; i.e., below the limit of detection of the assay.









TABLE 8







Affinity of immunogens for motavizumab and ITC measurements













Immunogen





Name used in
production
Biacore
ITC













Name
Table 9
system
Kd (nM)
dH
−TdS
Kd (nM)
















1lp1b_001

HEK293
181





1lp1b_002

HEK293
700


1lp1b_003
1lp1b
HEK293
66
1.1
−10.5
126


1lp1b_003
1lp1b(Ecoli)

E. coli



1lp1b_004

HEK293
296


1lp1b_003_K272E

HEK293
nd


1l1pb_003_Glyc1
1lp1b_Glyc1
HEK293
400


1lp1b_003_Glyc2
1lp1b_Glyc2
HEK293
812


1l1pb_003_Surf1
1lp1b_Surf1
HEK293
120


1l1pb_003_Surf8
1lp1b_Surf8
HEK293
117


1s2xa_001


E. coli

4755


1s2xa_002


E. coli

3638


1s2xa_003
1s2xa or Cag-Z

E. coli

3005
−19.2
9.9
147


1s2xa_004


E. coli

3950
−19.6
10.7
275


2eiaa_001

HEK293
nd


1lp1b_003_K46A
K46A
HEK293
148
−5.3
−5.1
21


1lp1b_003_Q52A
Q52A
HEK293
233
−1.8
−8.8
16


1lp1b_003_I13L_F27A
I13L F27A
HEK293
541
−19.4
10.1
159


1lp1b-003_L41I_L42A
L41I L42A
HEK293
247
−1.3
−9.0
24


1lp1b_003_L41I_L42V
L41I L42V
HEK293
77
−1.7
−7.9
90


1lp1b_003_I13A_L42A
I13A L42A
HEK293
>10,000
−25.6
18.8
10,700


1lp1b_003_L19A
L19A
HEK293
>10,000
−27.3
20.9
21,000


1lp1b_003_L19A_L41I
L19A L41I
HEK293
>10,000


1lp1b_003_I13A
I13A
HEK293
2300
−16.2
7.4
380


1lp1b_003_L16A
L16A
HEK293
2300
−12.3
3.4
341


1lp1b_003_F27A
F27A
HEK293
490
−12.4
2.7
83


1lp1b_003_L41A
L41A
HEK293
310
−4.0
−5.7
79


1lp1b_003_L42A
L42A
HEK293
218
−4.4
−5.1
93


1lp1b_003_ferritin
1lp1b_ferritin
HEK293


1lp1b_003_eumS
eumS
HEK293


1lp1b_003_eumSP
eumSP
HEK293


1lp1b_003_eumL
eumL
HEK293


1lp1b_003_eumLP
eumLP
HEK293


1lp1b_003_Neg1
Neg1
HEK293









The results indicate that a number of the immunogens bind motavizumab with a Kd within approximately an order of magnitude of the Kd of RSV F peptide for motavizumab (Kd of ˜250 nM). Immunogen 1lp1b003_K272E has a mutation resulting in removal of a key contact residue within the motavizumab-binding domain; as such, that immunogen would not be expected to bind to motavizumab.


Example 8
Immunogenicity Data: Binding and Neutralization

This Example demonstrates the ability of immunogens of the embodiments to elicit a humoral immune response that yields immune sera capable of binding to F protein, F peptide (having SEQ ID NO:2), and scaffold immunogens. The Example further demonstrates the ability of immunogens of the embodiments to elicit a neutralizing humoral immune response against RSV.


Mice were immunized as follows: Six- to eight-week old female C57BL/6 mice (Jackson Laboratory, Bar Harbor, Me.) were used for all experiments. Mice were immunized with immunogen and 25 μg CpG/mouse intramuscularly. Table 9 indicates which immunogens were tested, as well as the immunogen doses, administration regimen (including identification of immunogen administered as a prime (dose 1), identification of immunogen(s) administered as a boost(s), and number of boosts (i.e., dose 2, dose 3, etc.), interval of time between doses, and number of mice used. Please note that the immunogen names in Table 9 are an abbreviated form of immunogens listed in Table 8; Table 8 provides both names. Table 9 also indicates if the immunogens did not include a PADRE motif (-PADRE). Sera were collected on day 10 or day 14 and tested as described herein.


The abilities of the murine immune sera to bind 1lp1b003 immunogen (labeled as 1lp1b in Table 9), 1lp1b(K272E) protein (the mutation causing loss of motavizumab binding, and labeled as 1lp1b(K272E) in Table 9), RSV F peptide, and RSV F protein were tested using a kinetic ELISA as follows: The RSV F protein and scaffold proteins were diluted in PBS and coated onto 96-well flat bottom ELISA plates at a concentration of 1 μg/ml and incubated overnight at 4° C. For RSV F peptide binding, a biotinylated RSV F peptide (biotin-peptide) was used; the biotin-peptide was coated onto a Neutravidin plate (Thermo Scientific, Rockford, Ill.) and incubated for 2 hours at room temperature. For all samples, nonspecific adsorption was prevented with 200 μL/well of blocking buffer (2% BSA in PBS) for 1 hour at 37° C. Plates were then washed four times on an automated plate washer (Bio-Tek Instruments, Winooski, Vt.) with wash buffer (0.02% Tween-20 in PBS). One hundred μL of diluted test sera (1:100 in blocking buffer) or positive serum control were added to each well. Plates were incubated for one hour at room temperature, washed four times, and incubated for 1 hour at room temperature with HRP-conjugated goat anti-mouse IgG antibody (Jackson ImmunoResearch Laboratories, West Grove, Pa.). Plates were washed with wash buffer four times followed by distilled water. One hundred μl of Super AquaBlue ELISA substrate (eBioscience, San Diego, Calif.) was added to each well, and plates were read immediately using a Dynex Technologies microplate reader (Chantilly, Va.). The rate of color change in mOD/min was read at a wavelength of 405 nm every 9 seconds for 5 minutes with the plates shaken before each measurement. The mean mOD/min reading of duplicate wells was calculated, and the background mOD/min was subtracted from the corresponding control well.


Neutralization activity was measured using a flow cytometric neutralization assay as described in the Examples herein.


The abilities of immune sera elicited by immunogens of the embodiments to bind to RSV F protein and RSV F peptide are indicated in Table 9. Table 9 also provides data from the neutralization assay, expressed as number of mice (between 0 and 5) showing a specified result (Frequency) and reciprocal dilution of immune sera at which EC50 was achieved (Magnitude). For certain immunogens, the immune sera, while not achieving 50% neutralization under the stipulated conditions, did show lower amounts of neutralization. Those results are shown in the last column (Comments). These data are expressed as % of neutralization at a specified dilution of sera. For example, “40% neutralization in 1:10” means that all 5 mice showed 40% neutralization when the respective immune sera were diluted by a factor of 10. “3/5 20% neutralization in 1:10” means that 3 of the 5 mice showed 20% neutralization when the respective immune sera were diluted by a factor of 10. A blank square indicates that the respective immunogen was not tested in that assay. “nd” means not detectable; i.e., below the limit of detection of the assay.









TABLE 9





Immunogenicity of immune sera elicited by immunogens






























Neutral-




Immuno-





ization



gen


1lp1b
RSV F
RSV F
assay



Dose

1lp1b
(K272E)
Peptide
protein
(flow



(immuno-

binding
binding
binding
binding
cytometry)





















gen +





(kinetic
(kinetic
(kinetic
(kinetic
Fre-
Mag-




25 μg





ELISA)
ELISA)
ELISA)
ELISA)
quen-
ni-


Trial
CpG)
dose 1
dose 2
dose 3
Interval
N
mean
mean
mean
mean
cy
tude
Comments





2
10 μg
1lp1b
1lp1b

3 weeks
5
75.8
67.4

nd
nd
nd



10 μg
1lp1b(−)
1lp1b(−)


5
2.8
2.3

0.2
nd
nd




PADRE
PADRE



10 μg
1s2xa
1s2xa


5
0.2
0.8

1.2
nd
nd




(E. coli)
(E. coli)



10 μg
1lp1b
1lp1b


5
0.4
0.2

0.4
nd
nd




(E. coli)
(E. coli)



10 μg
1lp1b
1lp1b
1lp1b
4 weeks
5
113
88

nd
nd
nd



10 μg
1lp1b
1lp1b
1lp1b

5
3.8
4.2

nd
nd
nd



10 μg
1s2xa
1s2xa
1s2xa

5
0.2
0.4

nd
nd
nd




(E. coli)
(E. coli)
(E. coli)



10 μg
1lp1b
1lp1b
1lp1b

5
1
nd

2.21
nd
nd




(E. coli)
(E. coli)
(E. coli)


3
20 μg
1lp1b_Ferritin
1lp1b_Ferritin
1lp1b_Ferritin
2 weeks
5
12
15.33


nd
nd



20 μg
1lp1b_Glyc 1
1lp1b_Glyc 1
1lp1b_Glyc 1

5
3.6
2


nd
nd



20 μg
1lp1b_surf 1
1lp1b_Glyc 1
1lp1b_surf 1

5
0.6
1


nd
nd



20 μg
1lp1b_surf 8
1lp1b_Glyc 1
1lp1b_surf 8

5
0.8
0.4


nd
nd


4
20 μg
1lp1b
1lp1b

2 weeks
5


1.8
2.2
nd
nd



20 μg
L41A
L41A


5


0.4
1.6
nd
nd



20 μg
L42A
L42A


5


0  
2.6
nd
nd



20 μg
Glyc 2
Glyc 2


5


2.6
0.8
nd
nd



20 μg
113L F27A
113L F27A


5


0.4
1.4
nd
nd



20 μg
113L F27A
L42A


5


0.4
1
nd
nd
Additional















Neutral-















ization















Results:



20 μg
1lp1b
1lp1b
1lp1b-
2 weeks
5
18.8
19
0.2
1.8
2
15-20
40%






Ferritin








neutral-















ization















in 1:10



20 μg
L41A
L41A
1lp1b-

5
15.2
15.6
0.2
1.2
2
10-17
40%






Ferritin








neutral-















ization















in 1:10



20 μg
L42A
L42A
1lp1b-

5
12.8
10.2
nd
2.2
2
 15
40%






Ferritin








neutral-















ization















in 1:10



20 μg
Glyc 2
Glyc 2
1lp1b-

5
1.8
1.8
0.6
0.4
2
10-13
40%






Ferritin








neutral-















ization















in 1:10



20 μg
113L F27A
113L F27A
1lp1b-

5
6.2
5.4
nd
0.6
2
 10
40%






Ferritin








neutral-















ization















in 1:10



20 μg
113L F27A
L42A
Glyc 2

5
11.4
11.4
0.4
0.8
3
 9-15
40%















neutral-















ization















in 1:10


5
20 μg
k46A
k46A
1lp1b_Ferritin
2 weeks
5
14.8
6.6
nd
1.75
4
10-20



20 μg
q52A
q52A
1lp1b-

5
7.8
7.2
nd
3.6
4
11-20






Ferritin



20 μg
L141L/L42V
L141L/L42V
1lp1b-

5
19
12.8
nd
2.75
3
10-16






Ferritin



20 μg
eumS
eumS
1lp1b-

5
12
11.6
nd
2.2
3
10-24






Ferritin



20 μg
eumSP
eumSP
1lp1b-

5
25.4
22.8
nd
3
3
10-20






Ferritin



20 μg
eumL
eumL
1lp1b-

5
15.4
14.8
0.4
3.4
nd
~10
50%






Ferritin








neutral-















ization















in 1:10



20 μg
eumLP
eumLP
1lp1b-

5
13.5
15
0.2
2.8
4
13-27






Ferritin


6
20 μg
L16A
L16A
L16A
2 weeks
5
13
15.4
0.6
1.4
nd
nd
3/5 20%















neutral-















ization















in 1:10



20 μg
Neg 1
Neg 1
Neg 1

5
10.6
14.2
0.2
3
nd
nd
3/4 50%















neutral-















ization















in 1:10



20 μg
113A
113A
113A

5
1.8
2
0.4
1.2
nd
nd
3/5 35%















neutral-















ization















in 1:10



20 μg
F27A
F27A
F27A

5
3
2.2
0.2
2.8
nd
nd
3/5 25%















neutral-















ization















in 1:10





























1lp1b
RSV F
RSV F










1lp1b
(K272E)
Peptide
protein









binding
binding
binding
binding


PROSCI






(kinetic
(kinetic
(kinetic
(kinetic
Neutralization assay


Trial
Vaccination
dose 1, 2
dose 3, 4
dose 5, 6
Interval
N
ELISA)
ELISA)
ELISA)
ELISA)
(flow cytometry)






















1139
20 μg
2X 1lp1b
2X 1lp1b
2X 1lp1b_Ferritin
2 weeks
5


nd
4.6
nd
nd


1140
scaffold +
2X 1lp1b
2X 1s2xa
2X 1lp1b_Ferritin

5


4.25
4.75
nd
nd



50 μg CpG-

(CagZ)


1141
25 μl in 2%
2X 1lp1b
2X


5


nd
59
nd
nd



alum

1lpb1_surf1


1142

2X 1s2xa
2X 1s2xa


5


0.4
1.4
nd
nd




(CagZ)
(CagZ)


1143

2X 1s2xa
2X 1lp1b


5


0.2
3.8
nd
nd




(CagZ)


1144

2X pepide
2X 1lp1b


5


49.8
9.4
nd
nd









The results in Table 9 indicate that immune sera elicited by immunogens of the embodiments demonstrated binding to 1lp1b003 (referred to in Table 9 as 1lp1b) under the conditions tested. Some binding was also observed to 1lp1b003_K272E under the conditions tested. A number of the immune sera also bound to RSV F peptide or RSV F protein under the conditions tested. Interestingly, immune sera from all 5 mice immunized twice with 1lp1b003 followed by two boosts with 1lp1b003_Surf1 (trial 1141) showed high binding to RSV F protein. Some of the immunogens also elicited a neutralizing humoral immune response. Interestingly, boosting with a multivalent immunogen comprising ferritin stimulated a neutralizing humoral immune response. It is to be appreciated that while a number of immunogen immunizations led to undetectable neutralization, those results do not preclude other prime and/or boost conditions being found that could enable neutralization with such immunogens.


While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims.

Claims
  • 1. An RSV immunogen comprising an amino acid sequence of 1LP1_b (SEQ ID NO: 11) having from one to twenty amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of: (a) substitution of a serine at amino acid position N-25 in SEQ ID NO:11;(b) substitution of a leucine at amino acid position I-28 in SEQ ID NO:11;(c) substitution of a serine at amino acid position Q-29 in SEQ ID NO: 11;(d) substitution of an isoleucine at amino acid position L-31 in SEQ ID NO:11;(e) substitution of an asparagine at amino acid position K-32 in SEQ ID NO:11;(f) substitution of an aspartic acid at amino acid position K-4 in SEQ ID NO:11;(g) substitution of a lysine at amino acid position Q-6 in SEQ ID NO:11;(h) substitution of a lysine at amino acid position Q-7 in SEQ ID NO:11;(i) substitution of a leucine at amino acid position N-8 in SEQ ID NO:11;(j) substitution of a serine at amino acid position F-10 in SEQ ID NO:11; and(k) substitution of an asparagine at amino acid position Y-11 in SEQ ID NO:11.
  • 2. (canceled)
  • 3. The RSV immunogen of claim 1, wherein said RSV immunogen comprises an amino acid sequence that is at least 95% identical to an amino acid sequence of a protein selected from the group consisting of 1lp1b—001 (SEQ ID NO:18), 1lp1b—002 (SEQ ID NO:21), 1lp1b—003 (SEQ ID NO:24), and 1lp1b—004 (SEQ ID NO:149).
  • 4. The RSV immunogen of claim 1, wherein said RSV immunogen comprises an amino acid sequence of a protein selected from the group consisting of 1lp1b—001 (SEQ ID NO:18), 1lp1b—002 (SEQ ID NO:21), 1lp1b—003 (SEQ ID NO:24), 1lp1b—004 (SEQ ID NO:149), mota—1lp1b.m1.c1.d1_glyc1 (SEQ ID NO:55), mota—1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56), 1lp1b—003_Glyc1 (SEQ ID NO:39), 1lp1b—003_Glyc2 (SEQ ID NO:42), 1lp1b—003_Glyc3 (SEQ ID NO:45), 1lp1b—003_Glyc4 (SEQ ID NO:48), 1lp1b—003_Glyc5 (SEQ ID NO:51), 1lp1b—003_Glyc6 (SEQ ID NO:54), mota—1lp1b.m1.c1.d1_des1—1 (SEQ ID NO:66), mota—1lp1b.m1.c1.d1_des1—2 (SEQ ID NO:67), mota—1lp1b.m1.c1.d1_des1—3 (SEQ ID NO:68 mota—1lp1b.m1.c1.d1_des1—5 (SEQ ID NO:69), mota—1lp1b.m1.c1.d1_des1—6 (SEQ ID NO:70), mota—1lp1b.m1.c1.d1_des1—7 (SEQ ID NO:71), mota—1lp1b.m1.c1.d1_des1—8 (SEQ ID NO:72), mota—1lp1b.m1.c1.d1_des1—9 (SEQ ID NO:73), mota—1lp1b.m1.c1.d1_des1—10 (SEQ ID NO:74), mota—1lp1b.m1.c1.d1_des1—11 (SEQ ID NO:75), 1lp1b—003_Surf1 (SEQ ID NO:59), 1lp1b—003_Surf6 (SEQ ID NO:62), 1lp1b—003_Surf8 (SEQ ID NO:65), 1lp1b—003_ferritin (SEQ ID NO:138), 1lp1b—003_eumS (SEQ ID NO:140), 1lp1b—003_eumSP (SEQ ID NO:142), 1lp1b—003_eumL (SEQ ID NO:144), 1lp1b—003_eumLP (SEQ ID NO:146), 1lp1b—003_K46A (SEQ ID NO:78), 1lp1b—003_Q52A (SEQ ID NO:81), 1lp1b—003_I13L_F27A (SEQ ID NO:87), 1lp1b—003_L41I_L42V (SEQ ID NO:90), 1lp1b—003_L41I_L42A (SEQ ID NO:93), lp1b—003_I13A (SEQ ID NO:105), 1lp1b—003_L16A (SEQ ID NO:108), 1lp1b—003_F27A (SEQ ID NO:111), 1lp1b—003_L41A (SEQ ID NO:114), 1lp1b—003_L42A (SEQ ID NO:117), and 1lp1b—003_Neg1 (SEQ ID NO:132).
  • 5-10. (canceled)
  • 11. An RSV immunogen comprising an amino acid sequence of truncated 1S2X_a (SEQ ID NO:13) having from one to twenty-five amino acid substitutions, wherein at least one amino acid substitution is selected from the group consisting of: (a) substitution of a serine at amino acid position 92 in SEQ ID NO:13;(b) substitution of a leucine at amino acid position 95 in SEQ ID NO:13;(c) substitution of a serine at amino acid position 96 in SEQ ID NO:13;(d) substitution of an isoleucine at amino acid position 98 in SEQ ID NO:13;(e) substitution of an asparagine at amino acid position 99 in SEQ ID NO:13;(f) substitution of an aspartic acid at amino acid position 100 in SEQ ID NO:13;(g) substitution of an asparagine at amino acid position 105 in SEQ ID NO:13;(h) substitution of an aspartic acid at amino acid position 106 in SEQ ID NO:13;(i) substitution of a lysine at amino acid position 108 in SEQ ID NO:13;(j) substitution of a lysine at amino acid position 109 in SEQ ID NO:13;(k) substitution of a leucine at amino acid position 110 in SEQ ID NO:13;(l) substitution of a serine at amino acid position 112 in SEQ ID NO:13; and(m) substitution of an asparagine at amino acid position 113 in SEQ ID NO:13.
  • 12. (canceled)
  • 13. The RSV immunogen of claim 11, wherein said RSV immunogen comprises an amino acid sequence that is at least 95% identical to an amino acid sequence of a protein selected from the group consisting of 1s2xa—001 (SEQ ID NO:152), 1s2xa—002 (SEQ ID NO:155), 1s2xa—003 (SEQ ID NO:158), and 1s2xa—004 (SEQ ID NO:164).
  • 14. The RSV immunogen of claim 11, wherein said RSV immunogen comprises an amino acid sequence of a protein selected from the group consisting of 1s2xa—001 (SEQ ID NO:152), 1s2xa—002 (SEQ ID NO:155), 1s2xa—003 (SEQ ID NO:158), and 1s2xa—004 (SEQ ID NO:164).
  • 15. The RSV immunogen of claim 1, wherein said RSV immunogen comprises at least one characteristic selected from the group consisting of binding a motavizumab antibody, eliciting a humoral immune response against RSV and failing to elicit a cellular immune response.
  • 16-18. (canceled)
  • 19. The RSV immunogen of claim 1, wherein said RSV immunogen has a motavizumab antibody-binding domain comprising less than 9 consecutive amino acids from a motavizumab antibody-binding domain of RSV fusion protein.
  • 20. A nucleic acid molecule comprising a nucleic acid sequence that encodes an immunogen of claim 11.
  • 21. A nucleic acid molecule comprising a nucleic acid sequence that encodes an immunogen of claim 1.
  • 22-26. (canceled)
  • 27. A method selected from the group consisting of a method to elicit a neutralizing humoral immune response against RSV and a method to protect a patient from RSV infection, said method comprising administering an RSV immunogen of claim 1.
  • 28. (canceled)
  • 29. An immunogen comprising an antibody-binding domain that binds an antibody selected from the group consisting of motavizumab and 101F antibody, wherein the three-dimensional structure of said antibody-binding domain of said immunogen spatially corresponds to a three-dimensional structure of an antibody-binding domain of a fusion (F) peptide derived from respiratory syncytial virus (RSV) fusion (F) protein in a complex selected from the group consisting of: (a) a complex between a F peptide consisting of amino acid sequence SEQ ID NO:2 and motavizumab, said complex being set forth in the three-dimensional model defined by the atomic coordinates specified in Protein Data Bank accession code 3IXT; and(b) a complex between a F peptide consisting of amino acid sequence SEQ ID NO:4 and 101F antibody, said complex being set forth in the three-dimensional model defined by the atomic coordinates specified in Protein Data Bank accession code 3O41;wherein said antibody-binding domain of said immunogen comprises less than 12 consecutive amino acids from a motavizumab-binding domain or a 101F antibody-binding domain of RSV F protein, and wherein said immunogen elicits a humoral immune response against RSV.
  • 30. The immunogen of claim 29, wherein said antibody-binding domain of said immunogen comprises contact residues selected from the group consisting of: (a) contact residues that have a spatial orientation represented by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms from the corresponding backbone atoms of contact residues in an RSV F peptide that contacts motavizumab or 101F antibody in a complex set forth in claim 29;(b) contact residues of the motavizumab-binding domain embedded in a protein scaffold comprising a three-dimensional structure having two alpha helices defined by atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the two alpha helices of the peptide consisting of amino acid sequence SEQ ID NO:2 when complexed with motavizumab, the three-dimensional model of said complex being defined by the coordinates specified in Protein Data Bank accession code 3IXT; and,(c) contact residues of the 101F antibody-binding domain embedded in a protein scaffold comprising a three-dimensional structure with atomic coordinates that have a root mean square deviation of protein backbone atoms of less than 10 angstroms when superimposed on the peptide consisting of amino acid sequence SEQ ID NO:4 when complexed with 101F antibody, the 3-dimensional model of said complex being defined by the coordinates specified in Protein Data Bank accession code 3O41.
  • 31-33. (canceled)
  • 34. An RSV immunogen of claim 29, wherein said antibody-binding domain of said immunogen comprises less than 9 consecutive amino acids from a motavizumab-binding domain or a 101F antibody-binding domain of RSV F protein.
  • 35. The immunogen of claim 29, wherein the motavizumab-binding domain of said immunogen comprises less than 15 amino acids of the motavizumab-binding domain from said RSV F peptide, wherein said amino acids are in clusters of no more than 8 consecutive amino acids per cluster.
  • 36. (canceled)
  • 37. The immunogen of claim 29, wherein the 101F antibody-binding domain of said immunogen comprises no more than 10 amino acids of the 101F antibody-binding domain of said RSV F peptide, wherein said amino acids are in clusters of no more than 8 consecutive amino acids per cluster.
  • 38. (canceled)
  • 39. The immunogen of claim 29, wherein said immunogen comprises an amino acid sequence of a protein selected from the group consisting of 1LP1_b (SEQ ID NO:11), 1S2X_a (SEQ ID NO:12), truncated 1S2X_a (SEQ ID NO:13), and 2EIA_a (SEQ ID NO:14).
  • 40. The immunogen of claim 29, wherein said immunogen comprises an amino acid sequence selected from the group consisting of 1LP1_b (SEQ ID NO:11), 1S2X_a (SEQ ID NO:12), truncated 1S2X_a (SEQ ID NO:13), and 2EIA_a (SEQ ID NO:14), wherein said amino acid sequence comprises at least one substitution selected from the group consisting of: (a) substitution of a serine for the amino acid spatially corresponding to the amino acid at position 2 of SEQ ID NO:2;(b) substitution of a leucine for the amino acid spatially corresponding to the amino acid at position 5 of SEQ ID NO:2;(c) substitution of a serine for the amino acid spatially corresponding to the amino acid at position 6 of SEQ ID NO:2;(d) substitution of an isoleucine for the amino acid spatially corresponding to the amino acid at position 8 of SEQ ID NO:2;(e) substitution of an asparagine for the amino acid spatially corresponding to the amino acid at position 9 of SEQ ID NO:2;(f) substitution of an aspartic acid for the amino acid spatially corresponding to the amino acid at position 10 of SEQ ID NO:2;(g) substitution of a threonine for the amino acid spatially corresponding to the amino acid at position 14 of SEQ ID NO:2;(h) substitution of an asparagine for the amino acid spatially corresponding to the amino acid at position 15 of SEQ ID NO:2;(i) substitution of an aspartic acid for the amino acid spatially corresponding to the amino acid at position 16 of SEQ ID NO:2;(j) substitution of a lysine for the amino acid spatially corresponding to the amino acid at position 18 of SEQ ID NO:2;(k) substitution of a lysine for the amino acid spatially corresponding to the amino acid at position 19 of SEQ ID NO:2;(l) substitution of a leucine for the amino acid spatially corresponding to the amino acid at position 20 of SEQ ID NO:2;(m) substitution of a serine for the amino acid spatially corresponding to the amino acid at position 22 of SEQ ID NO:2; and(n) substitution of an asparagine for the amino acid spatially corresponding to the amino acid at position 23 of SEQ ID NO:2.
  • 41. (canceled)
  • 42. The immunogen of claim 30, wherein said protein scaffold comprises an amino acid sequence comprising at least one substitution selected from the group consisting of: (a) substitution of a lysine for the amino acid spatially corresponding to the amino acid at position 1 of SEQ ID NO:4;(b) substitution of an arginine for the amino acid spatially corresponding to the amino acid at position 3 of SEQ ID NO:4;(c) substitution of a isoleucine for the amino acid spatially corresponding to the amino acid at position 5 of SEQ ID NO:4;(d) substitution of a isoleucine for the amino acid spatially corresponding to the amino acid at position 6 of SEQ ID NO:4;(e) substitution of a lysine for the amino acid spatially corresponding to the amino acid at position 7 of SEQ ID NO:4;(f) substitution of a threonine for the amino acid spatially corresponding to the amino acid at position 8 of SEQ ID NO:4;(g) substitution of a phenylalanine for the amino acid spatially corresponding to the amino acid at position 9 of SEQ ID NO:4; and,(h) substitution of a serine for the amino acid spatially corresponding to the amino acid at position 10 of SEQ ID NO: 4.
  • 43. (canceled)
  • 44. The immunogen of claim 29, wherein said immunogen comprises an amino acid sequence of a protein selected from the group consisting of 1lp1b—001 (SEQ ID NO:18), 1lp1b—002 (SEQ ID NO:21), 1lp1b—003 (SEQ ID NO:24), 1lp1b—004 (SEQ ID NO:149), 1s2xa—001 (SEQ ID NO:152), 1s2xa—002 (SEQ ID NO:155), 1s2xa—003 (SEQ ID NO:158), 1s2xa—004 (SEQ ID NO:164), 2eiaa—001 (SEQ ID NO:167), 2eiaa—002 (SEQ ID NO:170), mota—1lp1b.m1.c1.d1_glyc1 (SEQ ID NO:55), mota—1lp1b.m1.c1.d1_glyc2 (SEQ ID NO:56), 1lp1b—003_Glyc1 (SEQ ID NO:39), 1lp1b—003_Glyc2 (SEQ ID NO:42), 1lp1b—003_Glyc3 (SEQ ID NO:45), 1lp1b—003_Glyc4 (SEQ ID NO:48), 1lp1b—003_Glyc5 (SEQ ID NO:51), 1lp1b—003_Glyc6 (SEQ ID NO:54), mota—1lp1b.m1.c1.d1_des1—1 (SEQ ID NO:66), mota—1lp1b.m1.c1.d1_des1—2 (SEQ ID NO:67), mota—1lp1b.m1.c1.d1_des1—3 (SEQ ID NO:68), mota—1lp1b.m1.c1.d1_des1—5 (SEQ ID NO:69), mota—1lp1b.m1.c1.d1_des1—6 (SEQ ID NO:70), mota—1lp1b.m1.c1.d1_des1—7 (SEQ ID NO:71), mota—1lp1b.m1.c1.d1_des1—8 (SEQ ID NO:72), mota—1lp1b.m1.c1.d1_des1—9 (SEQ ID NO:73), mota—1lp1b.m1.c1.d1_des1—10 (SEQ ID NO:74), mota—1lp1b.m1.c1.d1_des1—11 (SEQ ID NO:75), 1lp1b—003_Surf1 (SEQ ID NO:59), 1lp1b—003_Surf6 (SEQ ID NO:62), and 1lp1b—003_Surf8 (SEQ ID NO:65), 1lp1b—003_ferritin (SEQ ID NO:138), 1lp1b—003_eumS (SEQ ID NO:140), 1lp1b—003_eumSP (SEQ ID NO:142), 1lp1b—003_eum_L (SEQ ID NO:144), 1lp1b—003eum_LP (SEQ ID NO:146), 1lp1b—003_K46A (SEQ ID NO:78), 1lp1b—003_Q52A (SEQ ID NO:81), 1lp1b—003_I13L_F27A (SEQ ID NO:87), 1lp1b—003_L41I_L42V (SEQ ID NO:90), 1lp1b-003_L41I_L42A (SEQ ID NO:93), 1lp1b—003_I13A (SEQ ID NO:105), 1lp1b—003_L16A (SEQ ID NO:108), 1lp1b—003_F27A (SEQ ID NO:111), 1lp1b—003_L41A (SEQ ID NO:114), 1lp1b—003_L42A (SEQ ID NO:117), and 1lp1b—003_Neg1 (SEQ ID NO:132).
  • 45-49. (canceled)
  • 50. The RSV immunogen of claim 29, wherein said immunogen elicits a humoral immune response against RSV, but not a cellular immune response.
  • 51. An immunogen comprising a protein comprising an amino acid sequence of a protein selected from the group consisting of RSV F0 Fd (SEQ ID NO:174), RSV F Fd (SEQ ID NO:175), and RSV F0 Fd GAG (SEQ ID NO:176).
  • 52. A nucleic acid molecule comprising a nucleic acid sequence that encodes an immunogen of claim 29.
  • 53-59. (canceled)
  • 60. A method selected from the group consisting of: (a) a method to elicit a neutralizing humoral immune response against RSV and,(b) a method to protect a patient from RSV infection, said method comprising administering an RSV immunogen of claim 29.
  • 61. (canceled)
  • 62. An antibody protein selected from the group consisting of: A) an antibody protein comprising a heavy chain comprising SEQ ID NO:5, except that said antibody protein comprises at least one amino acid substitution selected from the group consisting of:(a) substitution of the amino acid corresponding to position 32 of SEQ ID NO:5, is a histidine or a glutamic acid;(b) substitution of the amino acid at position 35 of SEQ ID NO:5 is substituted with an alanine;(c) substitution of the amino acid at position 52 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of lysine, histidine, threonine, serine and arginine;(d) substitution of the amino acid at position 53 of SEQ ID NO:5 is substituted with a histidine or a serine;(e) substitution of the amino acid at position 54 of SEQ ID NO:5 is substituted with a phenylalanine or an arginine;(f) substitution of the amino acid at position 56 of SEQ ID NO:5 is substituted with an amino acid selected from the group consisting of isoleucine, serine, glutamic acid, and aspartic acid;(g) substitution of the amino acid at position 58 of SEQ ID NO:5 is substituted with a tyrosine or a tryptophan;(h) substitution of the amino acid at position 97 of SEQ ID NO:5 is substituted with an aspartic acid, a histidine or an arginine;(i) substitution of the amino acid at position 99 of SEQ ID NO:5 is substituted with aspartic acid;(j) substitution of the amino acid at position 100 of SEQ ID NO:5 is substituted with a tyrosine, a tryptophan or a histidine; and(k) substitution of the amino acid at position 100A of SEQ ID NO:5 is substituted with a serine or a threonine; and,B). an antibody protein comprising a heavy chain comprising SEQ ID NO:6, except that said antibody protein comprises at least one amino acid substitution selected from the group consisting of:(a) substitution of the amino acid at position 32 of SEQ ID NO:6 is substituted with a phenylalanine;(b) substitution of the amino acid at position 49 of SEQ ID NO:6 is substituted with a histidine or an arginine;(c) substitution of the amino acid at position 92 of SEQ ID NO:6 is substituted with a lysine;(d) substitution of the amino acid at position 94 of SEQ ID NO:6 is substituted with a histidine; and(e) substitution of the amino acid at position 96 of SEQ ID NO:6 is substituted with a histidine.
  • 63. (canceled)
  • 64. A nucleic acid molecule comprising a nucleic acid sequence that encodes an antibody protein of claim 62.
  • 65-68. (canceled)
  • 69. A method to protect a patient from RSV infection comprising administering to said patient an antibody protein of claim 62, wherein said administration protects said patient from RSV infection.
Parent Case Info

This application claims the benefit of U.S. Provisional Patent Application No. 61/253,826, filed Oct. 21, 2009, which is hereby expressly incorporated by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/US2010/053558 10/21/2010 WO 00 8/24/2012
Provisional Applications (1)
Number Date Country
61253826 Oct 2009 US