Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
EPITOPES IN AMYLOID BETA AND CONFORMATIONALLY-SELECTIVE ANTIBODIES THERETO
Document Type and Number:
WIPO Patent Application WO/2017/079834
Kind Code:
A1
Abstract:
The disclosure pertains to epitopes identified in A-beta including conformational epitopes, antibodies thereto and methods of making and using immunogens and antibodies specific thereto.

Inventors:
CASHMAN NEIL R (CA)
PLOTKIN STEVEN S (CA)
Application Number:
PCT/CA2016/051304
Publication Date:
May 18, 2017
Filing Date:
November 09, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV BRITISH COLUMBIA (CA)
International Classes:
C07K14/47; A61K39/00; A61K39/385; A61K47/60; A61K47/68; A61K49/00; A61K51/10; A61P25/28; A61P37/04; C07K5/00; C07K5/10; C07K7/06; C07K7/64; C07K16/18; C07K16/46; C12N5/10; C12N15/12; C12N15/13; G01N33/53; G01N33/577
Domestic Patent References:
WO2009052439A22009-04-23
Foreign References:
US20130136747A12013-05-30
Other References:
See also references of EP 3374382A4
Attorney, Agent or Firm:
BERESKIN & PARR LLP/S.E.N.C.R.L., S.R.L. (CA)
Download PDF:
Claims:
Claims:

I . A compound, preferably a cyclic compound, comprising an A-beta peptide the peptide comprising EDV and up to 8, 7 or 6 A-beta residues, and a linker, wherein the linker is covalently coupled to the A-beta peptide N-terminus residue and the A-beta C-terminus residue.

2. The compound of claim 1 , wherein the A-beta peptide is selected from a peptide having a sequence of any one of SEQ ID NOS: 1 -7, optionally selected from AEDV (SEQ ID NO: 1 ), AEDVG (SEQ ID NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), FAED (SEQ ID NO: 7) and EDVG (SEQ ID NO: 6).

3. The cyclic compound of claim 1 or 2, wherein the cyclic compound is a cyclic peptide.

4. The cyclic compound of any one of claims 1 to 3, comprising i) a curvature increase of V in the cyclic compound of at least 10%, at least 20%, or at least 30% increased compared to V in the context of a corresponding linear compound; ii) at least one residue selected from E, D and V, wherein at least one dihedral angle of said residue is different by at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, or at least 80 degrees compared to the corresponding dihedral angle in the context of a corresponding linear compound; iii) an O-C-Ca-C dihedral angle that is different by at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees or at least 80 degrees compared to the corresponding dihedral angle in the context of a corresponding linear compound; and/or iv) a conformation for D and/or V as measured by entropy that is at least 10%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40% more constrained compared to a corresponding linear compound.

5. The cyclic compound of any one of claims 1 to 4, wherein the A-beta peptide is AEDV.

6. The cyclic compound of any one of claims 1 to 5, wherein the compound further comprises a detectable label.

7. The compound of any one of claims 1 to 6, wherein the linker comprises or consists of 1 -8 amino acids and/or equivalently functioning molecules and/or one or more functional izable moieties.

8. The compound of claim 7 wherein the linker amino acids are selected from A and G, and/or wherein the functionalizable moiety is C.

9. The compound of any one of claims 1 to 8, wherein the linker comprises or consists of amino acids GCG or CGC.

10. The compound of any one of claims 1 to 9, wherein the linker comprises a PEG molecule.

I I . The cyclic compound of claim 1 , wherein the cyclic compound is selected from the following structures: Cyclo(CGAEDVG)

Chemical Formula: C24H37N7O-HS Molecular Weight: 631.66

Cyclo(C-PEG2-AEDVG)

Chemical Formula: C28H45N7O-13S Molecular Weight: 719.76 and

Cyclo(CGAEDV-PEG2)

Chemical Formula: C28H45N7013S

Molecular Weiqht: 7 9.76

12. An immunogen comprising the cyclic compound of any one of claims 1 to 1 1.

13. The immunogen of claim 12 wherein the cyclic compound is coupled to a carrier protein or immunogenicity enhancing agent.

14. The immunogen of claim 13, wherein the carrier protein is bovine serum albumin (BSA) or the immunogenicity-enhancing agent is keyhole limpet haemocyanin (KLH).

15. A composition comprising the compound of any one of claims 1 to 1 1 or the immunogen of any one of claims 12 to 14.

16. The composition of claim 15, further comprising an adjuvant.

17. The composition of claim 16, wherein the adjuvant is aluminum phosphate or aluminum hydroxide.

18. An isolated antibody that specifically binds to an A-beta peptide having a sequence of AEDV or a related epitope sequence, optionally as set forth in any one of SEQ ID NOS: 1 -7.

19. The antibody of claim 18, wherein the antibody specifically binds an epitope on A-beta, wherein the epitope comprises at least two consecutive amino acid residues predominantly involved in binding to the antibody, wherein the at least two consecutive amino acids are DV embedded within EDV or AEDV (SEQ ID NO: 1 ).

20. The antibody of claim 18, wherein the epitope comprises or consists of EDV, AEDV (SEQ ID NO: 1 ), AEDVG (SEQ ID NO: 2), AEDVGS (SEQ I D NO: 3), FAEDV (SEQ ID NO: 5), FAED (SEQ ID NO: 7) and EDVG (SEQ ID NO: 6).

21. The antibody of any one of claims 18 to 20, wherein the antibody is a conformation specific and/or selective antibody that specifically or selectively binds to AEDV or a related epitope peptide presented in a cyclic compound, optionally a cyclic compound of any one of claims 1 to 11 , preferably a cyclic peptide having a sequence as set forth in SEQ ID NO: 4, 23 or 24. 22. The antibody of any one of claims 18 to 21 , wherein the antibody selectively binds A-beta oligomer over A-beta monomer and/or A-beta fibril.

23. The antibody of claim 21 , wherein the selectivity is at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 100 fold, at least 500 fold, at least 1000 fold more selective for A-beta oligomer over A-beta monomer and/or A-beta fibril.

24. The antibody of any one of claims 18 to 23, wherein the antibody does not specifically and/or selectively bind a linear peptide comprising sequence AEDV (SEQ ID NO: 1 ) or a related epitope, optionally wherein the sequence of the linear peptide is a linear version of a cyclic compound used to raise the antibody, optionally a linear peptide having a sequence as set forth in SEQ ID NO: 4, 23 or 24.

25. The antibody of any one of claims 18 to 24, wherein the antibody lacks or has negligible binding to A-beta monomer and/or A-beta fibril plaques in situ.

26. The antibody of any one of claims 18 to 25 wherein the antibody is a monoclonal antibody or a polyclonal antibody.

27. The antibody of any one of claims 18 to 26, wherein the antibody is a humanized antibody.

28. The antibody of any one of claims 18 to 27, therein the antibody is an antibody binding fragment selected from Fab, Fab', F(ab')2, scFv, dsFv, ds-scFv, dimers, nanobodies, minibodies, diabodies, and multimers thereof.

29. The antibody of any one of claims 18 to 28, comprising a light chain variable region and a heavy chain variable region, optionally fused, the heavy chain variable region comprising complementarity determining regions CDR-H1 , CDR-H2 and CDR-H3, the light chain variable region comprising complementarity determining region CDR-L1 , CDR-L2 and CDR-L3 and with the amino acid sequences of said CDRs comprising the sequences:

CDR-H 1 GFSLTSYG (SEQ ID NO: 12)

CDR-H2 IWAGGST (SEQ ID NO: 13)

CDR-H3 FQPSYYYGMDY (SEQ ID NO: 14) CDR-L1 QTIVHSNGDTY (SEQ ID NO: 15)

CDR-L2 SVS (SEQ ID NO: 16)

CDR-L3 FQGSHVPYT (SEQ ID NO: 17)

30. The antibody of any one of claims 18 to 29, wherein the antibody comprises a heavy chain variable region comprising: i) an amino acid sequence as set forth in SEQ ID NO: 19; ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ ID NO: 19, wherein the CDR sequences are as set forth in SEQ ID NO: 12, 13 and 14, or iii) a conservatively substituted amino acid sequence i).

31. The antibody of any one of claims 18 to 30, wherein the antibody comprises a light chain variable region comprising i) an amino acid sequence as set forth in SEQ ID NO: 21 , ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ I D NO: 21 , wherein the CDR sequences are as set forth in

SEQ ID NO: 15, 16 and 17, or iii) a conservatively substituted amino acid sequence of i).

32. The antibody of any one of claims 18 to 31 , wherein the heavy chain variable region amino acid sequence is encoded by a nucleotide sequence as set forth in SEQ ID NO: 18 or a codon degenerate or optimized version thereof; and/or the antibody comprises a light chain variable region amino acid sequence encoded by a nucleotide sequence as set out in SEQ

ID NO: 20 or a codon degenerate or optimized version thereof.

33. The antibody of any one of claims 18 to 32, wherein the heavy chain variable region comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 19 and/or the light chain variable region comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 21.

34. The antibody of any one of claims 18 to 28, wherein the antibody competes for binding to human A-beta with an antibody comprising the CDR sequences as recited in Table 10.

35. An immunoconjugate comprising the antibody of any one of claims 18-34 and a detectable label or cytotoxic agent.

36. The immunoconjugate of claim wherein the detectable label comprises a positron emitting radionuclide, optionally for use in subject imaging such as PET imaging.

37. A composition comprising the antibody of any one of claims 18 to 34, or the immunoconjugate of claim 35 or 36, optionally with a diluent.

38. A nucleic acid molecule encoding a proteinaceous portion of the compound or immunogen of any one of claims 1 to 14, the antibody of any one of claims 18 to 34 or proteinaceous immunoconjugates of claim 35 and 36.

39. A vector comprising the nucleic acid of claim 38.

40. A cell expressing an antibody of any one of claims 1 to 34, optionally wherein the cell is a hybridoma comprising the vector of claim 39.

41. A kit comprising the compound of any one of claims 1 -11 , the immunogen of any one of claims 12-14, the antibody of any one of claims 18 to 34, the immunoconjugate of claim 35 or 36 the composition of claim 37, the nucleic acid molecule of claim 38, the vector of claim 39 or the cell of claim 40.

42. A method of making the antibody of any one of claims 18 to 35, comprising administering the compound or immunogen of any one of claims 1 - 14 or a composition comprising said compound or immunogen to a subject and isolating antibody and/or cells expressing antibody specific or selective for the compound or immunogen administered and/or A-beta oligomers, optionally lacking or having negligible binding to a linear peptide comprising the A-beta peptide and/or lacking or having negligible plaque binding.

43. A method of determining if a biological sample comprises A-beta, the method comprising: a. contacting the biological sample with an antibody of any one of claims 18 to 35 or the immunoconjugate of claim 36 or 37; and

b. detecting the presence of any antibody complex.

44. The method of claim 43 for determining if the biological sample contains A-beta oligomer the method comprising:

a. contacting the sample with the antibody of any one of claims 18 to 35 or the immunoconjugate of claim 36 or 37 that is specific and/or selective for A-beta oligomers under conditions permissive for forming an antibody:A-beta oligomer complex; and b. detecting the presence of any complex;

wherein the presence of detectable complex is indicative that the sample may contain A-beta oligomer.

45. The method of claim 43, wherein the amount of complex is measured.

46. The method of any one of claims 43-45 wherein the sample comprises brain tissue or an extract thereof, whole blood, plasma, serum and/or CSF.

47. The method of any one of claims 43 to 46, wherein the sample is a human sample.

48. The method of any one of claims 43 to 47, wherein the sample is compared to a control, optionally a previous sample.

49. The method of any one of claims 43 to 48, wherein the level of A-beta is detected by SPR. 50. A method of measuring a level of A-beta in a subject, the method comprising

administering to a subject at risk or suspected of having or having AD, an immunoconjugate comprising an antibody of claims 35 or 36 wherein the antibody is conjugated to a detectable label; and detecting the label, optionally quantitatively detecting the label.

51. The method of claim 50, wherein the label is a positron emitting radionuclide.

52. A method of inducing an immune response in a subject, comprising administering to the subject a compound or combination of compounds of any one of claims 1 to 1 1 , optionally a cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope peptide sequence, an immunogen and/or composition comprising said compound or said immunogen; and optionally isolating cells and/or antibodies that specifically or selectively bind the A-beta peptide in the compound or immunogen administered.

53. A method of inhibiting A-beta oligomer propagation, the method comprising contacting a cell or tissue expressing A-beta with or administering to a subject in need thereof an effective amount of an A-beta oligomer specific or selective antibody or immunoconjugate of any one of claims 1 to 36, to inhibit A-beta aggregation and/or oligomer propagation.

54. A method of treating AD and/or other A-beta amyloid related diseases, the method comprising administering to a subject in need thereof i) an effective amount of an antibody or immunoconjugate of any one of claims 1 -36, optionally an A-beta oligomer specific or selective antibody, or a pharmaceutical composition comprising said antibody; 2) administering an isolated cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope sequence or immunogen or pharmaceutical composition comprising said cyclic compound, or 3) a nucleic acid or vector comprising a nucleic acid encoding the antibody of 1 or the immunogen of 2, to a subject in need thereof.

55. The method of claim 54, wherein a biological sample from the subject to be treated is assessed for the presence or levels of A-beta using an antibody described herein.

56. The method of any one of claims 53 to 55, wherein more than one antibody or immunogen is administered.

57. The method of claim any one of claims 53 to 56, wherein the antibody, immunoconjugate, immunogen, composition or nucleic acid or vector is administered directly to the brain or other portion of the CNS.

58. The method of any one of claims 53 to 57, wherein the composition is a pharmaceutical composition comprising the compound or immunogen in admixture with a pharmaceutically acceptable, diluent or carrier.

59. An isolated peptide comprising an A beta peptide consisting of the sequence of any one of the sequences set forth in SEQ ID NOS: 1 -7.

60. The isolated peptide of claim 59, wherein the peptide is a cyclic peptide comprising a linker wherein the linker is covalently coupled to the A-beta peptide N-terminus residue and/or the A-beta C-terminus residue.

61. The isolated peptide of claim 59 or 60 comprising a detectable label.

62. A nucleic acid sequence encoding the isolated peptide of any one of claims 59 to 61.

63. A hybridoma cell or cell line expressing the antibody of any one of claims 18 to 34.

Description:
Title: Epitopes in Amyloid beta and conformational ly-selective antibodies thereto Related Applications

[0001] This is a PCT application which claims the benefit of priority of United States Patent

Application Serial Number 62/253044, filed November 9, 2015; United States Patent Application Serial Number 62/309,765, filed on March 17, 2016; United States Patent Application Serial Number 62/365,634, filed on July 22, 2016; and United States Patent Application Serial Number 62/393,615, filed on September 12, 2016, each of which are incorporated herein by reference.

Field

[0002] The present disclosure relates to Amyloid beta (A-beta or Αβ) epitopes and antibodies thereto and more specifically to conformational A-beta epitopes that are for example selectively accessible in A-beta oligomers, and related antibody compositions.

Background

[0003] Amyloid-beta (A-beta), which exists as a 36-43 amino acid peptide, is a product released from amyloid precursor protein (APP) by the enzymes β and γ secretase. In AD patients, A- beta can be present in soluble monomers, insoluble fibrils and soluble oligomers. In monomer form, A-beta exists as a predominantly unstructured polypeptide chain. In fibril form, A-beta can aggregate into distinct morphologies, often referred to as strains. Several of these structures have been determined by solid-state NMR.

[0004] For, example, structures for several stains of fibrils are available in the Protein Data

Bank (PDB), a crystallographic database of atomic resolution three dimensional structural data, including a 3-fold symmetric A/3 structure (PDB entry, 2M4J); a two-fold symmetric structure of A/3-40 monomers (PDB entry 2LMN), and a single-chain, parallel in-register structure of A/3-42 monomers (PDB entry 2MXU).

[0005] The structure of 2M4J is reported in Lu et al [8], and the structure of 2MXU is reported in Xiao et al [9]. The structure of 2LMN is reported in Petkova et al [10].

[0006] A-beta oligomers have been shown to kill cell lines and neurons in culture and block a critical synaptic activity that subserves memory, referred to as long term potentiation (LTP), in slice cultures and living animals.

[0007] The structure of the oligomer has not been determined to date. Moreover, NMR and other evidence indicates that the oligomer exists not in a single well-defined structure, but in a conformationally-plastic, malleable structural ensemble with limited regularity. Moreover, the concentration of toxic A-beta oligomer species is far below either that of the monomer or fibril (estimates vary but are on the order of 1000-fold below or more), making this target elusive.

[0008] Antibodies that bind Abeta have been described. [0009] WO2009086539A2 titled TREATMENT AND PROPHYLAXIS OF AMYLOIDOSIS is directed to Amyloidosis and amyloid light chain amyloidosis, by administering peptides comprising neoepitopes, such as amyloid protein A (AA) fragments from a C-terminal region of AA, and antibodies specific for neoepitopes of aggregated amyloid proteins, for example, antibodies specific for the C-terminal region of AA fibrils.

[0010] WO2003070760 titled ANTI-AMYLOID BETA ANTIBODIES AND THEIR USE is directed towards antibody molecules capable of specifically recognizing two regions of the R-A4 peptide, wherein the first region comprises the amino acid sequence AEFRHDSGY or a fragment thereof and wherein the second region comprises the amino acid sequence VHHAEDVFFAEDVG or a fragment thereof.

[0011] WO2006066089 titled HU MANIZED AMYLOID BETA ANTIBODIES FOR USE IN

I MPROVING COGNITION is directed to improved agents and methods for treatment of diseases associated with beta amyloid and in particular to the identification and characterization of a monoclonal antibody, 12A1 1 , that specifically binds to Αβ peptide and is effective at reducing plaque burden associated with amyloidogenic disorders (e.g. , AD).

[0012] WO2007068429 titled ANTIBODIES AGAINST AMYLOID BETA 4 WITH

GLYCOSYLATED IN THE VARIABLE REGION is directed to a purified antibody molecule preparation being characterized in that at least one antigen binding site comprises a glycosylated asparagine (Asn) in the variable region of the heavy chain (V H ).

[0013] WO 03/016466 is directed variant 266 antibodies that are engineered to lack an N- glycosylation site within the CDR2 of the heavy chain, pharmaceutical compositions thereof, and polynucleotide sequences, vectors, and transformed cells useful to express the variant antibodies. The variants are described to sequester soluble A-beta peptide from human biological fluids and specifically bind an epitope contained within position 13-28 of the amyloid beta peptide.

[0014] Yu et al. describes a hexavalent foldable Αβ1 -15 (6Αβ15) fused to PADRE or toxin-derived carrier proteins. Wang et al 2016 report that peripheral administration of this antibody mitigates Alzheimer's disease like pathology and cognitive decline in a transgenic animal of aged Alzheimer Disease [11 ], [12].

[0015] Antibodies that preferentially or selectively bind A-beta oligomers are desirable.

Summary

[0016] Described herein are epitopes, and more particularly conformational epitopes, in A- beta consisting of residues AEDV (SEQ ID NO: 1 ) or related epitopes, and antibodies that specifically and/or selectively bind said epitopes. The epitopes may be selectively exposed in the oligomeric species of A-beta, in a conformation that distinguishes oligomeric species from that in the monomer and/or fibril.

[0017] An aspect of the disclosure includes a compound, preferably a cyclic compound, comprising an A-beta peptide, the peptide comprising EDV, optionally AEDV (SEQ ID NO: 1 ) and up to 8, up to 7 or up to 6 A-beta residues, and a linker, wherein the linker is covalently coupled to the A- beta peptide N-terminus residue and the A-beta C-terminus residue.

[0018] In an embodiment, the A-beta peptide is selected from a peptide having a sequence of any one of SEQ ID NOS: 1 -7, optionally selected from AEDV (SEQ ID NO: 1 ), AEDVG (SEQ I D NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), FAED (SEQ ID NO: 7) and EDVG (SEQ ID NO: 6).

[0019] In another embodiment, the cyclic compound is a cyclic peptide.

[0020] In another embodiment, the cyclic compound described herein comprises i) a curvature increase of V in the cyclic compound of at least 10%, at least 20%, or at least 30% increased compared to V in the context of a corresponding linear compound; ii) at least one residue selected from E, D and V, wherein at least one dihedral angle of said residue is different by at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, or at least 80 degrees compared to the corresponding dihedral angle in the context of a corresponding linear compound; iii) an O-C-Ca-C dihedral angle that is different by at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees or at least 80 degrees compared to the corresponding dihedral angle in the context of a corresponding linear com pound; and/or iv) a conformation for D and/or V as measured by entropy that is at least 10%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40% more constrained compared to a corresponding linear compound.

[0021] In another embodiment, the A-beta peptide is AEDV.

[0022] In another embodiment, the compound further comprises a detectable label.

[0023] In another embodiment, the linker comprises or consists of 1 -8 amino acids and/or equivalently functioning molecules and/or one or more functionalizable moieties.

[0024] In another embodiment, the linker amino acids are selected from A and G, and/or wherein the functionalizable moiety is C.

[0025] In another embodiment, the linker comprises or consists of amino acids GCG or

CGC.

[0026] In another embodiment, the linker comprises a PEG molecule. [0027] In another embodiment, the cyclic compound is selected from the following structures:

Cyclo(CGAEDVG)

Chemical Formula: C2.H3 7 N 7 OHS

Molecular Weight: 631.66

Cyclo(C-PEG2-AEDVG)

and

Cyclo(CGAEDV-PEG2)

Chemical Formula: 0 28 Η 45 Ν 7 0 13 ε

Molecular Weiaht: 719.76 [0028] Another aspect of the disclosure provides an immunogen comprising the cyclic compound described herein.

[0029] In one embodiment, the compound is coupled to a carrier protein or immunogenicity enhancing agent.

[0030] In another embodiment, the carrier protein is bovine serum albumin (BSA) or the immunogenicity-enhancing agent is keyhole limpet haemocyanin (KLH).

[0031] Another aspect provides a composition comprising the compound of described herein or the immunogen described herein.

[0032] In one embodiment, the composition further comprises an adjuvant.

[0033] In another embodiment, the adjuvant is aluminum phosphate or aluminum hydroxide.

[0034] Another aspect provides an isolated antibody that specifically binds to an A-beta peptide having a sequence of AEDV or a related epitope sequence, optionally as set forth in any one of SEQ ID NOS: 1 -7.

[0035] In one embodiment, the antibody specifically binds an epitope on A-beta, wherein the epitope comprises at least two consecutive amino acid residues predominantly involved in binding to the antibody, wherein the at least two consecutive amino acids are DV embedded within EDV or AEDV (SEQ ID NO: 1 ).

[0036] In another embodiment, the epitope comprises or consists of EDV, AEDV (SEQ ID

NO: 1 ), AEDVG (SEQ ID NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), FAED (SEQ ID NO: 7) and EDVG (SEQ ID NO: 6).

[0037] In another embodiment, the antibody is a conformation specific and/or selective antibody that specifically or selectively binds to AEDV or a related epitope peptide presented in a cyclic compound, optionally a cyclic compound of any one of claims 1 to 1 1 , preferably a cyclic peptide having a sequence as set forth in SEQ ID NO: 4, 23 or 24.

[0038] In another embodiment, the antibody selectively binds A-beta oligomer over A-beta monomer and/or A-beta fibril.

[0039] In another embodiment, the selectivity is at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 100 fold, at least 500 fold, at least 1000 fold more selective for A-beta oligomer over A-beta monomer and/or A-beta fibril.

[0040] In another embodiment, the antibody does not specifically and/or selectively bind a linear peptide comprising sequence AEDV (SEQ ID NO: 1 ) or a related epitope, optionally wherein the sequence of the linear peptide is a linear version of a cyclic compound used to raise the antibody, optionally a linear peptide having a sequence as set forth in SEQ ID NO: 4, 23 or 24.

[0041] In another embodiment, the antibody lacks or has negligible binding to A-beta monomer and/or A-beta fibril plaques in situ. [0042] In another embodiment, the antibody is a monoclonal antibody or a polyclonal antibody.

[0043] In another embodiment, the antibody is a humanized antibody.

[0044] In another embodiment, the antibody is an antibody binding fragment selected from

Fab, Fab', F(ab')2, scFv, dsFv, ds-scFv, dimers, nanobodies, minibodies, diabodies, and multimers thereof.

[0045] In another embodiment, the antibody comprises a light chain variable region and a heavy chain variable region, optionally fused, the heavy chain variable region comprising complementarity determining regions CDR-H1 , CDR-H2 and CDR-H3, the light chain variable region comprising complementarity determining region CDR-L1 , CDR-L2 and CDR-L3 and with the amino acid sequences of said CDRs comprising the sequences:

CDR-H 1 GFSLTSYG (SEQ ID NO: 12)

CDR-H2 IWAGGST (SEQ ID NO: 13)

CDR-H3 FQPSYYYGMDY (SEQ ID NO: 14)

CDR-L1 QTIVHSNGDTY (SEQ ID NO: 15) CDR-L2 SVS (SEQ ID NO: 16)

CDR-L3 FQGSHVPYT (SEQ ID NO: 17)

[0046] In another embodiment, the antibody comprises a heavy chain variable region comprising: i) an amino acid sequence as set forth in SEQ ID NO: 19; ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ ID NO: 19, wherein the CDR sequences are as set forth in SEQ ID NO: 12, 13 and 14, or iii) a conservatively substituted amino acid sequence i).

[0047] In another embodiment, the antibody comprises a light chain variable region comprising i) an amino acid sequence as set forth in SEQ ID NO: 21 , ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ I D NO: 21 , wherein the CDR sequences are as set forth in SEQ ID NO: 15, 16 and 17, or iii) a conservatively substituted amino acid sequence of i).

[0048] In another embodiment, the heavy chain variable region amino acid sequence is encoded by a nucleotide sequence as set forth in SEQ ID NO: 18 or a codon degenerate or optimized version thereof; and/or the antibody comprises a light chain variable region amino acid sequence encoded by a nucleotide sequence as set out in SEQ ID NO: 20 or a codon degenerate or optimized version thereof. [0049] In another embodiment, the heavy chain variable region comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 19 and/or the light chain variable region comprises or consists of an amino acid sequence as set forth in SEQ ID NO: 21.

[0050] In another embodiment, the antibody competes for binding to human A-beta with an antibody comprising the CDR sequences as recited in Table 10.

[0051] Another aspect provides an immunoconjugate comprising the antibody described herein and a detectable label or cytotoxic agent.

[0052] In one embodiment, the detectable label comprises a positron emitting radionuclide, optionally for use in subject imaging such as PET imaging.

[0053] Another aspect provides a composition comprising the antibody described herein, or the immunoconjugate described herein, optionally with a diluent.

[0054] Another aspect provides a nucleic acid molecule encoding a proteinaceous portion of the compound or immunogen described herein, the antibody described herein or proteinaceous immunoconjugates described herein.

[0055] Another aspect provides a vector comprising the nucleic acid described herein.

[0056] Another aspect provides a cell expressing an antibody described herein, optionally wherein the cell is a hybridoma comprising the vector described herein

[0057] Another aspect provides a kit comprising the compound described herein, the immunogen described herein, the antibody described herein, the immunoconjugate described herein the composition described herein, the nucleic acid molecule described herein, the vector described herein or the cell described herein.

[0058] Another aspect provides a method of making the antibody described herein, comprising administering the compound or immunogen described herein or a composition comprising said compound or immunogen to a subject and isolating antibody and/or cells expressing antibody specific or selective for the compound or immunogen administered and/or A-beta oligomers, optionally lacking or having negligible binding to a linear peptide comprising the A-beta peptide and/or lacking or having negligible plaque binding.

[0059] Another aspect provides a method of determining if a biological sample comprises A- beta, the method comprising:

a. contacting the biological sample with an antibody described herein or the immunoconjugate described herein; and

b. detecting the presence of any antibody complex.

[0060] In one embodiment, the method comprises determining if the biological sample contains A-beta oligomer the method comprising: a. contacting the sample with the antibody described herein or the immunoconjugate described herein that is specific and/or selective for A-beta oligomers under conditions permissive for forming an antibody:A-beta oligomer complex; and

b. detecting the presence of any complex

wherein the presence of detectable complex is indicative that the sample may contain A-beta oligomer.

[0061] In one embodiment, the amount of complex is measured.

[0062] In another embodiment, the sample comprises brain tissue or an extract thereof, whole blood, plasma, serum and/or CSF.

[0063] In another embodiment, the sample is a human sample.

[0064] In another embodiment, the sample is compared to a control, optionally a previous sample.

[0065] In another embodiment, the level of A-beta is detected by SPR.

[0066] Another aspect provides a method of measuring a level of A-beta in a subject, the method comprising administering to a subject at risk or suspected of having or having AD, an immunoconjugate comprising an antibody described herein wherein the antibody is conjugated to a detectable label; and detecting the label, optionally quantitatively detecting the label.

[0067] In one embodiment, the label is a positron emitting radionuclide.

[0068] Another aspect provides a method of inducing an immune response in a subject, comprising administering to the subject a compound or combination of compounds described herein optionally a cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope peptide sequence, an immunogen and/or composition comprising said compound or said immunogen; and optionally isolating cells and/or antibodies that specifically or selectively bind the A-beta peptide in the compound or immunogen administered.

[0069] Another aspect provides a method of inhibiting A-beta oligomer propagation, the method comprising contacting a cell or tissue expressing A-beta with or administering to a subject in need thereof an effective amount of an A-beta oligomer specific or selective antibody or immunoconjugate described herein, to inhibit A-beta aggregation and/or oligomer propagation.

[0070] Another aspect provides a method of treating AD and/or other A-beta amyloid related diseases, the method comprising administering to a subject in need thereof i) an effective amount of an antibody or immunoconjugate described herein, optionally an A-beta oligomer specific or selective antibody, or a pharmaceutical composition comprising said antibody; 2) administering an isolated cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope sequence or immunogen or pharmaceutical composition comprising said cyclic compound, or 3) a nucleic acid or vector comprising a nucleic acid encoding the antibody of 1 or the immunogen of 2, to a subject in need thereof.

[0071] In one embodiment, a biological sample from the subject to be treated is assessed for the presence or levels of A-beta using an antibody described herein.

[0072] In another embodiment, more than one antibody or immunogen is administered.

[0073] In another embodiment, the antibody, immunoconjugate, immunogen, composition or nucleic acid or vector is administered directly to the brain or other portion of the CNS

[0074] In another embodiment, the composition is a pharmaceutical composition comprising the compound or immunogen in admixture with a pharmaceutically acceptable, diluent or carrier

[0075] Another aspect provides an isolated peptide comprising an A-beta peptide consisting of the sequence of any one of the sequences set forth in SEQ ID NOs: 1 -7.

[0076] In one embodiment, the peptide is a cyclic peptide comprising a linker wherein the linker is covalently coupled to the A-beta peptide N-terminus residue and/or the A-beta C-terminus residue.

[0077] In another embodiment, the isolated peptide comprises a detectable label.

[0078] Another aspect provides nucleic acid sequence encoding the isolated peptide described herein.

[0079] Another aspect provides a hybridoma cell or cell line expressing the antibody described herein.

[0080] Other features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the disclosure are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description. Brief description of the drawings

[0081] An embodiment of the present disclosure will now be described in relation to the drawings in which:

[0082] FIG. 1 : Predicted epitope for partially unfolded A-beta strain/structure 2M4J. The epitope AEDV (SEQ ID NO: 1 ) is predicted for chain A, B, and C of this structure, with an activation cost of approximately 1 1 kCal/mol. The abscissa of the boxes indicates the center residue index of the epitope, here residue ID 23 (x-axis), and the ordinate indicates a length 4 (y-axis), thus consisting of residue 21 -24 or AEDV (SEQ ID NO: 1 ). For purposes of plotting predicted epitopes of even-integer length, the algorithm assigns residues just right of center to be the "center" residue. [0083] FIG. 2: Landscape of partially unfolding 2MXU, chain L. The left marked epitope has length 5 residues with free energy cost 4.8 kcal/mol, with center at residue 23, thus residues 21 -25 or AEDVG (SEQ ID NO: 2) is predicted as an epitope for this chain. The right epitope has length 6 residues with free energy cost 4.9 kcal/mol, with center at residue 24, thus residues 21 -26 or AEDVGS (SEQ ID NO: 3) is predicted as an epitope for this chain but with slightly higher energy cost.

[0084] FIG. 3: Curvature as a function of residue index. Mean curvature in the equilibrium ensemble for the cyclic peptide CGAEDVG (SEQ ID NO: 4) is shown (dashed black), along with the curvature for the linear peptide (solid black), and the curvature of the various monomers in the fibril (thin curves).

[0085] FIG. 4: O-C-Ca-Cp dihedral angle distribution for the side chain heavy atoms of A21.

[0086] FIG. 5: Dihedral angle distribution for the angle O-C-Ca-Cp involving the side chain heavy atoms of E22. Purely as a comparison, the N-Ca-Cp-C γ dihedral distribution is very similar for all three isoforms.

[0087] FIG. 6: Dihedral angle distributions for angles involving the side chain heavy atoms of

D23.

[0088] FIG. 7: Dihedral angle distributions for angles involving the side chain heavy atoms of

V24.

[0089] FIG. 8: Top left panel: Entropy change of the linear and cyclic peptides relative to the entropy in the fibril. Top right panel: entropy change of dihedral angles in E22. Bottom left panel: entropy of dihedral angles in D23. Bottom right panel: entropy change of dihedral angles in V24.

[0090] FIG. 9: Equilibrium Ramachadran angles for residues A, E, D and V, in both the linear and cyclic forms of the peptide CGAEDVG (SEQ ID NO: 4).

[0091] FIG. 10: Likelihood of exposure as a function of sequence, as determined by the collective coordinates method. The AEDV (SEQ ID NO: 1 ) epitope emerges as a prediction for both structures 2LMN and 2MXU.

[0092] FIG. 11 : Solubility vs residue index for A-beta42 peptide. AEDV (SEQ ID NO: 1 ) has values of -1.1 , -0.46, +0.16, and +0.89 respectively. The solubility as one continues towards the C- terminus increases, which places emphasis on the valine being more exposed due to its local sequence environment.

[0093] FIG. 12: Plots of the solvent accessible surface area (SASA), the weighted SASA, σ, SASA,, and σ, SASA, - (σ, SASA,) ftM . When SASA is weighted by the solubility of the residue, more emphasis is placed on the C-terminal residues in the group AEDV (SEQ ID NO: 1 ).

[0094] FIG. 13: Two separate views of the root mean squared deviation (RMSD) values to the centroids of the three largest clusters of the linear peptide ensemble. Each point corresponds to a given conformation taken from either the linear peptide, cyclic peptide, or fibril equilibrium ensembles. The cyclic peptide ensemble is conformationally distinct from either the linear peptide or fibril ensembles.

[0095] FIG. 14: Cyclic peptides comprising AEDV (SEQ ID NO: 1 ).

[0096] FIG. 15: Representative conformations taken from the cyclic and linear peptide ensembles. Two views of the same image are shown. The cyclic peptide is shown, and the sidechain orientations present for a representative conformation of the linear peptide ensemble are shown in black, superimposed on the cyclic peptide.

[0097] FIG. 16: Surface plasmon resonance (SPR) direct binding assay of IgG antibodies to cyclic peptide and linear peptide in Panel A, and A-beta oligomer and A-beta monomer in Panel B.

[0098] FIG. 17: Primary screening of clones from tissue culture supernatants using ELISA and SPR direct binding assay. Plot comparing clone binding in SPR direct binding assay versus ELISA.

[0099] FIG. 18: SPR direct binding assay data for select clones, for binding to cyclic peptide

(circles), linear peptide (squares), A-beta monomer (upward pointing triangles), and A-beta oligomer (downward pointing triangles).

[00100] FIG. 19: Immunohistochemical staining of plaque from cadaveric AD brain using

6E 0 positive control antibody (A) and an antibody (302-24-10A4) raised against cyclo(CGAEDVG) (SEQ ID NO:4) (B)..

[00101] FIG. 20: Secondary screening of selected and purified antibodies using an SPR indirect (capture) binding assay. SPR binding response of A-beta oligomer to captured antibody minus binding response of A-beta monomer to captured antibody (circle); SPR binding response of pooled soluble brain extract from AD patients to captured antibody minus binding response of pooled brain extract from non-AD controls to captured antibody (triangle); SPR binding response of pooled cerebrospinal fluid (CSF) from AD patients to captured antibody minus binding response of pooled CSF from non-AD controls to captured antibody (square).

[00102] FIG. 21 : Verification of antibody binding to stable A-beta oligomers. SPR sensorgrams and binding response plots of varying concentrations of commercially-prepared stable A-beta oligomers binding to immobilized antibodies. Panel A shows results with the positive control mAb6E10, Panel B with the negative isotype control and Panel C with antibody raised against cyclo (CGAEDVG) (SEQ ID NO: 4)— antibody 302-24 in Panel D. Panel D plots binding of two antibody clones raised against cyclic peptide comprising AEDV (SEQ ID No: 1 ), with A-beta oligomer at a concentration of 1 micromolar. Isotype control lgG1 and positive control 6E10 are also shown.

[00103] A plot of the propagation of A-beta aggregation in vitro in the presence or absence of a representative antibody raised using a cyclic peptide comprising AEDV (SEQ ID NO: 1 ) (not shown).

[00104] Table 1 shows the peak values of the dihedral angle distribution for those dihedral angles whose distributions show significant differences between the cyclic peptide and other species.

11

RECTIFIED SHEET (RULE 91.1) Column 1 is the specific dihedral considered, column 2 is the peak value of the dihedral distribution for that angle in the context of the linear peptide CGAEDVG (SEQ ID NO: 4), column 3 is the peak value of the dihedral distribution for that angle in the context of the cyclic peptide CGAEDVG (SEQ ID NO: 4), column 4 is the difference of the peak values of the dihedral distributions for the linear and cyclic peptides, and column 5 is the peak value of the dihedral distribution for the peptide AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J. See Figures 5-7

[00105] Table 2 shows peak values of the Ramachandran backbone phi/psi angle distributions; peak values for all phi/psi angles showed significant differences between the cyclic peptide and other species for all residues. The first column is the residue considered, which manifests two angles, phi and psi, indicated in parenthesis. The 2 nd column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ ID NO: 1 ) in the context of the linear peptide CGAEDVG (SEQ ID NO: 4), while the 3 rd column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ ID NO: 1 ) in the context of the cyclic peptide CGAEDVG (SEQ ID NO: 4), and the last column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J. See Figure 9.

[00106] Table 3 shows Ramachandran backbone and side chain dihedral angles shown for the cyclic peptide in FIG 15, and for a representative conformation from the linear peptide ensemble. For the orientations taken by the linear peptide, side chains are shown in black in FIG 15, superimposed on the cyclic peptide.

[00107] Table 4 is a table of mean curvature values for each residue in the cyclic, linear, and 2M4J fibril ensembles.

[00108] Table 5 shows the binding properties of selected tissue culture supernatant antibody clones.

[00109] Table 6 shows the binding properties summary for selected purified antibodies.

[00110] Table 7 lists the A-beta oligomer SPR binding value minus monomer SPR binding value for an antibody raised against cyclo(CGAEDVG) (SEQ ID NO: 4).

[00111] Table 8 lists properties of antibodies tested on formalin fixed tissues.

[00112] Table 9 is an exemplary toxicity assay.

[00113] Table 10 lists CDR sequences.

[00114] Table 1 1 lists heavy chain and light chain variable sequences.

[00115] Table 12 is a table of A-beta epitope sequences and select sequences with linker.

[00116] Table 13 provides the full A-beta 1 -42 human polypeptide sequence.

Detailed description of the Disclosure [00117] Provided herein are antibodies, immunotherapeutic compositions and methods which may target epitopes preferentially accessible in toxic oligomeric species of A-beta, including oligomeric species associated with Alzheimer's disease. A region in A-beta has been identified that may be specifically and/or selectively accessible to antibody binding in oligomeric species of A-beta.

[00118] As demonstrated herein, generation of oligomer-specific or oligomer selective antibodies was accomplished through the identification of targets on A-beta peptide that are not present, or present to a lesser degree, on either the monomer and/or fibril. Oligomer-specific epitopes need not differ in primary sequence from the corresponding segment in the monomer or fibril, however they would be conformationally distinct in the context of the oligomer. That is, they would present a distinct conformation in terms of backbone and/or side-chain orientation in the oligomer that would not be present (or would be unfavourable) in the monomer and/or fibril.

[00119] Antibodies raised to linear peptide regions may not to be selective for oligomer, and thus may bind to monomer or A-beta plaques as well.

[00120] As described herein, to develop antibodies that may be selective for oligomeric forms of A-beta, the inventors sought to identify regions of A-beta sequence that are prone to disruption in the context of the fibril, and that may be exposed on the surface of the oligomer.

[00121] As described in the Examples, the inventors have identified a region they have determined to be prone to disruption in the context of the fibril. The inventors designed cyclic compounds comprising the identified target region to satisfy criteria of an alternate conformation such as higher curvature, higher exposed surface area, alternative dihedral angle distributions, and/or did not readily align by root mean squared deviation (RMSD) to either the linear or fibril ensembles.

[00122] Antibodies could be raised using a cyclic peptide comprising the target region, that selectively bound the cyclic peptide compared to a linear peptide of the same sequence (e.g. corresponding linear sequence). Experimental results are described and identify epitope-specific and conformationally selective antibodies that bind synthetic oligomer selectively compared to synthetic monomers, bind CSF from AD patients preferentially over control CSF and/or bind soluble brain extract from AD patients preferentially over control soluble brain extract. Further staining of AD brain tissue identified antibodies that show no or negligible plaque binding and in vitro studies found that the antibodies inhibited Αβ oligomer propagation and aggregation.

I. Definitions

[00123] As used herein, the term 'A-beta' may alternately be referred to as 'amyloid beta',

'amyloid β', Abeta, A-beta or Άβ'. Amyloid beta is a peptide of 36-43 amino acids and includes all wild-type and mutant forms of all species, particularly human A-beta. A-beta40 refers to the 40 amino acid form; A-beta42 refers to the 42 amino acid form, etc. The amino acid sequence of human wildtype A-beta42 is shown in SEQ I D NO: 22.

[00124] As used herein, the term "A-beta monomer" herein refers to any of the individual subunit forms of the A-beta (e.g. 1 -40, 1-42, 1-43) peptide. [00125] As used herein, the term "A-beta oligomer" herein refers to a plurality of any of the A- beta subunits wherein several (e.g. at least two) A-beta monomers are non-covalently aggregated in a conformationally-flexible, partially-ordered, three-dimensional globule of less than about 100, or more typically less than about 50 monomers. For example, an oligomer may contain 3 or 4 or 5 or more monomers. The term "A-beta oligomer" as used herein includes both synthetic A-beta oligomer and/or native A-beta oligomer. " Native A-beta oligomer" refers to A-beta oligomer formed in vivo, for example in the brain and CSF of a subject with AD.

[00126] As used herein, the term "A-beta fibril" refers to a molecular structure that comprises assemblies of non-covalently associated, individual A-beta peptides which show fibrillary structure under an electron microscope. The fibrillary structure is typically a "cross beta" structure; there is no theoretical upper limit on the size of multimers, and fibrils may comprise thousands or many thousands of monomers. Fibrils can aggregate by the thousands to form senile plaques, one of the primary pathological morphologies diagnostic of AD.

[00127] The term "AEDV" means the amino acid sequence alanine, glutamic acid, aspartic acid and valine as shown in SEQ ID NO: 1 . Similarly EDV, EDVG (SEQ ID NO: 6), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), EDVG (SEQ ID NO: 6), FAED (SEQ ID NO: 7) refer to the amino acid sequence identified by the 1 -letter amino acid code. Depending on the context, the reference of the amino acid sequence can refer to a sequence in A-beta or an isolated peptide, such as the amino acid sequence of a cyclic compound.

[00128] The term "alternate conformation than occupied by A, E, D and/or V in the monomer and/or fibril" as used herein means having one or more differing conformational properties selected from solvent accessibility, entropy, curvature (e.g. in the context of peptide AEDV (SEQ ID NO: 1 )), and dihedral angle of one or more backbone or side chain dihedral angles compared to said property for A, E, D and/or V in A-beta in a linear compound, monomer and/or A-beta fibril as shown for example in PDB 2M4J or 2MXU and shown in FIGs. 1 -15. For example, FIG. 3 shows that the aspartic acid (D) in the cyclic peptide has somewhat higher curvature than that in the linear peptide, and substantially higher curvature than that in the fibril and that the valine (V) has substantially larger curvature in the cyclic peptide than either the linear peptide or fibril. FIG. 5 shows that the dihedral angle distribution for angle (O-C-Ca-C ) involving the side chain E22 reflects an alternate conformational distribution compared to either the monomer or fibril. FIG.s. 6 and 7 show that the dihedral angle distributions for angles involving the side chains of D23 and V24 and reflect alternate conformational distributions compared to either the monomer or fibril for residues D and V. The alternate conformation can be similarly, less or more "constrained" than the comparator conformation. For example FIG. 8 demonstrates that D and V in the cyclic compound described are more constrained than in the monomer. FIG. 9 demonstrates that the distributions of the Ramachandran dihedral angles for the backbone of cyclic peptides are substantially different than those for either monomer or fibril for A, E, D, and V. FIG. 13 shows that the structures of A, E, D, V cluster differently for the equilibrium ensemble in the cyclic peptide than the equilibrium ensembles of either the linear peptide or corresponding sequence in the fibril.

[00129] The term "amino acid" includes all of the naturally occurring amino acids as well as modified L-amino acids. The atoms of the amino acid can include different isotopes. For example, the amino acids can comprise deuterium substituted for hydrogen nitrogen-15 substituted for nitrogen-14, and carbon-13 substituted for carbon-12 and other similar changes.

[00130] The term "antibody" as used herein is intended to include, monoclonal antibodies, polyclonal antibodies, single chain, veneered, humanized and other chimeric antibodies and binding fragments thereof, including for example a single chain Fab fragment, Fab'2 fragment or single chain Fv fragment. The antibody may be from recombinant sources and/or produced in animals such as rabbits, llamas, sharks etc. Also included are human antibodies that can be produced in transgenic animals or using biochemical techniques or can be isolated from a library such as a phage library. Humanized or other chimeric antibodies may include sequences from one or more than one isotype or class or species.

[00131] The phrase "isolated antibody" refers to antibody produced in vivo or in vitro that has been removed from the source that produced the antibody, for example, an animal, hybridoma or other cell line (such as recombinant insect, yeast or bacteria cells that produce antibody). The isolated antibody is optionally "purified", which means at least: 80%, 85%, 90%, 95%, 98% or 99% purity.

[00132] The term "binding fragment" as used herein to a part or portion of an antibody or antibody chain comprising fewer amino acid residues than an intact or complete antibody or antibody chain and which binds the antigen or competes with intact antibody. Exemplary binding fragments include without limitations Fab, Fab', F(ab')2, scFv, dsFv, ds-scFv, dimers, nanobodies, minibodies, diabodies, and multimers thereof. Fragments can be obtained via chemical or enzymatic treatment of an intact or complete antibody or antibody chain. Fragments can also be obtained by recombinant means. For example, F(ab')2 fragments can be generated by treating the antibody with pepsin. The resulting F(ab')2 fragment can be treated to reduce disulfide bridges to produce Fab' fragments. Papain digestion can lead to the formation of Fab fragments. Fab, Fab' and F(ab')2, scFv, dsFv, ds- scFv, dimers, minibodies, diabodies, bispecific antibody fragments and other fragments can also be constructed by recombinant expression techniques.

[00133] The terms "IMGT numbering" or " Im MunoGeneTics database numbering", which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e. hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody, or antigen binding portion thereof.

[00134] When an antibody is said to specifically bind to an epitope such as AEDV (SEQ ID

NO: 1 ), what is meant is that the antibody specifically binds to a peptide containing the specified residues or a part thereof for example at least 2 residues of AEDV (SEQ ID NO: 1 ), with a minimum affinity, and does not bind an unrelated sequence or unrelated sequence spatial orientation greater than for example an isotype control antibody. Such an antibody does not necessarily contact each residue of AEDV (SEQ ID NO: 1 ) and every single amino acid substitution or deletion within said epitope does not necessarily significantly affect and/or equally affect binding affinity.

[00135] When an antibody is said to selectively bind an epitope such as a conformational epitope, such as AEDV (SEQ ID NO: 1 ), what is meant is that the antibody preferentially binds one or more particular conformations containing the specified residues or a part thereof with greater affinity than it binds said residues in another conformation. For example, when an antibody is said to selectively bind a cyclopeptide comprising AEDV (SEQ ID NO: 1 ), or related epitope relative to a corresponding linear peptide, the antibody binds the cyclopeptide with at least a 2 fold greater affinity than it binds the linear peptide.

[00136] As used herein, the term "conformational epitope" refers to an epitope where the epitope amino acid sequence has a particular three-dimensional structure wherein at least an aspect of the three-dimensional structure not present or less likely to be present in a corresponding linear peptide is specifically and/or selectively recognized by the cognate antibody. The epitope e.g. AEDV (SEQ ID NO: 1 ) may be partially or completely exposed on the molecular surface of oligomeric A-beta and partially or completely obscured from antibody recognition in monomeric or fibrillar plaque A-beta. Antibodies which specifically and/or selectively bind a conformation-specific epitope recognize the spatial arrangement of one or more of the amino acids of that conformation-specific/selective epitope. For example an AEDV (SEQ ID NO: 1 ) conformational epitope, refers to an epitope of AEDV (SEQ ID NO: 1 ) that is recognized by antibodies specifically and/or selectively, for example at least 2 fold, 3 fold, 5 fold, 10 fold, 50 fold, 100 fold, 250 fold, 500 fold or 1000 fold or greater, more selectively as compared to linear AEDV (SEQ ID NO: 1 ).

[00137] The term "related epitope" as used herein means at least two residues of AEDV (SEQ

ID NO: 1 ) that are antigenic and/or sequences comprising 1 or 2 amino acid residues in a A-beta either N-terminal or C-terminal to at least two residues of AEDV (SEQ ID NO: 1 ). For example it is shown herein that AEDV (SEQ ID NO: 1 ), AEDVG (SEQ ID NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), EDVG (SEQ ID NO: 6) and FAED (SEQ ID NO: 7) were identified as regions prone to disorder in an A-beta fibril. AEDVG (SEQ I D NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), EDVG (SEQ ID NO: 6) and FAED (SEQ ID NO: 7) are accordingly related epitopes. Exemplary related epitopes can include A-beta sequences included in Table 12.

[00138] The term "constrained conformation" as used herein with respect to an amino acid or a side chain thereof, within a sequence of amino acids (e.g. D or V in AEDV (SEQ ID NO: 1 )), or with respect to a sequence of amino acids in a larger polypeptide, means decreased rotational mobility of the amino acid dihedral angles, relative to a corresponding linear peptide sequence, or the sequence or larger polypeptide, resulting in a decrease in the number of permissible conformations. This can be quantified for example by finding the entropy reduction for the ensemble of dihedral angle degrees of freedom, and is plotted in FIG. 8 for AEDV (SEQ ID NO: 1 ). For example, if the side chains in the sequence have less conformational freedom than the linear peptide, the entropy will be reduced. Such conformational restriction would enhance the conformational selectivity of antibodies specifically raised to this antigen. The amino acid sequence AEDV (SEQ ID NO: 1 ) is most constrained in a fibril structure, where it has less conformational freedom than either the cyclic peptide or the monomer. FIG. 8 shows that on average, residues A21 and E22 have comparable entropy increase AS over the fibril. On the other hand, residues D23 and V24 have less entropy in the cyclic peptide than they do in the linear peptide. The side chains of D and V are more strongly constrained in the cyclic peptide than in the linear peptide.

[00139] The term "more constrained conformation" as used herein means that the dihedral angle distribution (ensemble of allowable dihedral angles) of one or more dihedral angles is at least 10% more constrained than in the comparator conformation, as determined for example by the entropy of the amino acids, for example D, and/or V (e.g. a more constrained conformation has lower entropy). Specifically, the average entropy change relative to the entropy in the fibril, S(constrained) - S(fibril), of AEDV (SEQ ID NO: 1 ) in the more constrained conformational ensemble is on average reduced by more than 10% or reduced by more than 20% or reduced by more than 30% or reduced by more than 40%, from the unconstrained conformational ensemble, e.g. of the quantity S(linear) - S(fibril) for the linear peptide (FIG. 8 plots this entropy reduction for the cyclic peptide ensemble, which exhibits approximately 30% entropy reduction for D23 and 45% entropy reduction for V24).

[00140] The term " no or negligible plaque binding" or "lacks or has negligible plaque binding" as used herein with respect to an antibody means that the antibody does not show typical plaque morphology staining on immunohistochemistry (e.g. in situ) and the level of staining is comparable to or no more than 2 fold the level seen with an IgG negative (e.g. irrelevant) isotype control.

[00141] The term "Isolated peptide" refers to peptide that has been produced, for example, by recombinant or synthetic techniques, and removed from the source that produced the peptide, such as recombinant cells or residual peptide synthesis reactants. The isolated peptide is optionally "purified", which means at least: 80%, 85%, 90%, 95%, 98% or 99% purity and optionally pharmaceutical grade purity.

[00142] The term "detectable label" as used herein refers to moieties such as peptide sequences (such a myc tag, HA-tag, V5-tag or NE-tag), fluorescent proteins that can be appended or introduced into a peptide or compound described herein and which is capable of producing, either directly or indirectly, a detectable signal. For example, the label may be radio-opaque, positron- emitting radionuclide (for example for use in PET imaging), or a radioisotope, such as 3 H, 3 N, 4 C, 8F, 32 P, 35 S, 23 l, 25 l, 3 1; a fluorescent (fluorophore) or chemiluminescent (chromophore) compound, such as fluorescein isothiocyanate, rhodamine or luciferin; an enzyme, such as alkaline phosphatase, beta-galactosidase or horseradish peroxidase; an imaging agent; or a metal ion. The detectable label may be also detectable indirectly for example using secondary antibody.

[00143] The term "epitope" as commonly used means an antibody binding site, typically a polypeptide segment, in an antigen that is specifically recognized by the antibody. As used herein "epitope" can also refer to the amino acid sequences or part thereof identified on A-beta using the collective coordinates method described. For example an antibody generated against an isolated peptide corresponding to a cyclic compound comprising the identified target region AEDV (SEQ ID NO: 1 ), recognizes part or all of said epitope sequence. An epitope is "accessible" in the context of the present specification when it is accessible to binding by an antibody.

[00144] The term "greater affinity" as used herein refers to a relative degree of antibody binding where an antibody X binds to target Y more strongly (K on ) and/or with a smaller dissociation constant (K off ) than to target Z, and in this context antibody X has a greater affinity for target Y than for Z. Likewise, the term "lesser affinity" herein refers to a degree of antibody binding where an antibody X binds to target Y less strongly and/or with a larger dissociation constant than to target Z, and in this context antibody X has a lesser affinity for target Y than for Z. The affinity of binding between an antibody and its target antigen, can be expressed as K A equal to 1/K D where K D is equal to k on /k off . The k on and k off values can be measured using surface plasmon resonance technology, for example using a Molecular Affinity Screening System (MASS-1 ) (Sierra Sensors GmbH, Hamburg, Germany). An antibody that is selective for a conformation presented in a cyclic compound optional a cyclic peptide for example has a greater affinity for the cyclic compound (e.g. cyclic peptide) compared to a corresponding sequence in linear form (e.g. the sequence non-cyclized).

[00145] Also as used herein, the term "immunogenic" refers to substances that elicit the production of antibodies, activate T-cells and other reactive immune cells directed against an antigenic portion of the immunogen.

[00146] The term "corresponding linear compound" with regard to a cyclic compound refers to a compound, optionally a linear peptide, comprising or consisting of the same sequence or chemical moieties as the cyclic compound but in linear (i.e. non-cyclized) form, for example having properties as would be present in solution of a linear peptide. For example, the corresponding linear compound can be the synthesized peptide that is not cyclized.

[00147] As used herein "specifically binds" in reference to an antibody means that the antibody recognizes an epitope sequence and binds to its target antigen with a minimum affinity. For example a multivalent antibody binds its target with a K D of at least 1 e-6, at least 1 e-7, at least 1 e-8, at least 1 e-9, or at least 1 e-10. Affinities greater than at least 1 e-8 may be preferred. An antigen binding fragment such as Fab fragment comprising one variable domain, may bind its target with a 10 fold or 100 fold less affinity than a multivalent interaction with a non-fragmented antibody.

[00148] The term "selectively binds" as used herein with respect to an antibody that selectively binds a form of A-beta (e.g. fibril, monomer or oligomer) or a cyclic compound means that the antibody binds the form with at least 2 fold, at least 3 fold, or at least 5 fold, at least 10 fold, at least 100 fold, at least 250 fold, at least 500 fold or at least 1000 fold or more greater affinity. Accordingly an antibody that is more selective for a particular conformation (e.g. oligomer) preferentially binds the particular form of A-beta with at least 2 fold etc greater affinity compared to another form and/or a linear peptide. [00149] The term "linker" as used herein means a chemical moiety that can be covalently linked to the peptide comprising AEDV (SEQ ID NO: 1 ) epitope peptide, optionally linked to AEDV (SEQ ID NO: 1 ) peptide N- and C- termini to produce a cyclic compound. The linker can comprise a spacer and/or one or more functionalizable moieties. The linker via the functionalizable moieties can be linked to a carrier protein or an immunogen enhancing agent such as Keyhole Limpet Hemocyanin (KLH).

[00150] The term "spacer" as used herein means any preferably non-immunogenic or poorly immunogenic chemical moiety that can be covalently-linked directly or indirectly to a peptide N- and C- termini to produce a cyclic compound of longer length than the peptide itself, for example the spacer can be linked to the N- and C- termini of a peptide consisting of AEDV (SEQ ID NO: 1 ) to produce a cyclic compound of longer backbone length than the AEDV (SEQ I D NO: 1 ) sequence itself. That is, when cyclized, the peptide with a spacer (for example of 3 amino acid residues) makes a larger closed circle than the peptide without a spacer. The spacer may include, but is not limited to, moieties such as G, A, or PEG repeats, e.g. GAEDV (SEQ ID NO: 8), GAEDVG (SEQ ID NO: 9), GGAEDVG (SEQ ID NO: 10), GAEDVGG (SEQ ID NO: 1 1 ), etc. (see FIG. 12 for specific embodiments). The spacer may comprise or be coupled to one or more functionalizing moieties, such as one or more cysteine (C) residues, which can be interspersed within the spacer or covalently linked to one or both ends of the spacer. Where a functionalizable moiety such as a C residue is covalently linked to one or more termini of the spacer, the spacer is indirectly covalently linked to the peptide. The spacer can also comprise the functionalizable moiety in a spacer residue as in the case where a biotin molecule is introduced into an amino acid residue.

[00151] The term "functionalizable moiety" as used herein refers to a chemical entity with a

"functional group" which as used herein refers to a group of atoms or a single atom that will react with another group of atoms or a single atom (so called "complementary functional group") to form a chemical interaction between the two groups or atoms. In the case of cysteine, the functional group can be -SH which can be reacted to form a disulfide bond. Accordingly the linker can for example be CCC. The reaction with another group of atoms can be covalent or a strong non-covalent bond, for example as in the case of biotin-streptavidin bonds, which can have Kd~1 e-14. A strong non-covalent bond as used herein means an interaction with a Kd of at least 1 e-9, at least 1 e-10, at least 1 e-1 1 , at least 1 e-12, at least 1 e-13 or at least 1 e-14.

[00152] Proteins and/or other agents may be functionalized (e.g. coupled) to the cyclic compound, either to aid in immunogenicity, or to act as a probe in in vitro studies. For this purpose, any functionalizable moiety capable of reacting (e.g. making a covalent or non-covalent but strong bond) may be used. In one specific embodiment, the functionalizable moiety is a cysteine residue which is reacted to form a disulfide bond with an unpaired cysteine on a protein of interest, which can be, for example, an immunogenicity enhancing agent such as Keyhole Limpet Hemocyanin (KLH), or a carrier protein such as Bovine serum albumin (BSA) used for in vitro immunoblots or immunohistochemical assays. [00153] The term "reacts with" as used herein generally means that there is a flow of electrons or a transfer of electrostatic charge resulting in the formation of a chemical interaction.

[00154] The term "animal" or "subject" as used herein includes all members of the animal kingdom including mammals, optionally including or excluding humans.

[00155] A "conservative amino acid substitution" as used herein, is one in which one amino acid residue is replaced with another amino acid residue without abolishing the protein's desired properties. Suitable conservative amino acid substitutions can be made by substituting amino acids with similar hydrophobicity, polarity, and R-chain length for one another. Examples of conservative amino acid substitution include:

[00156] The term "sequence identity" as used herein refers to the percentage of sequence identity between two polypeptide sequences or two nucleic acid sequences. To determine the percent identity of two amino acid sequences or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g. , gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e. , % identity=number of identical overlapping positions/total number of positions X 100%). In one embodiment, the two sequences are the same length. The determination of percent identity between two sequences can also be accomplished using a mathematical algorithm. A preferred, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, Proc. Natl. Acad . Sci. U.S.A. 87:2264-2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. U.S.A. 90:5873- 5877. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al. , 1990, J. Mol. Biol. 215:403. BLAST nucleotide searches can be performed with the NBLAST nucleotide program parameters set, e.g. , for score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid molecules of the present application. BLAST protein searches can be performed with the XBLAST program parameters set, e.g. , to score-50, wordlength=3 to obtain amino acid sequences homologous to a protein molecule described herein. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al. , 1997, Nucleic Acids Res. 25: 3389-3402. Alternatively, PSI-BLAST can be used to perform an iterated search which detects distant relationships between molecules (Id. ). When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., of XBLAST and NBLAST) can be used (see, e.g. , the NCBI website). Another preferred, non- limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, 1988, CABIOS 4: 1 1 -17. Such an algorithm is incorporated in the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM 120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically only exact matches are counted.

[00157] For antibodies, percentage sequence identities can be determined when antibody sequences maximally aligned by I MGT or other (e.g. Kabat numbering convention). After alignment, if a subject antibody region (e.g. , the entire mature variable region of a heavy or light chain) is being compared with the same region of a reference antibody, the percentage sequence identity between the subject and reference antibody regions is the number of positions occupied by the same amino acid in both the subject and reference antibody region divided by the total number of aligned positions of the two regions, with gaps not counted, multiplied by 100 to convert to percentage.

[00158] The term "nucleic acid sequence" as used herein refers to a sequence of nucleoside or nucleotide monomers consisting of naturally occurring bases, sugars and intersugar (backbone) linkages. The term also includes modified or substituted sequences comprising non-naturally occurring monomers or portions thereof. The nucleic acid sequences of the present application may be deoxyribonucleic acid sequences (DNA) or ribonucleic acid sequences (RNA) and may include naturally occurring bases including adenine, guanine, cytosine, thymidine and uracil. The sequences may also contain modified bases. Examples of such modified bases include aza and deaza adenine, guanine, cytosine, thymidine and uracil; and xanthine and hypoxanthine. The nucleic acid can be either double stranded or single stranded, and represents the sense or antisense strand. Further, the term "nucleic acid" includes the complementary nucleic acid sequences as well as codon optimized or synonymous codon equivalents. The term "isolated nucleic acid sequences" as used herein refers to a nucleic acid substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors, or other chemicals when chemically synthesized. An isolated nucleic acid is also substantially free of sequences which naturally flank the nucleic acid (i.e. sequences located at the 5' and 3' ends of the nucleic acid) from which the nucleic acid is derived.

[00159] "Operatively linked" is intended to mean that the nucleic acid is linked to regulatory sequences in a manner which allows expression of the nucleic acid. Suitable regulatory sequences may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, or insect genes. Selection of appropriate regulatory sequences is dependent on the host cell chosen and may be readily accomplished by one of ordinary skill in the art. Examples of such regulatory sequences include: a transcriptional promoter and enhancer or RNA polymerase binding sequence, a ribosomal binding sequence, including a translation initiation signal. Additionally, depending on the host cell chosen and the vector employed, other sequences, such as an origin of replication, additional DNA restriction sites, enhancers, and sequences conferring inducibility of transcription may be incorporated into the expression vector.

[00160] The term "vector" as used herein comprises any intermediary vehicle for a nucleic acid molecule which enables said nucleic acid molecule, for example, to be introduced into prokaryotic and/or eukaryotic cells and/or integrated into a genome, and include plasmids, phagemids, bacteriophages or viral vectors such as retroviral based vectors, Adeno Associated viral vectors and the like. The term "plasmid" as used herein generally refers to a construct of extrachromosomal genetic material, usually a circular DNA duplex, which can replicate independently of chromosomal DNA.

[00161] By "at least moderately stringent hybridization conditions" it is meant that conditions are selected which promote selective hybridization between two complementary nucleic acid molecules in solution. Hybridization may occur to all or a portion of a nucleic acid sequence molecule. The hybridizing portion is typically at least 15 (e.g. 20, 25, 30, 40 or 50) nucleotides in length. Those skilled in the art will recognize that the stability of a nucleic acid duplex, or hybrids, is determined by the Tm, which in sodium containing buffers is a function of the sodium ion concentration and temperature (Tm = 81.5°C - 16.6 (Log10 [Na+]) + 0.41 (%(G+C) - 600/I), or similar equation). Accordingly, the parameters in the wash conditions that determine hybrid stability are sodium ion concentration and temperature. In order to identify molecules that are similar, but not identical, to a known nucleic acid molecule a 1 % mismatch may be assumed to result in about a 1 °C decrease in Tm, for example if nucleic acid molecules are sought that have a >95% identity, the final wash temperature will be reduced by about 5°C. Based on these considerations those skilled in the art will be able to readily select appropriate hybridization conditions. In preferred embodiments, stringent hybridization conditions are selected. By way of example the following conditions may be employed to achieve stringent hybridization: hybridization at 5x sodium chloride/sodium citrate (SSC)/5x Denhardt's solution/1.0% SDS at Tm - 5°C based on the above equation, followed by a wash of 0.2x SSC/0.1 % SDS at 60°C. Moderately stringent hybridization conditions include a washing step in 3x SSC at 42°C. It is understood, however, that equivalent stringencies may be achieved using alternative buffers, salts and temperatures. Additional guidance regarding hybridization conditions may be found in: Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. , 2002, and in: Sambrook et al. , Molecular Cloning: a Laboratory Manual, Cold Spring Harbor Laboratory Press, 2001.

[00162] The term "treating" or "treatment" as used herein and as is well understood in the art, means an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, diminishment of the reoccurrence of disease, and remission (whether partial or total), whether detectable or undetectable. "Treating" and "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. "Treating" and "treatment" as used herein also include prophylactic treatment. For example, a subject with early stage AD can be treated to prevent progression can be treated with a compound, antibody, immunogen, nucleic acid or composition described herein to prevent progression.

[00163] The term "administered" as used herein means administration of a therapeutically effective dose of a compound or composition of the disclosure to a cell or subject.

[00164] As used herein, the phrase "effective amount" means an amount effective, at dosages and for periods of time necessary to achieve a desired result. Effective amounts when administered to a subject may vary according to factors such as the disease state, age, sex, and weight of the subject. Dosage regime may be adjusted to provide the optimum therapeutic response.

[00165] The term "pharmaceutically acceptable" means that the carrier, diluent, or excipient is compatible with the other components of the formulation and not substantially deleterious to the recipient thereof.

[00166] Compositions or methods "comprising" or "including" one or more recited elements may include other elements not specifically recited. For example, a composition that "comprises" or "includes" an antibody may contain the antibody alone or in combination with other ingredients.

[00167] In understanding the scope of the present disclosure, the term "consisting" and its derivatives, as used herein, are intended to be close ended terms that specify the presence of stated features, elements, components, groups, integers, and/or steps, and also exclude the presence of other unstated features, elements, components, groups, integers and/or steps.

[00168] The recitation of numerical ranges by endpoints herein includes all numbers and fractions subsumed within that range (e.g. 1 to 5 includes 1 , 1.5, 2, 2.75, 3, 3.90, 4, and 5). It is also to be understood that all numbers and fractions thereof are presumed to be modified by the term "about." Further, it is to be understood that "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. The term "about" means plus or minus 0.1 to 50%, 5-50%, or 10- 40%, preferably 10-20%, more preferably 10% or 15%, of the number to which reference is being made.

[00169] Further, the definitions and embodiments described in particular sections are intended to be applicable to other embodiments herein described for which they are suitable as would be understood by a person skilled in the art. For example, in the following passages, different aspects of the invention are defined in more detail. Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous.

[00170] The singular forms of the articles "a," "an," and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a compound" or "at least one compound" can include a plurality of compounds, including mixtures thereof.

III. Epitopes and binding proteins

[00171] The inventors have identified an "epitope region" in A-beta AEDV (SEQ ID NO: 1 ) at amino acids 21 to 24 of A-beta. They have further identified that the epitope region may be or comprise a conformational epitope, and that AEDV (SEQ ID NO: 1 ) may be selectively accessible to antibody binding in oligomeric species of A-beta.

[00172] Without wishing to be bound by theory, fibrils may present interaction sites that have a propensity to catalyze oligomerization. This may be strain-specific, and may only occur when selective fibril surface not present in normal individuals is exposed and thus able to have aberrant interactions with the monomer (i.e. is presented to the monomer). Environmental challenges such as low pH, osmolytes present during inflammation, or oxidative damage may induce disruption in fibrils that can lead to exposure of more weakly stable regions. There is interest, then, to predict these weakly-stable regions, and use such predictions to rationally design antibodies that could target them. Regions likely to be disrupted in the fibril may also be good candidates for exposed regions in oligomeric species.

[00173] Computer based systems and methods to predict contiguous protein regions that are prone to disorder are described in US Patent Application serial no. 62/253044, SYSTEMS AND M ETHODS FOR PREDICTING M ISFOLDED PROTEIN EPITOPES BY COLLECTIVE COORDINATE BIASING filed November 9, 2015 and US Patent Application serial no. 12/574,637, " Methods and Systems for Predicting M isfolded Protein Epitopes" filed October 6, 2009, each of which are hereby incorporated by reference in their entirety. As described in the Examples, the methods were applied to A-beta and identified an epitope that may be more accessible in A-beta oligomers.

[00174] As described in the Examples, cyclic peptide cyclo(CGAEDVG) (SEQ ID NO: 4) may capture one or more of the conformational differences of the AEDV (SEQ ID NO: 1 ) epitope in oligomers relative to the monomer and/or fibril species. For example, differences in solvent accessible surface area, curvature, RMSD structural alignment, and the dihedral angle distributions for several of the amino acids and dihedral angles in the cyclic 7-mer cyclo (CGAEDVG) (SEQ ID NO: 4) were found to be substantially different than either the monomer and/or fibril, suggesting that the cyclic peptide provides for a conformational epitope that is distinct from the linear epitope. Antibodies raised using an immunogen comprising cyclo(CGAEDVG) (SEQ ID NO: 4) selectively bound cyclo(CGAEDVG) (SEQ I D NO: 4) over linear CGAEDVG (SEQ ID NO: 4) and selectively bound synthetic and/or native oligomeric A-beta species compared to monomeric A-beta and A-beta fibril plaques. Further antibodies raised to cyclo(CGAEDVG) (SEQ ID NO: 4) were able to inhibit in vitro propagation of A-beta aggregation.

11. AEDV (SEQ ID NO: 1 ) "Epitope" Compounds

[00175] Accordingly, the present disclosure identifies an epitope in A-beta consisting of amino acids AEDV (SEQ ID NO: 1 ) or a part thereof, AEDV (SEQ ID NO: 1 ) corresponding to amino acids residues 21-24 on A-beta. As demonstrated in the Examples, epitopes AEDV (SEQ ID NO: 1 ), AEDVG (SEQ I D NO: 2), AEDVGS (SEQ ID NO: 3), FAEDV (SEQ ID NO: 5), EDVG (SEQ ID NO: 6) and FAED (SEQ ID NO: 7) (collectively referred herein as related epitopes) were identified as regions prone to disorder in an A-beta fibril.

[00176] An aspect includes a compound comprising an isolated A-beta peptide comprising or consisting of AEDV (SEQ ID NO: 1 ), sequence of a related epitope and/or part of any of the foregoing.

[00177] In an embodiment, the A-beta peptide is selected from an amino acid sequence comprising or consisting of AEDV (SEQ ID NO: 1 ), AEDVG (SEQ ID NO: 2), AEDVGS (SEQ I D NO: 3), FAEDV (SEQ ID NO: 5), EDVG (SEQ ID NO: 6) or FAED (SEQ ID NO: 7). In an embodiment the A-beta peptide has a sequence of an A-beta peptide as set forth in any one of the epitopes in Table

12. In an embodiment, the compound comprises the sequence as set for in any of SEQ ID No: 4, 23 and 24.

[00178] In an embodiment, the compound is a cyclic compound, such as a cyclopeptide. The terms cyclopeptide and cyclic peptide are used interchangeably herein.

[00179] In some embodiments, the peptide comprising EDV, or AEDV (SEQ ID NO: 1 ) can include 1 , 2 or 3 additional residues in A-beta N- and/or C- terminus of AEDV (SEQ ID NO: 1 ) for example FAEDV (SEQ ID NO: 5). For example, the 3 amino acids N-terminal to AEDV (SEQ ID NO: 1 ) in A-beta are VFF and the 3 amino acids C-terminal to AEDV (SEQ ID NO: 1 ) are GSN. I n an embodiment, the A-beta peptide is a maximum of 6 A-beta residues. In an embodiment, the A-beta peptide is a maximum of 5 A-beta residues.

[00180] In an embodiment, the compound further includes a linker. The linker comprises a spacer and/or one or more functionalizable moieties. The linker can for example comprise 1 , 2, 3, 4, 5, 6, 7 or 8 amino acids and/or equivalently functioning molecules such as polyethylene glycol (PEG) moieties, and/or a combination thereof. In an embodiment, the spacer amino acids are selected from non-immunogenic or poorly immunogenic amino acid residues such as G and A, for example the spacer can be GGG, GAG, G(PEG)G, PEG-PEG-GG and the like. One or more functionalizable moieties e.g. amino acids with a functional group may be included for example for coupling the compound to an agent or detectable tag or a carrier such as BSA or an immunogenicity enhancing agent such as KLH.

[00181] In an embodiment the linker comprises GC-PEG, PEG-GC, GCG or PEG-C-PEG. [00182] In an embodiment, the linker comprises 2, 3, 4, 5, 6, 7 or 8 amino acids.

[00183] In embodiments wherein the peptide comprising AEDV (SEQ ID NO: 1 ) includes 1 , 2 or 3 additional residues found in A-beta that are N- and/or C- terminal to AEDV (SEQ ID NO: 1 ) (e.g. AEDVGS (SEQ ID NO: 3)), the linker is covalently linked to the N- and/or C- termini of the A-beta residues (e.g. where the peptide is AEDVGS (SEQ ID NO: 3), the linker is covalently lined to A and S residues). Similarly, where the A-beta peptide is AEDV (SEQ ID NO: 1 ), the linker is covalently linked to residues A and V and where the A-beta peptide is EDV, the linker is covalently linked to residues E and V.

[00184] Proteinaceous portions of compounds may be prepared by chemical synthesis using techniques well known in the chemistry of proteins such as solid phase synthesis or synthesis in homogenous solution.

[00185] As mentioned, the compound can be a cyclic compound. Reference to the "cyclic peptide" herein can refer to a fully proteinaceous compound (e.g. wherein the linker is for example 1 , 2, 3, 4, 5, 6, 7 or 8 amino acids). It is understood that properties described for the cyclic peptide determined in the examples can be incorporated in other compounds (e.g. other cyclic compounds) comprising non-amino acid linker molecules. The terms "cyclopeptide" and "cyclic peptide" are used interchangeably herein.

[00186] An aspect therefore provides a cyclic compound comprising peptide AEDV (SEQ ID

NO: 1 ) (or a part thereof such as EDV) and a linker, wherein the linker is covalently coupled to the peptide comprising AEDV (SEQ ID NO: 1 ) (e.g. the A and the V residues when the peptide consists of AEDV (SEQ ID NO: 1 )), optionally wherein at least D and/or V is in an alternate conformation than D and/or V in a linear peptide comprising AEDV (SEQ ID NO: 1 ) as would be manifest in A-beta monomer, and optionally wherein at least D, or at least V, is in a more constrained conformation than the conformation occupied in a linear compound such as a linear peptide comprising AEDV (SEQ I D NO: 1 ), as may be manifest for example in A-beta monomer.

[00187] The linear peptide comprising the A-beta sequence can be comprised in a linear compound. The linear compound or the linear peptide comprising AEDV (SEQ ID NO: 1 ) is in an embodiment, a corresponding linear peptide. I n another embodiment, the linear peptide is any length of A-beta peptide comprising AEDV(SEQ ID NO: 1 ), including for example a linear peptide comprising A-beta residues 1 -35, or smaller portions thereof such as A-beta residues 10-20, 1 1-20, 12-20, 13-20, 10-19, 10-18 and the like etc. The linear peptide can in some embodiments also be a full length A- beta peptide.

[00188] In an embodiment, the cyclic compound comprises: an A-beta peptide comprising

EDV and up to 6 A-beta residues and a linker, wherein the linker is covalently coupled to the peptide N-terminus residue and the C-terminus residue of the A-beta peptide and optionally wherein at least D is in an alternate conformation than D in a linear peptide, optionally a corresponding linear peptide, comprising EDV, and/or the conformation of D in EDV in the fibril, optionally wherein at least E, or at least D, or at least V is in a more constrained conformation than the conformation occupied in the linear peptide comprising EDV.

[00189] The cyclic compound can be synthesized as a linear molecule with the linker covalently attached to the N-terminus or C-terminus of the peptide comprising the A-beta peptide, optionally AEDV (SEQ ID NO: 1 ), prior to cyclization. Alternatively part of the linker is covalently attached to the N-terminus and part is covalently attached to the C-terminus prior to cyclization. In either case, the linear compound is cyclized for example in a head to tail cyclization (e.g. amide bond cyclization).

[00190] In an embodiment the cyclic compound comprises peptide comprising or consisting of

AEDV (SEQ ID NO: 1 ) and a linker, wherein the linker is coupled to the N- and C- termini of the peptide (e.g. the A and the V residues when the peptide consists of AEDV (SEQ ID NO: 1 )). In an embodiment, at least D and/or V is in an alternate conformation in the cyclic compound than occupied by D and/or V in a linear peptide comprising AEDV (SEQ ID NO: 1 ).

[00191] In an embodiment, at least D and/or V is in an alternate conformation in the cyclic compound than occupied by a residue, optionally by D and/or V, in the A-beta monomer and/or fibril.

[00192] In an embodiment, the alternate conformation is a constrained conformation.

[00193] In an embodiment, at least D, optionally alone or in combination with at least V is in a more constrained conformation than the conformation occupied in a linear peptide comprising AEDV (SEQ ID NO: 1 ).

[00194] In an embodiment, the conformation of D and/or D in combination with one or more of E and/or V is comprised in the compound in an alternate conformation, optionally in a more constrained conformation.

[00195] As shown in FIG. 8 residues D and V are in a more constrained conformation in the cyclic compound compared to the corresponding linear peptide. FIG. 8 shows that there is approximately a 30% entropy reduction for D23 and approximately a 45% entropy reduction for V24 In an embodiment, the cyclic compound has a conformation D and/or V that is at least 10%, at least 20%, at least 25%, at least 30%, at least 35% or at least 40% more constrained compared to a corresponding linear compound, as quantified by that residue's reduction in entropy.

[00196] For example, the alternate conformation can include one or more differing dihedral angles in residues D, and optionally in D and/or V differing from the dihedral angles in the linear peptide and/or peptide in the context of the fibril.

[00197] As shown in FIG. 5, the dihedral angle distribution of E22 is substantially different in the cyclic peptide compared to the linear peptide. In an embodiment, the cyclic compound comprises an E comprising an O-C-Ca-Cp dihedral angle that is at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, or at least 80 degrees different, than the corresponding dihedral angle in the context of the linear peptide. For example, Table 1 indicates that for simulated linear and cyclic peptides, the difference in this dihedral angle is about 85 degrees.

[00198] Table 1 also identifies differences in the dihedral angle distributions for other angles, including those for example in residues D and V.

[00199] Accordingly in an embodiment the cyclic compound comprises a residue selected from E, D and V, wherein at least one dihedral angle is at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, at least 80 degrees, at least 90 degrees, at least 100 degrees, at least 1 10 degrees, at least 120 degrees, at least 130 degrees or at least 140 degrees different, than the corresponding dihedral angle in the context of the linear compound. For example, for the conformations depicted in FIG. 15 for which dihedral angles are listed in Table 3, the average side-chain dihedral angle difference between the cyclic and linear peptide is given as follows A: 4 degrees, E: 134 degrees, D: 150 degrees, V: 157 degrees.

[00200] The angle difference can for example positive or negative, (+) or (-).

[00201] The alternate conformation can comprise an alternate backbone orientation. For example, the backbone orientation that the cyclic epitope exposes for an antibody differs compared to linear or fibril form.

[00202] FIG. 9 plots the phi and psi angles sampled in equilibrium simulations, for residues

A21 , E22, D23, and V24 in both linear and cyclic peptides consisting of sequence CGEADVG, as well as AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J. From FIG. 9 it is seen that the distribution of backbone dihedral angles in the cyclic peptide is substantially different from the distribution of dihedral angles sampled for either the linear peptide, or for the peptide AEDV (SEQ ID NO: 1 ) in the context the fibril structure 2M4J. Table 2 lists differences for peak values of distributions of backbone phi/psi angles. Similarly Table 3 shows backbone phi/phi angles and the differences for the structures plotted in FIG. 15. For example, for the conformations depicted in FIG. 15 for which dihedral angles are listed in Table 3, the average backbone Ramachandran angle difference between the cyclic and linear peptide is given as follows A: 149 degrees, E: 88 degrees, D: 40 degrees, V: 109 degrees.

[00203] Accordingly, in an embodiment, the cyclic compound comprises at least one residue wherein backbone phi/psi angles is at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, at least 80 degrees different, at least 90 degrees, at least 100 degrees, at least 110 degrees, at least 120 degrees, or at least 130 degrees compared to the corresponding linear peptide or in a fibril PDB structure.

[00204] The alternate conformation can also include an increase in curvature centered around an amino acid or of the cyclic peptide AEDV (SEQ ID NO: 1 ) relative to a linear peptide and/or A-beta fibril.

[00205] In an embodiment, the alternate conformation AEDV (SEQ ID NO: 1 ) has an increased curvature relative to linear AEDV (SEQ ID NO: 1 ). As shown in the Examples, the curvature of the backbone at the positions of D23 and V24 in the cyclic compound is increased relative to the curvature at those positions in the linear peptide, or peptide in the context of the fibril (FIG. 3) as described in Example 3.

[00206] The values of the curvature were determined for A, E, D, V in cyclo(CGAEDVG)

(SEQ ID NO: 4), linear CGAEDVG (SEQ ID NO: 4), and AEDV (SEQ ID NO: 1 ) in the context of the fibril. As described in Example 3, these were:

Cyclic peptide: 0.94, 1.46, 1.55, 1.56

Linear Peptide: 1.42, 1.44, 1.43, 1.14

Fibril: 1.00, 1.32, 0.98, 1.10

The averages of these are:

Cyclic peptide: 1.38

Linear peptide: 1.36

Fibril: 1.10

[00207] Accordingly, the curvature of the D, and/or V in the alternate conformation is increased by at least 0.1 , 0.2, 0.3 or more radians compared to the linear peptide.

[00208] In an embodiment, the DV, EDV and/or AEDV (SEQ ID NO: 1 ) are in an alternate conformation, for example as compared to what is occupied by these residues in a non-oligomeric conformation, such as the linear peptide and/or fibril.

[00209] Further the entropy of the side chains is reduced in the cyclic peptide relative to the linear peptide, rendering the side chains in a more structured conformation than the linear peptide.

[00210] As demonstrated herein, the curvature of the cyclic epitope is for some amino acids different than that in the linear peptide or to the peptide in the context of the fibril (FIG. 3). For example the curvature of V in the context of the cyclic compound CGAEDVG (SEQ ID NO: 4) compared to the corresponding linear peptide is increased.

[00211] Accordingly in one embodiment, the curvature of V in the cyclic compound is increased by at least 10%, at least 20%, or at least 30% compared to a corresponding linear compound.

[00212] It is also demonstrated, that one or more of the dihedral angles in residues E, and/or

D, and/or V are significantly different from the dihedral angles in the linear peptide or peptide in the context of the fibril. For these amino acids, when SASA is weighted by the solubility, more emphasis is placed on the C-terminal residues. In addition, the entropy of the side chains of E is different in the cyclic peptide relative to the linear peptide

[00213] Cyclic compounds which show similar changes are also encompassed. [00214] The cyclic compound in some embodiments that comprises a peptide comprising

EDV or AEDV (SEQ ID NO: 1 ) can include 1 , 2, 3 or more residues in A-beta upstream and/or downstream of AEDV (SEQ ID NO: 1 ). In such cases the spacer is covalently linked to the N- and C- termini of the A-beta residues.

[00215] In some embodiments, the linker or spacer is indirectly coupled to the N- and C- terminus residues of the A-beta peptide.

[00216] In an embodiment, the cyclic compound is a compound in FIG. 14.

[00217] Methods for making cyclized peptides are known in the art and include SS-cyclization or amide cyclization (head-to-tail, or backbone cyclization). Methods are further described in Example 4. For example, a peptide with "C" residues at its N- and C- termini, e.g. CGAEDVGC (SEQ ID NO: 4), can be reacted by SS-cyclization to produce a cyclic peptide.

[00218] As described in Example 3, the cyclic compound of FIG. 14A was assessed for its relatedness to the conformational epitope identified. The cyclic compound comprising AEDV (SEQ ID NO: 1 ) peptide for example can be used to raise antibodies selective for one or more conformation features.

[00219] The epitope AEDV (SEQ ID NO: 1 ) and/or a part thereof, as described herein may be a potential target in misfolded propagating strains of A-beta involved in A-beta, and antibodies that recognize the conformational epitope may for example be useful in detecting such propagating strains.

[00220] Also provided in another aspect is an isolated peptide comprising an A-beta peptide sequence described herein, including linear peptides and cyclic peptides. Linear, uncyclized, peptides can for example be used for selecting antibodies for lack of binding thereto. The isolated peptide can comprise a linker sequence described herein. The linker can be covalently coupled to the N or C terminus or may be partially coupled to the N terminus and partially coupled to the C terminus as in CGAEDVG (SEQ ID NO: 4) linear peptide. In the cyclic peptide, the linker is coupled to the C- terminus and N-terminus directly or indirectly.

[00221] Another aspect includes an immunogen comprising a compound, optionally a cyclic compound described herein. The immunogen may also comprise for example EDV or AEDV (SEQ ID NO: 1 ) or additional A-beta sequence. The amino acids may be directly upstream and/or downstream (i.e. N-terminal and/or C-terminal) of EDV or AEDV (SEQ ID NO: 1 ) or related epitope sequence. Antibodies raised against such immunogens can be selected for example for binding to a cyclopeptide comprising AEDV (SEQ ID NO: 1 ) or a related epitope.

[00222] An immunogen is suitably prepared or formulated for administration to a subject, for example, the immunogen may be sterile, or purified.

[00223] In an embodiment, the immunogen is a cyclic peptide comprising AEDV or a related epitope. [00224] In an embodiment, the immunogen comprises immunogenicity enhancing agent such as Keyhole Limpet Hemocyanin (KLH) or a MAP antigen. The immunogenicity enhancing agent can be coupled to the compound either directly, such as through an amide bound, or indirectly through a functionalizable moiety in the linker. When the linker is a single amino acid residue (for example with the A-beta peptide in the cyclic compound is 6 amino acid residues) the linker can be the functionalizable moiety (e.g. a cysteine residue).

[00225] The immunogen can be produced by conjugating the cyclic compound containing the constrained epitope peptide to an immunogenicity enhancing agent such as Keyhole Limpet Hemocyanin (KLH) or a carrier such bovine serum albumin (BSA) using for example the method described in Lateef et al 2007, herein incorporated by reference. In an embodiment, the method described in Example 3 or 4 is used.

[00226] A further aspect is an isolated nucleic acid molecule encoding the proteinaceous portion of a compound or immunogen described herein.

[00227] In embodiment, the nucleic acid molecule encodes any one of the amino acid sequences sent forth in SEQ ID NOs: 1 -7.

[00228] In an embodiment, nucleic acid molecule encodes AEDV (SEQ ID NO: 1 ) or a related epitope and optionally a linker described herein.

[00229] A further aspect is a vector comprising said nucleic acid. Suitable vectors are described elsewhere herein.

III. Antibodies, Cells and Nucleic Acids

[00230] The compounds and particularly the cyclic compounds described above can be used to raise antibodies that specifically bind DV, EDV or AEDV (SEQ ID NO: 1 ) in A-beta and/or which recognize specific conformations of DV, EDV or AEDV in species of A-beta, for example oligomeric species of A-beta. Similarly cyclic compounds comprising for example AEDVG (SEQ ID NO: 2), AEDVGS (SEQ ID NO: 3), EDVG (SEQ ID NO: 6) and/or other related epitope sequences described herein can be used to raise antibodies that specifically bind AEDV (SEQ I D NO: 1 ) etc and/or specific conformational epitopes thereof. As demonstrated herein, antibodies were raised to cyclo(CGAEDVG) (SEQ ID NO: 4), which specifically and/or selectively bound cyclo(CGAEDVG) (SEQ ID NO: 4) over linear CGAEDVG (SEQ ID NO: 4).

[00231] Accordingly as aspect includes an antibody (including a binding fragment thereof) that specifically binds to an A-beta peptide having of a sequence AEDV (SEQ ID NO: 1 ) or a related epitope sequence, for example as set forth in any one of SEQ ID NOs: 1 to 7.

[00232] In an embodiment, the cyclic compound is a cyclic peptide. In an embodiment, A-beta peptide in the cyclic peptide is any one of SEQ ID NOs: 1-7. In a further embodiment, the cyclic peptide has a sequence as set forth in SEQ ID NOs: 4, 23 or 24. [00233] As described in the examples, antibodies having one or properties can be selected using assays described in the Examples.

[00234] In an embodiment, the antibody does not bind a linear peptide comprising the sequence AEDV (SEQ ID NO: 1 ), optionally wherein the sequence of the linear peptide is a linear version of a cyclic sequence used to raise the antibody, optionally as set forth in SEQ ID NOs: 4, 23 or 24.

[00235] In an embodiment, the antibody is selective for the A-beta peptide as presented in the cyclic compound relative to a corresponding linear compound comprising the A-beta peptide.

[00236] In an embodiment, the antibody specifically binds an epitope on A-beta, the epitope comprising or consisting AEDV (SEQ ID NO: 1 ) or a related epitope thereof.

[00237] In an embodiment, the epitope recognized specifically or selectively by the antibody on

A-beta is a conformational epitope.

[00238] In an embodiment the antibody is isolated.

[00239] In an embodiment, the antibody is an exogenous antibody.

[00240] As described in the Examples, E, D, V and/or DV residues of the identified residues in the cyclic compound may be predominantly accessible or exposed in conformations of A-beta that are distinct from a corresponding linear peptide and/or fibril forms.

[00241] Accordingly a further aspect is an antibody which specifically binds an epitope on A- beta, wherein the epitope comprises or consists of at least one amino acid residue predominantly involved in binding to the antibody, wherein the at least one amino acid is E, D or V embedded within the sequence AEDV (SEQ ID NO: 1 ). In an embodiment, the epitope comprises or consists of at least two consecutive amino acid residues predominantly involved in binding to the antibody, wherein the at least two consecutive amino acids are DV embedded within AEDV (SEQ ID NO: 1 ).

[00242] In another embodiment, the epitope consists of AEDV (SEQ ID NO: 1 ) or a related epitope.

[00243] In an embodiment, the antibody is a conformation selective antibody. I n an embodiment, the antibody selectively binds a cyclic compound comprising an epitope peptide sequence described herein compared to the corresponding linear sequence. For example an antibody that binds a particular epitope conformation can be referred to as a conformation specific antibody. Such antibodies can be selected using the methods described herein. The conformation selective antibody can differentially recognize a particular A-beta species or a group of related species (e.g. dimers, trimers, and other oligomeric species) and can have a higher affinity for one species or group of species compared to another (e.g. to either the monomer or fibril species).

[00244] In an embodiment, the antibody does not specifically bind monomeric A-beta. In an embodiment, the antibody does not specifically bind A-beta senile plaques, for example in situ in AD brain tissue. [00245] In another embodiment, the antibody does not selectively bind monomeric A-beta compared to native- or synthetic- oligomeric A-beta.

[00246] In an embodiment, the antibody specifically binds a cyclic compound comprising an epitope peptide described herein comprising at least one alternate conformational feature described herein (e.g. of the epitope in a cyclic compound compared to a linear compound).

[00247] For example, the antibody may specifically bind a cyclic compound comprises a residue selected from E, D and V, wherein at least one dihedral angle is at least 30 degrees, at least 40 degrees, at least 50 degrees, at least 60 degrees, at least 70 degrees, at least 80 deg rees, at least 90 degrees, at least 100 degrees, at least 1 10 degrees, at least 120 degrees, at least 130 degrees or at least 140 degrees different, than the corresponding dihedral angle in the context of the linear compound.

[00248] In an embodiment, the antibody selectively binds A-beta peptide in a cyclic compound, the A-beta comprising AEDV (SEQ I D NO: 1 ) or a part thereof, relative to a linear peptide comprising AEDV (SEQ ID NO: 1 ), such as a corresponding sequence. For example, in an embodiment the antibody selectively binds AEDV (SEQ ID NO: 1 ) in a cyclic conformation and has at least 2 fold, at least 3 fold, at least 5 fold, at least 10 fold at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 100 fold, at least 500 fold, at least 1000 fold greater selectivity (e.g. greater binding affinity) for AEDV (SEQ ID NO: 1 ) in the cyclic conformation compared to AEDV (SEQ ID NO: 1 ) in a linear peptide, for example as measured by ELISA, or optionally a method described herein.

[00249] In an embodiment, the cyclic compound is cyclo(CGAEDVG) (SEQ ID NO: 4) or the cyclic compound with sequence as set forth in SEQ ID NOs: 23 or 24 and/or having a structure shown in FIG. 14.

[00250] In an embodiment, the antibody selectively binds A-beta peptide in a cyclic compound and/or oligomeric A-beta. In an embodiment, the selectivity is at least 2 fold, at least 3 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 100 fold, at least 500 fold, at least 1000 fold more selective for the A-beta peptide in the cyclic compound and/or A-beta oligomer over a species of A-beta selected from A-beta monomer and/or A-beta fibril and/or a compound comprising a corresponding linear peptide.

[00251] In an embodiment, the antibody lacks A-beta fibril plaque (also referred to as senile plaque) staining. Absence of plaque staining can be assessed by comparing to a positive control such as A-beta-specific antibodies 6E10 and 4G8 (Biolegend, San Diego, CA), or 2C8 (Enzo Life Sciences Inc. , Farmingdale, NY) and an isotype control. An antibody described herein lacks or has negligible A- beta fibril plaque staining if the antibody does not show typical plaque morphology staining and the level of staining is comparable to or no more than 2 fold the level seen with an IgG negative isotype control. The scale can for example set the level of staining with isotype control at 1 and with 6E10 at 10. An antibody lacks A-beta fibril plaque staining if the level of staining on such a scale is 2 or less. In embodiment, the antibody shows minimal A-beta fibril plaque staining, for example on the foregoing scale, levels scored at less about or less than 3.

[00252] In an embodiment, the antibody is produced using a cyclic compound, optionally a cyclic peptide described herein.

[00253] In an embodiment, the antibody is a monoclonal antibody.

[00254] To produce monoclonal antibodies, antibody producing cells (B lymphocytes) can be harvested from a subject immunized with an immunogen described herein, and fused with myeloma cells by standard somatic cell fusion procedures thus immortalizing these cells and yielding hybridoma cells. Such techniques are well known in the art, (e.g. the hybridoma technique originally developed by Kohler and Milstein (Nature 256:495-497 (1975)) as well as other techniques such as the human B-cell hybridoma technique (Kozbor et al. , Immunol. Today 4:72 (1983)), the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al. , Methods Enzymol, 121 : 140-67 (1986)), and screening of combinatorial antibody libraries (Huse et al. , Science 246: 1275 (1989)). Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with the epitope sequences described herein and the monoclonal antibodies can be isolated.

[00255] Specific antibodies, or antibody fragments, reactive against particular antigens or molecules, may also be generated by screening expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria with cell surface components. For example, complete Fab fragments, VH regions and FV regions can be expressed in bacteria using phage expression libraries (see for example Ward et al. , Nature 41 :544-546 (1989); Huse et al. , Science 246: 1275-1281 (1989); and McCafferty et al. , Nature 348:552-554 (1990).

[00256] In an embodiment, the antibody is a humanized antibody.

[00257] The humanization of antibodies from non-human species has been well described in the literature. See for example EP-B1 0 239400 and Carter & Merchant 1997 (Curr Opin Biotechnol 8, 449-454, 1997 incorporated by reference in their entirety herein). Humanized antibodies are also readily obtained commercially (e.g. Scotgen Limited, 2 Holly Road, Twickenham, Middlesex, Great Britain. ).

[00258] Humanized forms of rodent antibodies are readily generated by CDR grafting

(Riechmann et al. Nature, 332:323-327, 1988). In this approach the six CDR loops comprising the antigen binding site of the rodent monoclonal antibody are linked to corresponding human framework regions. CDR grafting often yields antibodies with reduced affinity as the amino acids of the framework regions may influence antigen recognition (Foote & Winter. J Mol Biol, 224: 487-499, 1992). To maintain the affinity of the antibody, it is often necessary to replace certain framework residues by site directed mutagenesis or other recombinant techniques and may be aided by computer modeling of the antigen binding site (Co et al. J Immunol, 152: 2968-2976, 1994). [00259] Humanized forms of antibodies are optionally obtained by resurfacing (Pedersen et al.

J Mol Biol, 235: 959-973, 1994). In this approach only the surface residues of a rodent antibody are humanized.

[00260] Human antibodies specific to a particular antigen may be identified by a phage display strategy (Jespers et al. Bio/Technology, 12: 899-903, 1994). In one approach, the heavy chain of a rodent antibody directed against a specific antigen is cloned and paired with a repertoire of human light chains for display as Fab fragments on filamentous phage. The phage is selected by binding to antigen. The selected human light chain is subsequently paired with a repertoire of human heavy chains for display on phage, and the phage is again selected by binding to antigen. The result is a human antibody Fab fragment specific to a particular antigen. In another approach, libraries of phage are produced were members display different human antibody fragments (Fab or Fv) on their outer surfaces (Dower et al. , WO 91/17271 and McCafferty et al. , WO 92/01047). Phage displaying antibodies with a desired specificity are selected by affinity enrichment to a specific antigen. The human Fab or Fv fragment identified from either approach may be recloned for expression as a human antibody in mammalian cells.

[00261] Human antibodies are optionally obtained from transgenic animals (US Patent Nos.

6, 150,584; 6, 114,598; and 5,770,429). In this approach the heavy chain joining region (JH) gene in a chimeric or germ-line mutant mouse is deleted. Human germ-line immunoglobulin gene array is subsequently transferred to such mutant mice. The resulting transgenic mouse is then capable of generating a full repertoire of human antibodies upon antigen challenge.

[00262] Humanized antibodies are typically produced as antigen binding fragments such as

Fab, Fab' F(ab')2, Fd, Fv and single domain antibody fragments, or as single chain antibodies in which the heavy and light chains are linked by a spacer. Also, the human or humanized antibodies may exist in monomeric or polymeric form. The humanized antibody optionally comprises one non- human chain and one humanized chain (i.e. one humanized heavy or light chain).

[00263] Antibodies, including humanized or human antibodies, are selected from any class of immunoglobulins including: IgM, IgG, IgD, IgA or IgE; and any isotype, including: lgG1 , lgG2, lgG3 and lgG4. A chimeric, humanized or human antibody may include sequences from one or more than one isotype or class.

[00264] Additionally, antibodies specific for the epitopes described herein are readily isolated by screening antibody phage display libraries. For example, an antibody phage library is optionally screened by using cyclic compounds comprising peptides corresponding to epitopes disclosed herein to identify antibody fragments specific for conformation specific antibodies. Antibody fragments identified are optionally used to produce a variety of recombinant antibodies that are useful with different embodiments described herein. Antibody phage display libraries are commercially available, for example, through Xoma (Berkeley, California) Methods for screening antibody phage libraries are well known in the art. [00265] A further aspect is antibody and/or binding fragment thereof comprising a light chain variable region and a heavy chain variable region, the heavy chain variable region comprising complementarity determining regions CDR-H1 , CDR-H2 and CDR-H3, the light chain variable region comprising complementarity determining region CDR-L1 , CDR-L2 and CDR-L3 and with the amino acid sequences of said CDRs comprising the sequences set forth below:

[00266] In an embodiment, the antibody is a monoclonal antibody. In an embodiment, the antibody is a chimeric antibody such as a humanized antibody comprising the CDR sequences as recited in Table 10.

[00267] Also provided in another embodiment, is an antibody comprising the CDRs in Table 10 and a light chain variable region and a heavy chain variable region, optionally in the context of a single chain antibody.

[00268] In yet another aspect, the antibody comprises a heavy chain variable region comprises: i) an amino acid sequence as set forth in SEQ ID NO: 19; ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ ID NO: 19, wherein the CDR sequences are as set forth in SEQ ID NOs: 12, 13 and 14, or iii) a conservatively substituted amino acid sequence i). In another aspect the antibody comprises a light chain variable region comprising i) an amino acid sequence as set forth in SEQ ID NO: 21 , ii) an amino acid sequence with at least 50%, at least 60%, at least 70%, at least 80% or at least 90% sequence identity to SEQ ID NO: 21 , wherein the CDR sequences are as set forth in SEQ ID NOs: 15, 16 and 17, or iii) a conservatively substituted amino acid sequence of i). In another embodiment, the heavy chain variable region amino acid sequence is encoded by a nucleotide sequence as set out in SEQ ID NO: 18 or a codon degenerate optimized version thereof. In another embodiment, the antibody comprises a light chain variable region amino acid sequence encoded by a nucleotide sequence as set out in SEQ ID NO: 20 or a codon degenerate or optimized version thereof. In an embodiment, the heavy chain variable region comprises an amino acid sequence as set forth in SEQ ID NO: 19.

[00269] Another aspect is an antibody that specifically binds a same epitope as the antibody with CDR sequences as recited in Table 10. [00270] Another aspect includes an antibody that competes for binding to human A-beta with an antibody comprising the CDR sequences as recited in Table 10.

[00271] Competition between antibodies can be determined for example using an assay in which an antibody under test is assessed for its ability to inhibit specific binding of a reference antibody to the common antigen. A test antibody competes with a reference antibody if an excess of a test antibody (e.g. , at least a 2 fold, 5, fold, 10 fold or 20 fold) inhibits binding of the reference antibody by at least 50%, at least 75%, at least 80%, at least 90% or at least 95% as measured in a competitive binding assay.

[00272] In an embodiment, the antibody is produced using a cyclic compound described herein.

[00273] A further aspect is an antibody conjugated to a therapeutic, detectable label or cytotoxic agent. In an embodiment, the detectable label is a positron-emitting radionuclide. A positron- emitting radionuclide can be used for example in PET imaging.

[00274] A further aspect relates to an antibody complex comprising an antibody described herein and/or a binding fragment thereof and oligomeric A-beta.

[00275] A further aspect is an isolated nucleic acid encoding an antibody or part thereof described herein.

[00276] Nucleic acids encoding a heavy chain or a light chain are also provided, for example encoding a heavy chain comprising CDR-H1 , CDR-H2 and/or CDR-H3 regions described herein or encoding a light chain comprising CDR-L1 , CDR-L2 and/or CDR-L3 regions described herein.

[00277] The present disclosure also provides variants of the nucleic acid sequences that encode for the antibody and/or binding fragment thereof disclosed herein. For example, the variants include nucleotide sequences that hybridize to the nucleic acid sequences encoding the antibody and/or binding fragment thereof disclosed herein under at least moderately stringent hybridization conditions or codon degenerate or optimized sequences In another embodiment, the variant nucleic acid sequences have at least 50%, at least 60%, at least 70%, most preferably at least 80%, even more preferably at least 90% and even most preferably at least 95% sequence identity to nucleic acid sequences encoding SEQ ID NOs: 18 and 20.

[00278] In an embodiment, the nucleic acid is an isolated nucleic acid.

[00279] Another aspect is an expression cassette or vector comprising the nucleic acid herein disclosed. In an embodiment, the vector is an isolated vector.

[00280] The vector can be any vector, including vectors suitable for producing an antibody and/or binding fragment thereof or expressing a peptide sequence described herein.

[00281] The nucleic acid molecules may be incorporated in a known manner into an appropriate expression vector which ensures expression of the protein. Possible expression vectors include but are not limited to cosmids, plasmids, or modified viruses (e.g. replication defective retroviruses, adenoviruses and adeno-associated viruses). The vector should be compatible with the host cell used. The expression vectors are "suitable for transformation of a host cell", which means that the expression vectors contain a nucleic acid molecule encoding the peptides corresponding to epitopes or antibodies described herein.

[00282] In an embodiment, the vector is suitable for expressing for example single chain antibodies by gene therapy. The vector can be adapted for specific expression in neural tissue, for example using neural specific promoters and the like. In an embodiment, the vector comprises an IRES and allows for expression of a light chain variable region and a heavy chain variable region. Such vectors can be used to deliver antibody in vivo.

[00283] Suitable regulatory sequences may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, or insect genes.

[00284] Examples of such regulatory sequences include: a transcriptional promoter and enhancer or RNA polymerase binding sequence, a ribosomal binding sequence, including a translation initiation signal. Additionally, depending on the host cell chosen and the vector employed, other sequences, such as an origin of replication, additional DNA restriction sites, enhancers, and sequences conferring inducibility of transcription may be incorporated into the expression vector.

[00285] In an embodiment, the regulatory sequences direct or increase expression in neural tissue and/or cells.

[00286] In an embodiment, the vector is a viral vector.

[00287] The recombinant expression vectors may also contain a marker gene which facilitates the selection of host cells transformed, infected or transfected with a vector for expressing an antibody or epitope peptide described herein.

[00288] The recombinant expression vectors may also contain expression cassettes which encode a fusion moiety (i. e. a "fusion protein") which provides increased expression or stability of the recombinant peptide; increased solubility of the recombinant peptide; and aid in the purification of the target recombinant peptide by acting as a ligand in affinity purification, including for example tags and labels described herein. Further, a proteolytic cleavage site may be added to the target recombinant protein to allow separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Typical fusion expression vectors include pGEX (Amrad Corp. , Melbourne, Australia), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the recombinant protein.

[00289] Systems for the transfer of genes for example into neurons and neural tissue both in vitro and in vivo include vectors based on viruses, most notably Herpes Simplex Virus, Adenovirus, Adeno-associated virus (AAV) and retroviruses including lentiviruses. Alternative approaches for gene delivery include the use of naked, plasmid DNA as well as liposome-DNA complexes. Another approach is the use of AAV plasmids in which the DNA is polycation-condensed and lipid entrapped and introduced into the brain by intracerebral gene delivery (Leone et al. US Application No. 2002076394).

[00290] Accordingly, in another aspect, the compounds, immunogens, nucleic acids, vectors and antibodies described herein may be formulated in vesicles such as liposomes, nanoparticles, and viral protein particles, for example for delivery of antibodies, compounds, immunogens and nucleic acids described herein. In particular synthetic polymer vesicles, including polymersomes, can be used to administer antibodies.

[00291] Also provided in another aspect is a cell, optionally an isolated and/or recombinant cell, expressing an antibody described herein or comprising a vector herein disclosed.

[00292] The recombinant cell can be generated using any cell suitable for producing a polypeptide, for example suitable for producing an antibody and/or binding fragment thereof. For example to introduce a nucleic acid (e.g. a vector) into a cell, the cell may be transfected, transformed or infected, depending upon the vector employed.

[00293] Suitable host cells include a wide variety of prokaryotic and eukaryotic host cells. For example, the proteins described herein may be expressed in bacterial cells such as E. coli, insect cells (using baculovirus), yeast cells or mammalian cells.

[00294] In an embodiment, the cell is a eukaryotic cell selected from a yeast, plant, worm, insect, avian, fish, reptile and mammalian cell.

[00295] In another embodiment, the mammalian cell is a myeloma cell, a spleen cell, or a hybridoma cell. Also provided in another embodiment, is a hybridoma cell line.

[00296] In an embodiment, the cell is a neural cell.

[00297] Yeast and fungi host cells suitable for expressing an antibody or peptide include, but are not limited to Saccharomyces cerevisiae, Schizosaccharomyces pombe, the genera Pichia or Kluyveromyces and various species of the genus Aspergillus. Examples of vectors for expression in yeast S. cerivisiae include pYepSed , pMFa, pJRY88, and pYES2 ( Invitrogen Corporation, San Diego, CA). Protocols for the transformation of yeast and fungi are well known to those of ordinary skill in the art.

[00298] Mammalian cells that may be suitable include, among others: COS (e.g. , ATCC No.

CRL 1650 or 1651 ), BHK (e.g. ATCC No. CRL 6281 ), CHO (ATCC No. CCL 61 ), HeLa (e.g., ATCC No. CCL 2), 293 (ATCC No. 1573) and NS-1 cells. Suitable expression vectors for directing expression in mammalian cells generally include a promoter (e.g. , derived from viral material such as polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40), as well as other transcriptional and translational control sequences. Examples of mammalian expression vectors include pCDM8 and pMT2PC.

A further aspect is a hybridoma producing an antibody specific for an epitope described herein.

IV. Compositions [00299] A further aspect is a composition comprising a compound, immunogen, nucleic acid, vector or antibody described herein.

[00300] In an embodiment, the composition comprises a diluent.

[00301] Suitable diluents for nucleic acids include but are not limited to water, saline solutions and ethanol.

[00302] Suitable diluents for polypeptides, including antibodies or fragments thereof and/or cells include but are not limited to saline solutions, pH buffered solutions and glycerol solutions or other solutions suitable for freezing polypeptides and/or cells.

[00303] In an embodiment, the composition is a pharmaceutical composition comprising any of the peptides, immunogens, antibodies, nucleic acids or vectors disclosed herein, and optionally comprising a pharmaceutically acceptable carrier.

[00304] The compositions described herein can be prepared by per se known methods for the preparation of pharmaceutically acceptable compositions that can be administered to subjects, optionally as a vaccine, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.

[00305] Pharmaceutical compositions include, without limitation, lyophilized powders or aqueous or non-aqueous sterile injectable solutions or suspensions, which may further contain antioxidants, buffers, bacteriostats and solutes that render the compositions substantially compatible with the tissues or the blood of an intended recipient. Other components that may be present in such compositions include water, surfactants (such as Tween), alcohols, polyols, glycerin and vegetable oils, for example. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, tablets, or concentrated solutions or suspensions. The composition may be supplied, for example but not by way of limitation, as a lyophilized powder which is reconstituted with sterile water or saline prior to administration to the patient.

[00306] Pharmaceutical compositions may comprise a pharmaceutically acceptable carrier. Suitable pharmaceutically acceptable carriers include essentially chemically inert and nontoxic compositions that do not interfere with the effectiveness of the biological activity of the pharmaceutical composition. Examples of suitable pharmaceutical carriers include, but are not limited to, water, saline solutions, glycerol solutions, ethanol, N-(1 (2,3-dioleyloxy)propyl)N, N, N-trimethylammonium chloride (DOTMA), diolesylphosphotidyl-ethanolamine (DOPE), and liposomes. Such compositions should contain a therapeutically effective amount of the compound, together with a suitable amount of carrier so as to provide the form for direct administration to the patient.

[00307] The composition may be in the form of a pharmaceutically acceptable salt which includes, without limitation, those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc. , and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylarnino ethanol, histidine, procaine, etc. [00308] In an embodiment comprising a compound or immunogen described herein, the composition comprises an adjuvant.

[00309] Adjuvants that can be used for example, include intrinsic adjuvants (such as lipopolysaccharides) that normally are the components of killed or attenuated bacteria used as vaccines. Extrinsic adjuvants are immunomodulators which are typically non-covalently linked to antigens and are formulated to enhance the host immune responses. Aluminum hydroxide, aluminum sulfate and aluminum phosphate (collectively commonly referred to as alum) are routinely used as adjuvants. A wide range of extrinsic adjuvants can provoke potent immune responses to immunogens. These include saponins such as Stimulons (QS21 , Aquila, Worcester, Mass. ) or particles generated therefrom such as ISCOMs and (immunostimulating complexes) and ISCOMATRIX, complexed to membrane protein antigens (immune stimulating complexes), pluronic polymers with mineral oil, killed mycobacteria and mineral oil, Freund's complete adjuvant, bacterial products such as muramyl dipeptide (MDP) and lipopolysaccharide (LPS), as well as lipid A, and liposomes.

[00310] In an embodiment, the adjuvant is aluminum hydroxide. In another embodiment, the adjuvant is aluminum phosphate. Oil in water emulsions include squalene; peanut oil; MF59 (WO 90/14387); SAF (Syntex Laboratories, Palo Alto, Calif. ); and Ribi™ (Ribi Immunochem, Hamilton, Mont. ). Oil in water emulsions may be used with immunostimulating agents such as muramyl peptides (for example, N-acetylmuramyl-L-threonyl-D-isoglutamine (thr-MDP), -acetyl-normuramyl-L-alanyl-D- isoglutamine (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutamyl-L-alanine-2-(1 '-2'dipalmitoyl-sn- glycero-3-hydroxyphosphoryloxy)-ethylamine ( MTP-PE), N-acetylglucsaminyl-N-acetylmuramyl-L-AI- D-isoglu-L-Ala-dipalmitoxy propylamide (DTP-DPP) theramide(TM)), or other bacterial cell wall components.

[00311] The adjuvant may be administered with an immuogen as a single composition.

Alternatively, an adjuvant may be administered before, concurrent and/or after administration of the immunogen.

[00312] Commonly, adjuvants are used as a 0.05 to 1.0 percent solution in phosphate - buffered saline. Adjuvants enhance the immunogenicity of an immunogen but are not necessarily immunogenic themselves. Adjuvants may act by retaining the immunogen locally near the site of administration to produce a depot effect facilitating a slow, sustained release of immunogen to cells of the immune system. Adjuvants can also attract cells of the immune system to an immunogen depot and stimulate such cells to elicit immune responses. As such, embodiments may encompass compositions further comprising adjuvants.

[00313] Adjuvants for parenteral immunization include aluminum compounds (such as aluminum hydroxide, aluminum phosphate, and aluminum hydroxy phosphate). The antigen can be precipitated with, or adsorbed onto, the aluminum compound according to standard protocols. Other adjuvants such as RIBI (ImmunoChem, Hamilton, MT) can also be used in parenteral administration. [00314] Adjuvants for mucosal immunization include bacterial toxins (e.g., the cholera toxin

(CT), the E. coli heat-labile toxin (LT), the Clostridium difficile toxin A and the pertussis toxin (PT), or combinations, subunits, toxoids, or mutants thereof). For example, a purified preparation of native cholera toxin subunit B (CTB) can be of use. Fragments, homologs, derivatives, and fusion to any of these toxins are also suitable, provided that they retain adjuvant activity. Preferably, a mutant having reduced toxicity is used. Suitable mutants have been described (e.g., in WO 95/1721 1 (Arg-7-Lys CT mutant), WO 96/6627 (Arg-192-Gly LT mutant), and WO 95/34323 (Arg-9-Lys and Glu-129-Gly PT mutant)). Additional LT mutants that can be used in the methods and compositions include, for example Ser-63-Lys, Ala-69-Gly, Glu-1 10-Asp, and Glu-1 12-Asp mutants. Other adjuvants (such as a bacterial monophosphoryl lipid A (MPLA) of various sources (e.g. , E. coli, Salmonella minnesota, Salmonella typhimurium, or Shigella flexneri, saponins, or polylactide glycolide (PLGA) microspheres) can also be used in mucosal administration.

[00315] Other adjuvants include cytokines such as interleukins for example IL-1 , IL-2 and IL-

12, chemokines, for example CXCL10 and CCL5, macrophage stimulating factor, and/or tumor necrosis factor. Other adjuvants that may be used include CpG oligonucleotides (Davis. Curr Top Microbiol Immunol. , 247: 171 -183, 2000).

[00316] Oil in water emulsions include squalene; peanut oil; MF59 (WO 90/14387); SAF

(Syntex Laboratories, Palo Alto, Calif. ); and Ribi™ (Ribi Immunochem, Hamilton, Mont. ). Oil in water emulsions may be used with immunostimulating agents such as muramyl peptides (for example, N- acetylmuramyl-L-threonyl-D-isoglutamine (thr-MDP), -acetyl-normuramyl-L-alanyl-D-isoglutamine (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutamyl-L-alanine-2-(1 '-2'dipalmitoyl-sn-glycero-3- hydroxyphosphoryloxy)-ethylamine (MTP-PE), N-acetylglucsaminyl-N-acetylmuramyl-L-AI-D-isoglu-L- Ala-dipalmitoxy propylamide (DTP-DPP) theramide(TM)), or other bacterial cell wall components.

[00317] Adjuvants useful for both mucosal and parenteral immunization include polyphosphazene (for example, WO 95/2415), DC-chol (3 b-(N-(N', N'-dimethyl aminomethane)- carbamoyl) cholesterol (for example, U.S. Patent No. 5,283, 185 and WO 96/14831 ) and QS-21 (for example, WO 88/9336).

[00318] An adjuvant may be coupled to an immunogen for administration. For example, a lipid such as palmitic acid, may be coupled directly to one or more peptides such that the change in conformation of the peptides comprising the immunogen does not affect the nature of the immune response to the immunogen.

[00319] In an embodiment, the composition comprises an antibody described herein. In another embodiment, the composition comprises an antibody described herein and a diluent. In an embodiment, the composition is a sterile composition.

V. Kits

[00320] A further aspect relates to a kit comprising i) an antibody and/or binding fragment thereof, ii) a nucleic acid, iii) peptide or immunogen, iv) composition or v) recombinant cell described herein, comprised in a vial such as a sterile vial or other housing and optionally a reference agent and/or instructions for use thereof.

[00321] In an embodiment, the kit further comprises one or more of a collection vial, standard buffer and detection reagent.

VI. Methods

[00322] Included are methods for making and using the compounds, immunogens and antibodies described herein.

[00323] In particular, provided are methods of making an antibody specific and/or selective for a conformational epitope of AEDV (SEQ ID NO: 1 ) or related epitope comprising administering to a subject, optionally a non-human subject, a conformationally restricted compound comprising an epitope sequence described herein, optionally cyclic compound comprising AEDV (SEQ ID NO: 1 ) or related epitope, and isolating antibody producing cells or antibodies that specifically or selectively bind the cyclic compound and optionally i) specifically or selectively bind synthetic and/or native oligomers and/or that have no or negligible senile plaque binding in situ tissue samples or no or negl igible binding to a corresponding linear peptide. The cyclic compound can for example comprise any of the "epitopes" described herein containing cyclic compounds described herein.

[00324] In an embodiment, the method is for making a monoclonal antibody using for example a method as described herein.

[00325] In an embodiment, the method is for making a humanized antibody using for example a method described herein.

[00326] Antibodies produced using a cyclic compound are selected as described herein and in the Examples. In an embodiment, the method comprises isolating antibodies that specifically or selectively bind cyclic peptide over linear peptide, are specific for the epitope sequence, specifically bind oligomer and/or lack or negligibly bind plaque in situ and/or corresponding linear peptide, optionally using a method described herein.

[00327] A further aspect provides a method of detecting whether a biological sample comprises A-beta, the method comprising contacting the biological sample with an antibody described herein and detecting the presence of any antibody complex. In an embodiment, the method is for detecting whether a biological sample comprises A-beta wherein at least D and/or V is in an alternate conformation than occupied by D and/or V in a non-oligomeric conformation.

[00328] In an embodiment the method is for detecting whether the biologic sample comprises oligomeric A-beta.

[00329] In an embodiment, the method comprises:

a. contacting the biologic sample with an antibody described herein that is specific and/or selective for A-beta oligomer herein under conditions permissive to produce an antibody:A- beta oligomer complex; and b. detecting the presence of any complex;

wherein the presence of detectable complex is indicative that the sample may contain A-beta oligomer.

[00330] In an embodiment, the level of complex formed is compared to a test antibody such as a suitable Ig control or irrelevant antibody.

[00331] In an embodiment, the detection is quantitated and the amount of complex produced is measured. The measurement can for example be relative to a standard.

[00332] In an embodiment, the measured amount is compared to a control.

[00333] In another embodiment, the method comprises:

(a) contacting a biological sample of said subject with an antibody described herein, under conditions permissive to produce an antibody-antigen complex;

(b) measuring the amount of the antibody-antigen complex in the test sample; and

(c) comparing the amount of antibody-antigen complex in the test sample to a control; wherein detecting antibody-antigen complex in the biological sample as compared to the control indicates that the sample comprises A-beta.

[00334] The control can be a sample control (e.g. from a subject without AD, or from a subject with a particular form of AD, mild, moderate or advanced), or be a previous sample from the same subject for monitoring changes in A-beta oligomer levels in the subject.

[00335] In an embodiment, an antibody described herein is used.

[00336] In an embodiment, the antibody specifically and/or selectively recognizes a conformation of A-beta comprising an AEDV (SEQ I D NO: 1 ) or related conformational epitope, and detecting the antibody antigen complex in the biological sample is indicative that sample comprises A- beta oligomer.

[00337] In an embodiment, the sample is a biological sample. In an embodiment, the sample comprises brain tissue or an extract thereof and/or CSF. In an embodiment, the sample comprises whole blood, plasma or serum. In an embodiment, the sample is obtained from a human subject. In an embodiment, the subject is suspected of, at a risk of or has AD.

[00338] A number of methods can be used to detect an A-beta: antibody complex and thereby determine if A-beta comprising a AEDV (SEQ ID NO: 1 ) or related conformational epitope and/or A- beta oligomers is present in the biological sample using the antibodies described herein, including immunoassays such as flow cytometry, immunoblots, ELISA, and immunoprecipitation followed by SDS-PAGE, and immunocytochemistry.

[00339] As described in the Examples surface plasmon resonance technology can be used to assess conformation specific binding. If the antibody is labelled or a detectably labelled secondary antibody specific for the complex antibody is used, the label can be detected. Commonly used reagents include fluorescent emitting and H P labelled antibodies. In quantitative methods, the amount of signal produced can be measured by comparison to a standard or control. The measurement can also be relative.

[00340] A further aspect includes a method of measuring a level of- or imaging A-beta in a subject or tissue, optionally where the A-beta to be measured or imaged is oligomeric A-beta. In an embodiment, the method comprises administering to a subject at risk or suspected of having or having AD, an antibody conjugated to a detectable label; and detecting the label, optionally quantitatively detecting the label. The label in an embodiment is a positron emitting radionuclide which can for example be used in PET imaging.

[00341] A further aspect includes a method of inducing an immune response in a subject, comprising administering to the subject a compound described herein, optionally a cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope peptide sequence, an immunogen and/or composition comprising said compound or said immunogen; and optionally isolating cells and/or antibodies that specifically or selectively bind the A-beta peptide in the compound or immunogen administered. In an embodiment, the composition is a pharmaceutical composition comprising the compound or immunogen in admixture with a pharmaceutically acceptable, diluent or carrier.

[00342] In an embodiment, the subject is a non-human subject such as a rodent. Antibody producing cells generated are used in an embodiment to produce a hybridoma cell line.

[00343] In an embodiment, the immunogen administered comprises a compound illustrated in

FIG. 14.

[00344] It is demonstrated herein that antibodies raised against cyclo(CGAEDVG) (SEQ ID

NO: 4), can specifically and/or selectively bind A-beta oligomers and lack A-beta plaque staining. Oligomeric A-beta species are believed to be the toxic propagating species in AD. Further, antibody raised using cyclo(CGAEDVG) (SEQ ID NO: 4) and specific for oligomers, inhibited A-beta aggregation and A-beta oligomer propagation. Accordingly, also provided are methods of inhibiting A- beta oligomer propagation, the method comprising contacting a cell or tissue expressing A-beta with or administering to a subject in need thereof an effective amount of an A-beta oligomer specific or selective antibody described herein to inhibit A-beta aggregation and/or oligomer propagation. In vitro the assay can be monitored as described in Example 12.

[00345] The antibodies may also be useful for treating AD and/or other A-beta amyloid related diseases. For example, variants of Lewy body dementia and in inclusion body myositis (a muscle disease) exhibit similar plaques as AD in the brain and muscle respectively, and A-beta can also form in aggregates implicated in cerebral amyloid angiopathy. Moreover, "mixed" pathology in neurodegenerative diseases (including Parkinson's disease and frontotemporal dementia) is recognized in which features of AD pathology can be observed without a frank AD clinical syndrome. As mentioned, antibodies raised to cyclo(CGAEDVG) (SEQ ID NO: 4) bind oligomeric A-beta which is believed to be a toxigenic species of A-beta in AD and inhibit formation of toxigenic A-beta oligomers.

45

RECTIFIED SHEET (RULE 91.1) [00346] Accordingly a further aspect is a method of treating AD and/or other A-beta amyloid related diseases, the method comprising administering to a subject in need thereof i) an effective amount of an antibody described herein, optionally an A-beta oligomer specific or selective or a pharmaceutical composition comprising said antibody; or 2) administering an isolated cyclic compound comprising AEDV (SEQ ID NO: 1 ) or a related epitope sequence or immunogen or pharmaceutical composition comprising said cyclic compound, to a subject in need thereof.

[00347] In an embodiment, a biological sample from the subject to be treated is assessed for the presence or levels of A-beta using an antibody described herein. In an embodiment, a subject with detectable A-beta levels (e.g. A-beta antibody complexes measured in vitro or measured by imaging) is treated with the antibody.

[00348] The antibody and immunogens can for example be comprised in a pharmaceutical composition as described herein, and formulated for example in vesicles for improving delivery.

[00349] One or more antibodies targeting AEDV (SEQ ID NO: 1 ) and/or related antibodies can be administered in combination. In addition the antibodies disclosed herein can be administered with one or more other treatments such as a beta-secretase inhibitor or a cholinesterase inhibitor.

[00350] In an embodiment, the antibody is a conformation specific/selective antibody, optionally that specifically or selectively binds A-beta oligomer.

[00351] Also provided are uses of the compositions, antibodies, isolated peptides, immunogens and nucleic acids for treating AD.

[00352] The compositions, compounds, antibodies, isolated peptides, immunogens and nucleic acids, vectors etc. described herein can be administered for example, by parenteral, intravenous, subcutaneous, intramuscular, intracranial, intraventricular, intrathecal, intraorbital, ophthalmic, intraspinal, intracisternal, intraperitoneal, intranasal, aerosol or oral administration.

[00353] In certain embodiments, the pharmaceutical composition is administered systemically.

[00354] In other embodiments, the pharmaceutical composition is administered directly to the brain or other portion of the CNS. For example such methods include the use of an implantable catheter and a pump, which would serve to discharge a pre-determined dose through the catheter to the infusion site. A person skilled in the art would further recognize that the catheter may be implanted by surgical techniques that permit visualization of the catheter so as to position the catheter adjacent to the desired site of administration or infusion in the brain. Such techniques are described in Elsberry et al. U.S. Patent 5,814,014 "Techniques of Treating Neurodegenerative Disorders by Brain I nfusion", which is herein incorporated by reference. Also contemplated are methods such as those described in US patent application 20060129126 (Kaplitt and During "Infusion device and method for infusing material into the brain of a patient". Devices for delivering drugs to the brain and other parts of the CNS are commercially available (e.g. SynchroMed ® EL Infusion System; Medtronic, Minneapolis, Minnesota). [00355] In another embodiment, the pharmaceutical composition is administered to the brain using methods such as modifying the compounds to be administered to allow receptor-mediated transport across the blood brain barrier.

[00356] Other embodiments contemplate the co-administration of the compositions, compounds, antibodies, isolated peptides, immunogens and nucleic acids described herein with biologically active molecules known to facilitate the transport across the blood brain barrier.

[00357] Also contemplated in certain embodiments, are methods for administering the compositions, compounds, antibodies, isolated peptides, immunogens and nucleic acids described herein across the blood brain barrier such as those directed at transiently increasing the permeability of the blood brain barrier as described in US patent 7012061 "Method for increasing the permeability of the blood brain barrier", herein incorporated by reference.

[00358] A person skilled in the art will recognize the variety of suitable methods for administering the compositions, compounds, antibodies, isolated peptides, immunogens and nucleic acids described herein directly to the brain or across the blood brain barrier and be able to modify these methods in order to safely administer the products described herein.

[00359] The above disclosure generally describes the present application. A more complete understanding can be obtained by reference to the following specific examples. These examples are described solely for the purpose of illustration and are not intended to limit the scope of the application. Changes in form and substitution of equivalents are contemplated as circumstances might suggest or render expedient. Although specific terms have been employed herein, such terms are intended in a descriptive sense and not for purposes of limitation.

[00360] The following non-limiting examples are illustrative of the present disclosure:

Examples

Example 1

I. Go MODEL METHOD FOR PREDICTING A-BETA OLIGOMER SPECIFIC EPITOPES

[00361] One epitope prediction model is based on the free energy landscape of partial protein unfolding from the native state. The native state is taken to be an experimentally-derived fibril structure. When the protein is partially unfolded from the native state by a given amount of primary sequence, epitope candidates are contiguous sequence segments that cost the least free energy to disorder. The free energy of a given protein conformation arises from several contributions, including conformational entropy and solvation of polar functional groups that favor the unfolded state, as well as the loss of electrostatic and van der Waals intra-protein interactions that enthalpically stabilize the native state.

A. Go -like Model of Protein Partially Unfolding Landscape

[00362] An approximate model to account for the free energetic changes that take place during unfolding assigns a fixed energy to all contacts in the native state, where a contact is defined as a pair of heavy (non-hydrogen) atoms within a fixed cut-off distance r cu tof f Go-like models have been successfully implemented in previous studies of protein folding. The Go- like model isolates the effects arising from the topology of native protein interactions, and in practice the unfolding free energy landscape can be readily calculated from a single native state structure.

[00363] The total free energy cost of unfolding a segment depends on the number of interactions to be disrupted, together with the conformational entropy term of the unfolded region.

[00364] In the following equations, lower case variables refer to atoms, while upper case variables refer to residues. Let T be the set of all residues in the protein, U be the set of residues unfolded in the protein, and F be the subset of residues folded in the protein (thus T = U uF ). The unfolding mechanism at high degrees of nativeness consists of multiple contiguous strands of disordered residues. Here the approximation of a single contiguous unfolded strand was adopted, and the free energy cost to disorder this contiguous strand was calculated.

[00365] The total free energy change AFQ^ (U ) for unfolding the set of residues U is

AFQ O (U ) = AE G o (U ) - TAS G - 0 (U ) ( 1 )

[00366] The unfolding enthalpy function AEQ~^ {U ) is given by the number of interactions disrupted by unfolding of the set of U residues: i>j

/·.; ,·„; ∑ θ( Γ ∞ίο// - | Γί - | ) (2)

Atoms i ΐ .;■ / ' /

[00367] In Equation 2, the sum on ,j is over all unique pairs of heavy atoms that have either one or both atoms in the unfolded region, 17 and l are the coordinates of atoms / and j, r cu toff (taken to be 4.8A) is the interaction distance cut-off. Θ(χ) is the Heaviside function defined by Θ(χ) = 1 if x is positive and 0 otherwise. The energy per contact a may be chosen to recapitulate the overall experimental stability AFExp{U )\u = T on completely unfolding the protein at room temperature:

[00368] The results do not depend on this value; it merely sets the overall global energy scale in the problem. In the present model, this free energy was taken to be a constant number equal to 4.6 kcal/mol. This value is not a primary concern as it is the relative free energy cost for the different regions of the same protein that is sought to be disordered in the method of epitope prediction. [00369] The calculation of the unfolding entropy term ASQ 0 (U ) is discussed in B below.

B. Entropy calculation

[00370] The number of microstates accessible to the protein in the unfolded state is much greater than the number accessible in the native state, so there is a favorable gain of conformational entropy on unfolding. T e total entropy of the unfolding segment U by summing over all the residues K in the unfolded region is calculated = { Sbb.K— (1— ,A ) Sbu→eo!.K + 3 ex→so i < ) W where ASbb ,K · ^bu→ex,K · &Sex→sol, K are the three conformational entropic components of residue K as listed in reference [3]: ASbb,K is the backbone entropy change from native state to unfolded state, ASbu→ex, K is the entropy change for side-chain from buried inside protein to the surface of the protein, and, and AS ex so i t K is the entropy obtained for the side-chain from the surface to the solution.

[00371] A correction is applied to the unfolded state conformational entropies, since in the single sequence approximation the end points of the partially unfolded strand are fixed in their positions in the native structure. This means that there is a loop entropy penalty to be paid for constraining the ends in the partially unfolded structure, which is not present in the fully unfolded state Sreturn = ~ t< B lll( fw (R| Λ Γ ) Δτ ) (5)

[00372] Here f W (R\N)Ar is found by calculating the probability an ideal random walk returns to a box of volume AT centered at position R after N steps, without penetrating back into the protein during the walk. For strand lengths shorter than about n ~ 20 residues, the size of the melted strand is much smaller than the protein diameter and the steric excluded volume of the protein is well treated as an impenetrable plane. The number of polymeric states of the melted strand must be multiplied by the fraction of random walks that travel from an origin on the surface of the protein to a location where the melted polymer re-enters the protein without touching or crossing the impenetrable plane. The above fraction of states can be written in the following form: where R is the end to end distance between the exit and entrance locations, N is the number of residues of the melted region, and a, I, V c are parameters determined by fitting to unfolded polypeptide simulations. The parameter / is the effective arc length between two C a atoms, and V c is the average excluded volumes for each residue. By fitting the Equation 6 into the simulation results, the values of the parameters a = 0.0217, / = 4.867, V c = 3.291 are obtained. This entropy penalty is general and independent of the sequence.

[00373] Disulfide bonds require additional consideration in the loop entropy term since they further restrict the motion of the unfolded segment. When present, the disulfide is treated as an additional node through which the loop must pass, in effect dividing the full loop into two smaller loops both subject to the boundary conditions described above.

C. Epitope prediction from Free Energy Landscape

[00374] Once the free energy landscape of partially unfolding the protein is obtained, a variable energy threshold Efh is applied, and the segments that contains no fewer than 3 amino acids and with free energy cost below the threshold are predicted as epitope candidates. The prediction is stable with respect to varying the threshold value Ef

[00375] Epitopes predicted using this method are described in Example 3.

Example 2

Mi. COLLECTIVE COORDINATES PREDICTIONS

[00376] A second method for predicting misfolded epitopes is provided by a method referred to as "Collective Coordinates biasing" which is described in US Patent Application serial no. 62/253044, SYSTEMS AND METHODS FOR PREDICTING MISFOLDED PROTEIN EPITOPES BY COLLECTIVE COORDINATE BIASING filed November 9, 2015, and is incorporated herein by reference. As described therein, the method uses molecular-dynamics-based simulations which impose a global coordinate bias on a protein (or peptide-aggregate) to force the protein (or peptide- aggregate) to misfold and then predict the most likely unfolded regions of the partially unstructured protein (or peptide aggregate). Biasing simulations were performed and the solvent accessible surface area (SASA) corresponding to each residue index (compared to that of the initial structure of the protein under consideration). SASA represents a surface area that is accessible to H20. A positive change in SASA (compared to that of the initial structure of the protein under consideration) may be considered to be indicative of unfolding in the region of the associated residue index. The method was applied to three A-beta strains, each with its own morphology: a three-fold symmetric structure of A/3-40 peptides (or monomers) (PDB entry 2M4J), a two-fold symmetric structure of A/3-40 monomers (PDB entry 2LMN), and a single-chain, parallel in-register (e.g. a repeated beta sheet where the residues from one chain interact with the same residues from the neighboring chains) structure of A/3-42 monomers (PDB entry 2MXU). [00377] Simulations were performed for each initial structure using the collective coordinates method as described in US Patent Application serial no. 62/253044, SYSTEMS AND METHODS FOR PREDICTING MISFOLDED PROTEIN EPITOPES BY COLLECTIVE COORDINATE BIASING and the CHARMM force-field parameters described in: K. Vanommeslaeghe, E. Hatcher, C.Acharya, S. Kundu, S. Zhong, J. Shim, E. Darian, O. Guvench, P. Lopes, I. Vorobyov, and A. D. Mackerell. Charmm general force field: A force field for drug-like molecules compatible with the charmm all-atom additive biological force fields. Journal of Computational Chemistry, 31 (4):671-690, 2010; and P. Bjelkmar, P. Larsson, M. A. Cuendet, B. Hess, and E. Lindahl. Implementation of the CHARM M force field in GROMACS: analysis of protein stability effects from correlation maps, virtual interaction sites, and water models. J. Chem. Theo. Comp. , 6:459-466, 2010, both of which are hereby incorporated herein by reference, with TIP3P water.

[00378] Epitopes predicted using this method are described in Example 3.

Example 3

I. OLIGOMER-SELECTIVE EPITOPE - AEDV (SEQ ID NO: 1 )

[00379] This disclosure pertains to antibodies that may be selective for oligomeric A-beta peptide and particularly to toxic oligomers of Αβ peptide, a species of misfolded protein whose prion- like propagation and interference with synaptic vesicles are believed to be responsible for the synaptic dysfunction and cognitive decline that occurs in Alzheimer's disease (AD). Αβ is a peptide of length 36-43 amino acids that results from the cleavage of amyloid precursor protein (APP) by gamma secretase. In AD patients, it is present in monomers, fibrils, and in soluble oligomers. Αβ is the main component of the amyloid plaques found in the brains of AD patients.

[00380] In monomer form, Αβ exists as an unstructured polypeptide chain. In fibril form, Αβ can aggregate into distinct morphologies, often referred to as strains. Several of these structures have been determined by solid-state N MR- some fibril structures have been obtained from in vitro studies, and others obtained by seeding fibrils using amyloid plaques taken from AD patients.

[00381] The oligomer is suggested to be a toxic and propagative species of the peptide, recruiting and converting monomeric Αβ to oligomers, and eventually fibrils.

[00382] A prerequisite for the generation of oligomer-specific antibodies is the identification of targets on Αβ peptide that are not present on either the monomer or fibril. These oligomer-specific epitopes would not differ in primary sequence from the corresponding segment in monomer or fibril, however they would be conformationally distinct in the context of the oligomer. That is, they would present a distinct conformation in the oligomer that would not be present in the monomer or fibril.

[00383] The structure of the oligomer has not been determined to date, moreover, N MR evidence indicates that the oligomer exists not in a single well-defined structure, but in a conformationally-plastic, malleable structural ensemble with limited regularity. Moreover, the concentration of oligomer species is far below either that of the monomer or fibril (estimates vary but on the order of 1000-fold below or more), making this target elusive.

[00384] Antibodies directed either against contiguous strands of primary sequence (e.g. , linear sequence), or against fibril structures, may suffer from several problems limiting their efficacy. Antibodies raised to linear peptide regions tend not to be selective for oligomer, and thus bind to monomer as well. Because the concentration of monomer is substantially higher than that of oligomer, such antibody therapeutics may suffer from "target distraction", primarily binding to monomer and promoting clearance of functional Αβ, rather than selectively targeting and clearing oligomeric species. Antibodies raised to amyloid inclusions bind primarily to fibril, and have resulted in amyloid related imaging abnormalities (ARIA), including signal changes thought to represent vasogen ic edema and/or microhemorrhages.

[00385] To develop antibodies selective for oligomeric forms of Αβ, a region that may be disrupted in the fibril was identified. Without wishing to be bound to theory, it was hypothesized that disruptions in the context of the fibril may be exposed as well on the surface of the oligomer. On oligomers however, these sequence regions may be exposed in conformations distinct from either that of the monomer and/or that of the fibril. For example, being on the surface, they may be exposed in turn regions that have higher curvature, higher exposed surface area, and different dihedral angle distribution than the corresponding quantities exhibit in either the fibril or the monomer.

[00386] Cyclic peptides comprising the predicted sequences are described herein and shown in FIG. 14. The cyclic compounds have been designed to satisfy one or more of the above criteria of higher curvature, higher exposed surface area, and alternative dihedral angle distributions.

[00387] A potential benefit of identifying regions prone to disruption in the fibril is that it may identify regions involved in secondary nucleation processes where fibrils may act as a catalytic substrate to nucleate oligomers from monomers [3]. Regions of fibril with exposed side chains may be more likely to engage in aberrant interactions with nearby monomer, facilitating the accretion of monomers; such accreted monomers would then experience an environment of effectively increased concentration at or near the surface of the fibril, and thus be more likely to form multimeric aggregates including oligomers. Aged or damaged fibril with exposed regions of Αβ may enhance the production of toxic oligomer, and that antibodies directed against these disordered regions on the fibril could be effective in blocking such propagative mechanisms.

II. EPITOPES PREDICTIONS FROM A-beta DISRUPTED FIBRIL STRUCTURES

[00388] From the native fibril structure 2M4J, the unfolding free energy landscape was calculated using the Go-like model as described in Example 1. The result is shown in FIG. 1. The epitope AEDV (SEQ ID NO: 1 ) emerges as a candidate epitope in this analysis.

[00389] FIG. 1 shows a top view of the free energy landscape with a free energy cost for local protein unfolding of E th = 1 1 kcal/mol, and E th = 11.5kcal/mol, respectively. The x-axis in these plots represents the center of the segment, the y-axis represents the length of the segment, and the depth of shading (values also provided) represents the free energy needed to unfold the corresponding segment. From FIG. 1 , at the energy scale of roughly 11 kcal/mol, the epitope consisting of residues 21 -24 or sequence AEDV (SEQ ID NO: 1 ) emerges as a predicted epitope in chains A, B and C of the structure PDB 2M4J.

[00390] Likewise, for Αβ fibril structure 2MXU, an epitope of sequence 21-25 emerges for chain C (sequence AEDVG (SEQ ID NO: 2), FIG. 2); at a slightly higher energy cost epitope AEDVGS (SEQ ID NO: 3) emerges. These epitopes have low free energy to disorder with respect their surroundings.

III. CURVATURE OF THE CYCLIC PEPTIDE

[00391] The curvature profile of the cyclic peptide CGAEDVG (SEQ I D NO: 4) differentiates cyclic AEDV (SEQ ID NO: 1 ) from either the linear peptide or the fibril. The curvature profiles are shown in Figure 3. The alanine has a lower curvature than either the linear peptide or the fibril. The linear peptide tends to bend the backbone compared to the conformations explored by either the cyclic peptide or fibril. The glutamic acid residue (E) appears to be comparable between the linear peptide and the cyclic peptide on the other hand, while the curvature centered on this residue appears to be somewhat higher in the fibril. The aspartic acid (D) in the cyclic peptide has somewhat higher curvature than that in the linear peptide, and substantially higher curvature than that in the fibril. Likewise, the valine has substantially larger curvature in the cyclic peptide than either the linear peptide or fibril. These results imply that an antibody directed against the cyclic peptide could show selectivity for a species presenting a different conformational ensemble than that of either the monomer or fibril.

[00392] For the plots of curvature, and dihedral angle distributions discussed herein, the data are obtained from equilibrium simulations in explicit solvent (SPC) using the Charmm27. The simulation time and number of configurations for each ensemble are as follows. Cyclic peptide ensemble: simulation time 10ns, containing 1000 frames linear peptide ensemble: simulation time 10ns, containing 1000 frames 2m4j ensemble: 680ps, containing 68 frames.

Because the curvature of the cyclic epitope has a different profile than either the linear peptide or fibril, it is expected that the corresponding stretch of amino acids on an oligomer containing these residues would have a backbone orientation that is distinct from that in the fibril or monomer. However the degree of curvature would not be unphysical - values of curvature characterizing the cyclic peptide are obtained in several locations of the fibril.

IV. DIHEDRAL ANGLE DISTRIBUTIONS

[00393] Further computational support for the identification of an oligomer-selective epitope, is provided by both the side chain dihedral angle distributions, and the Ramachandran ° and ' distributions for the backbone dihedral angles in the cyclic peptide a proxy for an exposed epitope in the oligomer- are substantially different from the corresponding distributions in either the fibril or monomer.

[00394] The dihedral distributions can be examined for A, E, D, and V. The distribution of the 0-C-C a -Cp dihedral angle for A21 is slightly different in the cyclic peptide than either the monomer or fibril distributions (FIG. 4). The fibril shows several regions of dihedral angle not occupied by either the monomer or cyclic peptides. In the following descriptions and figures CA, Ca, or C a are alternatively used to describe the C-alpha atom, and similarly for CB, Cb, and C p , and so on.

[00395] The dihedral distributions are shown for E22 in FIG. 5. Specifically the 0-C-C a -C R dihedral is substantially different in the cyclic peptide than either the monomer or fibril.

[00396] The dihedral distributions are shown for D23 in FIG. 6. One can see that the dihedral angle distributions are substantially different for all dihedral angles in the cyclic conformational ensemble then they are in either the linear or fibril ensembles.

[00397] The dihedral distributions are shown for V24 in FIG. 7. The N-C a -C p -C Y1 and N-C a -

Cp-Cy 2 distributions are slightly shifted for the cyclic peptide, while the 0-C-C a -C p distribution is substantially different for the cyclic peptide.

[00398] According to the above analysis of side chain dihedral angle distributions, D23 and

V24 are the residues showing the largest discrepancy from the linear peptide and fibril ensembles. D23-V24 may be key residues on the epitope conferring conformational selectivity.

[00399] Based on the data shown in FIGs. 5-7, Table 1 lists the peak values of the dihedral angle distributions, for those dihedral angles whose distributions that show significant differences between the cyclic peptide and other species. Column 1 in Table 1 is the specific dihedral considered, column 2 is the peak value of the dihedral distribution for that angle in the context of the linear peptide CGAEDVG (SEQ ID NO: 4), column 3 is the peak value of the dihedral distribution for that angle in the context of the cyclic peptide CGAEDVG (SEQ ID NO: 4), column 4 is the difference of the peak values of the dihedral distributions for the linear and cyclic peptides, and column 5 is the peak value of the dihedral distribution for the peptide AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J.

Table 1 : Peak Values of the Dihedral Angle Distributions

Dihedral angle linear cyclic Diff(linear- fibril

cyclic)

22E: O-C-CA-CB -2.5 -87.5 85 77.5

23D: C-CA-CB-CG 67.5, 167.5 -67.5 135, 235 -67.5, 72.5,

167.5

23D: O-C-CA-CB -102.5 -62.5 -40 112.5

23D: N-CA-CB-CG -167.5, 57.5 -225, -120 62.5,

-62.5 -162.5,

-77.5

24V: N-CA-CB-CG 1 180, -72.5 -162.5 -17.5, 90 -57.5, 180 24V: N-CA-CB-CG2 -57.5, 52.5 -42.5 -15, 95 -57.5, 57.5

24V: O-C-CA-CB 72.5, 17.5 55, -120 97.5

-102.5

V. ENTROPY OF THE SIDE CHAINS

[00400] The side chain entropy of a residue may be calculated from

> J

Where the sum is over all dihedral angles in a particular residue's side chain, and p( is the dihedral angle distribution, as analyzed above.

[00401] A plot of the increase in residue entropy, over the entropy of the fibril, is shown in

FIG. 8. The entropy for both the linear and cyclic peptides is substantially increased over that in the fibril, up to approximately 15 k B = 0.125 kJ/mol/K, where k B is Boltzmann's constant. Residues D and V in the cyclic peptide have more constrained conformation than D and V in the linear peptide.

A. Dissection of entropy of residue side-chain moieties

[00402] The entropy of each dihedral angle was investigated in the respective side chains of E,D, and V (Alanine has only one dihedral for which the entropy is equal to 2.16 vs 1.72 k B in the linear and cyclic respectively). The entropy of the dihedral angles for E22, D23, and V24 are plotted in FIG. 8. The entropy tends to increase as dihedrals become further distal from the backbone, indicating that these parts of the sidechain are more exposed in the linear and cyclic peptides and so will be more accessible for antibody binding.

VI. RAMACHANDRAN ANGLES

[00403] The backbone orientation that the epitope exposes to an antibody differs depending on whether the peptide is in the linear, cyclic, or fibril form. This discrepancy can be quantified by plotting the Ramachandran # and angles phi and psi (or φ and ψ), along the backbone, for residues A, E, D,V in both the linear and cyclic peptides. Figure 9 plots the phi and psi angles sampled in equilibrium simulations, for residues A21 , E22, D23, and V24 in both linear and cyclic peptides consisting of sequence CGEADVG, as well as AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J. From FIG. 9 it is clear that the distribution of backbone dihedral angles in the cyclic peptide is substantially different from the distribution of dihedral angles sampled for either the linear peptide, or for the peptide AEDV (SEQ ID NO: 1 ) in the context the fibril structure 2M4J.

[00404] As a specific example, for residue V24, FIG. 9 shows a distribution of Ramachandran φ,ψ angles that for the cyclic peptide has peak values (most-likely values) at (φ,ψ)=(51 °, 64°). For the linear peptide, these most likely values are (φ,ψ)=(-125°, 128°), and for the fibril structure 2M4J, these most likely values are (φ,ψ)=(-71 °, 144°). This implies that antibodies selected for the cyclic epitope conformation will likely have lower affinity for the linear and fibril epitopes.

[00405] The peak values (most likely values) of the Ramachandran backbone φ,ψ distributions for A21 , E22, D23, and V24 are given in Table 2. Peak values for all phi/psi angles show significant differences between the cyclic peptide and other species for all residues. The first column in Table 2 gives the residue considered, which manifests two angles, phi and psi, indicated in parenthesis. The 2 nd column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ I D NO: 1 ) in the context of the linear peptide CGAEDVG (SEQ ID NO: 4), while the 3 rd column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ ID NO: 1 ) in the context of the cyclic peptide CGAEDVG (SEQ ID NO: 4), and the last column indicates the peak values of the Ramachandran phi/psi angles for AEDV (SEQ ID NO: 1 ) in the context of the fibril structure 2M4J.

Table 2. Peak values of distributions of backbone phi/psi angles

VII. COLLECTIVE COORDINATES PREDICTIONS

[00406] The epitope AEDV (SEQ ID NO: 1 ) also emerges from strain 2LMN and 2MXU from the collective coordinates approach described in Example 2. The corresponding figures showing the predicted epitopes are in Figure 10. For fibril structure 2LMN, 2 sequences bracketing AEDV (SEQ ID NO: 1 ) from the left and right, FAEDV (SEQ ID NO: 5) and AEDVG (SEQ ID NO: 2), are predicted, with AEDV (SEQ ID NO: 1 ) emerging more easily for less extreme external challenge (FIG. 10 left panel). For fibril structure 2MXU, again 2 sequences bracketing the left and right parts of AEDV (SEQ ID NO: 1 ), i.e. FAED (SEQ ID NO: 7) and EDVG (SEQ ID NO: 6), are predicted. From these, the overlapping consensus sequence AEDV (SEQ ID NO: 1 ) is again inferred as a likely exposed epitope.

VIII. SOLUBILITY AND ANTIGENICITY OF THE EPITOPE

[00407] The solubility of the residues of Abeta 42 according to the CamSol prediction scheme [4] is shown in the figure 1 1. Residues A21 -D24 are denoted by vertical lines. [00408] The more soluble a residue is, the more likely it is to be encountered on the surface of the oligomer. A relative solubility factor σί for residue i is introduced, as

S-i ' s ;;.' '.·■

(7i = —

^max

where s, is the solubility of residue i, and s max and s min are the maximum and minimum values of the solubility in a given range of residues, here specifically for the residues A21 -D24. The chosen range of residues is AEDV (SEQ ID NO: 1 ), and s max = 0.89 and s min = -1.1. By the above definition, σ, will be zero for the least soluble residue in the chosen range, in this case A21.

[00409] FIG. 12 plots the solvent accessible surface area (SASA), the SASA weighted by the solubility factor for each residue, σ, SASA,, and σ , SASA, minus the value in the fibril, i.e. the increase in this quantity in the monomer and cyclic peptide over the fibril, σ , ASASA,.

[00410] The SASA of the cyclic and linear peptides are comparable, and both larger than the SASA in the fibril. The solubility weighting factor takes the least soluble residue in the epitope and sets the weighted SASA for that residue to zero, in this case residue A21 at the N-terminus.

[00411] Weighting by the solubility results in the D and V residues having the most likelihood of differential exposure and availability for antibody binding, as compared to the conformation of AEDV (SEQ ID NO: 1 ) in the fibril structure.

IX. THE ENSEM BLE OF CYCLIC PEPTIDE CONFORMATIONS CLUSTERS DIFFERENTLY THAN THE ENSEMBLE OF EITHER LINEAR OR FIBRIL CONFORMATIONS

[00412] Definitive evidence that the sequence AEDV (SEQ ID NO: 1 ) displays a different conformation in the context of the cyclic peptide than in the linear peptide can be seen by using standard structural alignment metrics between conformations, and then implementing clustering analysis. Equilibrium ensembles of conformations are obtained for the linear and cyclic peptides CGAEDVG (SEQ ID NO: 4), as well as the full-length fibril in the 3-fold symmetric structure corresponding to PDB ID 2M4J. Snapshots of conformations from these ensembles for residues AEDV (SEQ ID NO: 1 ) are collected and then structurally aligned to the centroids of 3 largest clusters of the linear peptide ensemble, and the root mean squared deviation (RMSD) recorded. The clustering is performed here by the maxcluster algorithm ( http://www.sbg. bio. ic.ac.uk/maxcluster). The 3 corresponding RMSD values for the linear, cyclic, and fibril ensembles are plotted as a 3- dimensional scatter plot in FIG. 13.

[00413] It is evident from FIG. 13 that the 3 ensembles cluster differently from each other. In particular the cyclic peptide structural ensemble is distinct from either the linear or fibril ensembles, implying that antibodies specific to the cyclic peptide epitope will likely have low affinity to the conformations presented in the linear or fibril ensembles. [00414] Two views of a representative snapshot from the cyclic peptide ensemble of structures are shown in FIG. 15. As well, the sidechain orientations that are present for a representative conformation in the linear peptide ensemble are shown in black in FIG. 15, superimposed on the cyclic peptide, to make explicit their different orientations.

[00415] Table 3 lists values of the Ramachandran backbone and side chain dihedral angles undertaken for the cyclic peptide shown in FIG. 15, and for the representative conformation from the linear peptide ensemble. The large majority of dihedral angles in this table are significantly different, as described herein.

Table 3: Table of Ramachandran backbone and side chain dihedral angles shown for the cyclic peptide conformation in FIG. 15, and for a representative conformation from the linear peptide ensemble.

Example 4

Cyclic compound construction comprising a conformationally constrained epitope [00416] Peptides comprising AEDV (SEQ ID NO: 1 ) such as Cyclo(CGAEDVG) (SEQ ID NO:

4) can be cyclized head to tail.

[00417] A linear peptide comprising AEDV (SEQ ID NO: 1 ) and a linker, preferably comprising

2, 3, or 4 amino acids and/or PEG units, can be synthesized using known methods such as Fmoc based solid phase peptide synthesis alone or in combination with other methods. PEG molecules can be coupled to amine groups at the N terminus for example using coupling chemistries described in Hamley 2014 [6] and Roberts et al 2012 [7], each incorporated herein by reference. The linear peptide compound may be cyclized by covalently bonding 1 ) the amino terminus and the carboxy terminus of the peptide+linker to form a peptide bond (e.g. cyclizing the backbone), 2) the amino or carboxy terminus with a side chain in the peptide+linker or 3) two side chains in the peptide+linker.

[00418] The bonds in the cyclic compound may be all regular peptide bonds (homodetic cyclic peptide) or include other types of bonds such as ester, ether, amide or disulfide linkages (heterodetic cyclic peptide).

[00419] Peptides may be cyclized by oxidation of thiol- or mercaptan-containing residues at the N-terminus or C-terminus, or internal to the peptide, including for example cysteine and homocysteine. For example two cysteine residues flanking the peptide may be oxidized to form a disulphide bond. Oxidative reagents that may employed include, for example, oxygen (air), dimethyl sulphoxide, oxidized glutathione, cystine, copper (I I) chloride, potassium ferricyanide, thallium(l l l) trifluro acetate, or other oxidative reagents such as may be known to those of skill in the art and used with such methods as are known to those of skill in the art.

[00420] Methods and compositions related to cyclic peptide synthesis are described in US

Patent Publication 2009/0215172. US Patent publication 2010/0240865, US Patent Publication 2010/0137559, and US Patent 7,569,541 describe various methods for cyclization. Other examples are described in PCT Publication WO01/92466, and Andreu et al. , 1994. Methods in Molecular Biology 35:91-169.

[00421] More specifically, a cyclic peptide comprising the AEDV (SEQ ID NO: 1 ) epitope can be constructed by adding a linker comprising a spacer with cysteine residues flanking and/or inserted in the spacer. The peptide can be structured into a cyclic conformation by creating a disulfide linkage between the non-native cysteines residues added to the N- and C-termini of the peptide. It can also be synthesized into a cyclic compound by forming a peptide bond between the N- and C-termini amino acids (e.g. head to tail cyclization).

[00422] Peptide synthesis is performed by CPC Scientific Inc. (Sunnyvale CA, USA) following standard manufacturing procedures.

[00423] For example Cyclo(CGAEDVC) cyclic peptide comprising the conformational epitope

AEDV (SEQ ID NO: 1 ) is constructed in a constrained cyclic conformation using a disulfide linkage between cysteine residues added to the N- and C- termini of a peptide comprising AEDV (SEQ ID NO: 1 ). Two non-native cysteine residues were added to GAEDV (SEQ ID NO: 8) one at the C- terminus and one at the N-terminus. The two cysteines are oxidized under controlled conditions to form a disulfide bridge or reacted head to tail to produce a peptide bond.

[00424] As described above, the structure of the cyclic peptide was designed to mimic the conformation and orientation of the amino acid side changes of AEDV (SEQ ID NO: 1 ) in A-beta oligomer.

Cyclo(CGAEDVG) (SEQ ID NO: 4)

[00425] Cyclo(CGAEDVG) (SEQ ID NO: 4) was synthesized using the following method

(CPC Scientific Inc, Sunnyvale CA). The protected linear peptide was synthesized by standard conventional Fmoc-based solid-phase peptide synthesis on 2-chlorotrityl chloride resin, followed by cleavage from the resin with 30% HFIP/DCM. Protected linear peptide was cyclized to the corresponding protected cyclic peptide by using EDC. HCI/HOBt/DIEA in DMF at low concentration. The protected cyclic peptide was deprotected by TFA to give crude cyclic peptide and the crude peptide was purified by RP HPLC to give pure cyclic peptide after lyophilize.

[00426] Cyclo(CGAEDVG) (SEQ ID NO: 4) can be prepared by amide condensation of the linear peptide CGAEDVG.

[00427] Cyclo(C-PEG2-AEDVG) (SEQ ID NO: 23) can be prepared by amide condensation of the linear compound C-PEG2-AEDVG.

[00428] Linear(CGAEDVG) (SEQ I D NO: 4) was prepared (CPC Scientific Inc, Sunnyvale

CA) The protected linear peptide was synthesized by standard conventional Fmoc-based solid-phase peptide synthesis on Fmoc-Gly-Wang resin, then the protected peptide was cleaved by TFA to give crude peptide and the crude peptide was purified by RP HPLC to give pure peptide after lyophilize, and which was used to conjugate BSA.

Immunogen Construction

[00429] The cyclic compound Cyclo(CGAEDVG) (SEQ ID NO: 4) was synthesized as described above and then conjugated to BSA and/or KLH (CPC Scientific Inc, Sunnyvale CA). BSA or KLH was re-activated by SMCC in PBS buffer, then a solution of the pure peptide in PBS buffer was added to the conjugation mixture, the conjugation mixture was stirred at R.T. for 2h. Then the conjugation mixture was lyophilized after dialysis to give the conjugation product.

Example 5

Antibody Generation and Selection

[00430] A conformational constrained compound optionally a cyclic compound such as a cyclic peptide comprising AEDV (SEQ ID NO: 1 ) such as cyclo(CGAEDVG) (SEQ ID NO: 4) peptide is linked to Keyhole Limpet Hemocyanin (KLH). The cyclopeptide is sent for mouse monoclonal antibody production (ImmunoPrecise Antibodies LTD (Victoria BC, Canada), following protocols approved by the Canadian Council on Animal Care. Mouse sera are screened using either the conformational peptide used for producing the antibodies or a related peptide e.g. cyclo(CGAEDVG) (SEQ ID NO: 4) peptide, linked to BSA.

Hybridomas were made using an immunogen comprising cyclo(CGAEDVG) (SEQ ID NO: 4) as further described in Example 7. Hybridoma supernatants were screened by ELISA and SPR for preferential binding to cyclo(CGAEDVG) (SEQ ID NO: 4) peptide vs linear (unstructured) peptide as described herein. Positive IgG-secreting clones are subjected to large-scale production and further purification using Protein G.

Example 6

Assessing binding or lack thereof to plaques/fibrils

[00431] For immunostaining, antibodies described herein, positive control 6E10 (1 μg/ml) and isotype controls lgG1 , 2a and 2b (1 g/ml, Abeam) are used as primary antibodies. Sections are incubated overnight at 4 °C, and washed 3 x 5 min in TBS-T. Anti-mouse IgG Horseradish Peroxidase conjugated (1 : 1000, ECL) is applied to sections and incubated 45 min, then washed 3 x 5 min in TBS- T. DAB chromogen reagent (Vector Laboratories, Burlington ON, Canada) is applied and sections rinsed with distilled water when the desired level of target to background staining is achieved. Sections are counterstained with Mayer's hematoxylin, dehydrated and cover slips were applied. Slides are examined under a light microscope (Zeiss Axiovert 200M, Carl Zeiss Canada, Toronto ON, Canada) and representative images captured at 50, 200 and 400X magnification using a Leica DC300 digital camera and software (Leica Microsystems Canada Inc. , Richmond Hill, ON).

Example 7

Methods and Materials

Immunogen

[00432] Cyclic and linear peptides were generated at CPC Scientific, Sunnyvale, CA, USA.

Peptides were conjugated to KLH (for immunizing) and BSA (for screening) using a trifluoroacetate counter ion protocol. Peptides were desalted and checked by MS and HPLC and deemed 95% pure. Peptides were shipped to IPA for use in production of monoclonal antibodies in mouse.

Antibodies

[00433] A number of hybridomas and monoclonal antibodies were generated to cyclo(CGAEDVG) (SEQ I D NO: 4) linked to Keyhole Limpet Hemocyanin (KLH).

[00434] Fifty day old female BALB/c mice (Charles River Laboratories, Quebec) were immunized. A series of subcutaneous aqueous injections containing antigen but no adjuvant were given over a period of 19 days. Mice were immunized with 100μg of peptide per mouse per injection of a 0.5mg/mL solution in sterile saline of cyclic peptide-KLH. Mice were housed in a ventilated rack system from Lab Products. All 4 mice were euthanized on Day 19 and lymphocytes were harvested for hybridoma cell line generation. Fusion / Hybridoma Development

[00435] Lymphocytes were isolated and fused with murine SP2/0 myeloma cells in the presence of poly-ethylene glycol (PEG 1500). Fused cells were cultured using HAT selection. This method uses a semi-solid methylcellulose-based HAT selective medium to combine the hybridoma selection and cloning into one step. Single cell-derived hybridomas grow to form monoclonal colonies on the semi-solid media. 10 days after the fusion event, resulting hybridoma clones were transferred to 96-well tissue culture plates and grown in HT containing medium until mid-log growth was reached (5 days).

Hybridoma Analysis (Screening)

[00436] Tissue culture supernatants from the hybridomas were tested by indirect ELISA on screening antigen (cyclic peptide-BSA) (Primary Screening) and probed for both IgG and IgM antibodies using a Goat anti-lgG/lgM(H&L)-HRP secondary and developed with TMB substrate.

Clones >0.2 OD in this assay were taken to the next round of testing. Positive cultures were retested on screening antigen to confirm secretion and on an irrelevant antigen (Human Transferrin) to eliminate non-specific mAbs and rule out false positives. All clones of interest were isotyped by antibody trapping ELISA to determine if they are IgG or IgM isotype. All clones of interest were also tested by indirect ELISA on other cyclic peptide-BSA conjugates as well as linear peptide-BSA conjugates to evaluate cross-reactivity.

[00437] Mouse hybridoma antibodies were screened by Indirect ELISA using cyclo(CGAEDVG) (SEQ ID NO: 4) conjugated to BSA.

ELISA Antibody Screening

[00438] Briefly, the ELISA plates were coated with 0.1 ug/well cyclo(CGAEDVG) -conjugated -

BSA (SEQ ID NO: 4) at 100uL/well in carbonate coating buffer (pH 9.6) O/N at 4C and blocked with 3% skim milk powder in PBS for 1 hour at room temperature. Primary Antibody: Hybridoma supernatant at 100 uL/well incubated for 1 hour at 37C with shaking. Secondary Antibody 1 : 10,000 Goat anti-mouse lgG/lgM(H+L)-HRP at l OOuL/well in PBS-Tween for 1 hour at 37C with shaking. All washing steps were performed for 30 mins with PBS-Tween. The substrate 3,3', 5,5'- tetramethylbenzidine (TMB) was added at 50uL/well, developed in the dark and stopped with equal volume 1 M HCI.

[00439] Positive clones were selected for further testing. Positive clones of mouse hybridomas were tested for reactivity to cyclo(CGAEDVG) (SEQ ID NO: 4) conjugated BSA and human transferrin (HT) by indirect ELISA. Plates were coated with 1 ) 0.1 ug/well cyclo(CGAEDVG) - conjugated -BSA (SEQ ID NO: 4) at 100uL/well in carbonate coating buffer (pH 9.6) O/N at 4C; or 2) 0.25ug/well HT Antigen at 50 uL/well in dH20 O/N at 37C. Primary Antibody: Hybridoma supernatant at 100 uL/well incubated for 1 hour at 37C with shaking. Secondary Antibody 1 : 10,000 Goat anti- mouse lgG/lgM(H+L)-HRP at 100uL/well in PBS-Tween for 1 hour at 37C with shaking. All washing steps were performed for 30 mins with PBS-Tween. The substrate 3,3',5,5'-tetramethylbenzidine (TMB) was added at 50uL/well, developed in the dark and stopped with equal volume 1 M HCI.

ELISA Cvclo vs linear CGAEDVG (SEQ ID NO: 4) compound selectivity

[00440] ELISA plates were coated with 1 ) 0. "l ug/well cyclo(CGAEDVG) -conjugated -BSA

(SEQ ID NO:4) at "l OOuL/well in carbonate coating buffer (pH 9.6) O/N at 4C; 2) ) 0. "l ug/well linear CGAEDVG -conjugated -BSA (SEQ ID NO:4) at 100uL/well in carbonate coating buffer (pH 9.6) O/N at 4C; or 3) 0.1 ug/well Negative-Peptide at 100uL/well in carbonate coating buffer (pH 9.6) O/N at 4C.

Primary Antibody: Hybridoma supernatant at 100 uL/well incubated for 1 hour at 37C with shaking.

Secondary Antibody 1 : 10,000 Goat anti-mouse lgG/lgM(H+L)-HRP at l OOuL/well in PBS-Tween for 1 hour at 37C with shaking. All washing steps were performed for 30 mins with PBS-Tween. The substrate TMB was added at 50uL/well, developed in the dark and stopped with equal volume 1 M

HCI.

Isotypin

[00441] The hybridoma antibodies were isotyped using antibody trap experiments. Trap plates were coated with 1 : 10,000 Goat anti-mouse lgG/lgM(H&L) antibody at 100uL/well carbonate coating buffer pH9.6 overnight at 4C. No blocking step was used. Primary antibody (hybridoma supernatants) was added ( 100 ug/mL). Secondary Antibody 1 :5,000 Goat anti-mouse IgGy-HRP or 1 : 10,000 Goat anti-mouse IgMM-HRP at l OOuL/well in PBS-Tween for 1 hour at 37C with shaking. All washing steps were performed for 30 mins with PBS-Tween. The substrate TMB was added at 50uL/well, developed in the dark and stopped with equal volume 1 M HCI.

SPR Binding Assays - Primary and Secondary Screens

SPR analysis of Antibody binding to Abeta monomers and oligomers

[00442] A-beta Monomer and Oligomer Preparation Recombinant A-beta40 and 42 peptides

(California Peptide, Salt Lake City UT, USA) were dissolved in ice-cold hexafluoroisopropanol (HFIP). The HFIP was removed by evaporation overnight and dried in a SpeedVac centrifuge. To prepare monomers, the peptide film was reconstituted in DMSO to 5mM, diluted further to 100μ Μ in dH20 and used immediately. Oligomers were prepared by diluting the 5mM DMSO peptide solution in phenol red-free F12 medium (Life Technologies Inc. , Burlington ON, Canada) to a final concentration of 100μ Μ and incubated for 24 hours to 7 days at 4°C.

[00443] SPR Analysis All SPR measurements were performed using a Molecular Affinity

Screening System ( MASS-1 ) (Sierra Sensors GmbH, Hamburg, Germany), an analytical biosensor that employs high intensity laser light and high speed optical scanning to monitor binding interactions in real time. The primary screening of tissue culture supernatants was performed using an SPR direct binding assay, whereby BSA-conjugated peptides, A-beta42 Monomer and A-beta42 Oligomer are covalently immobilized on individual flow cells of a High Amine Capacity (HAC) sensorchip (Sierra Sensors GmbH, Hamburg, Germany) and antibodies flowed over the surface. Protein G purified mAbs were analyzed in a secondary screen using an SPR indirect (capture) binding assay, whereby the antibodies were captured on a protein A-derivatized sensorchip (XanTec Bioanalytics GmbH, Duesseldorf, Germany) and A-beta40 Monomer, A-beta42 Oligomer, soluble brain extracts and cerebrospinal fluid flowed over the surface. The specificity of the antibodies was verified in an SPR direct binding assay by covalently immobilizing A-beta42 Monomer and A-beta42 Oligomer on individual flow cells of a HAC sensorchip and flowing purified mAbs.

SPR analysis of soluble brain extracts and CSF samples

[00444] Soluble brain extract and CSF Preparation Human brain tissues and CSFs were obtained from patients assessed at the UBC Alzheimer's and Related Disorders Clinic. Clinical diagnosis of probable AD is based on NINCDS-ADRDA criteria [5]. CSFs are collected in polypropylene tubes, processed, aliquoted into 100 μΙ_ polypropylene vials, and stored at -80°C within 1 hour after lumbar puncture.

[00445] Homogenization: Human brain tissue samples were weighed and subsequently submersed in a volume of fresh, ice cold TBS (supplemented with EDTA-free protease inhibitor cocktail from Roche Diagnostics, Laval QC, Canada) such that the final concentration of brain tissue is 20% (w/v). Tissue is homogenized in this buffer using a mechanical probe homogenizer (3 x 30 sec pulses with 30 sec pauses in between, all performed on ice). TBS homogenized samples are then subjected to ultracentrifugation (70,000xg for 90 min). Supernatants are collected, aliquoted and stored at -80°C. The protein concentration of TBS homogenates is determined using a BCA protein assay (Pierce Biotechnology Inc, Rockford IL, USA).

[00446] SPR Analysis Brain extracts from 4 AD patients and 4 age-matched controls, and

CSF samples from 9 AD patients and 9 age-matched controls were pooled and analyzed. Purified mAbs were captured on separate flow cells of a protein A-derivatized sensor chip and diluted samples injected over the surfaces for 180 seconds, followed by 120 seconds of dissociation in buffer and surface regeneration. Binding responses were double-referenced by subtraction of mouse control IgG reference surface binding and assay buffer, and the different groups of samples compared

Assessing binding or lack thereof to A-beta monomers

[00447] In the primary screen of tissue culture supernatants, A-beta42 monomers and A- beta42 oligomers were used in a direct binding assay. In the secondary screen, A-beta40 monomers and A-beta42 oligomers, soluble brain extracts and CSF samples were used in an indirect (capture) binding assay.

[00448] Primary Screen

Tissue culture supernatants were screened for the presence of antibody binding against their cognate cyclic peptide. Each sample was diluted and injected in duplicate over the immobilized peptide and BSA reference surfaces for 120 seconds, followed by injection of running buffer only for a 300-second dissociation phase. After every analytical cycle, the sensor chip surfaces were regenerated. Sensorgrams were double-referenced by subtracting out binding from the BSA reference surfaces and blank running buffer injections, and binding response report points collected in the dissociation phase.

Oligomer Binding Assay

[00449] Next synthetic A-beta42 oligomers were generated and immobilized as above, antibody binding responses analyzed. Antibody binding responses to A-beta42 oligomers were compared to binding responses to cyclic.

Verifying binding to A-beta oligomers

[00450] To further verify and validate A-beta42 oligomer binding, antibodies were covalently immobilized, followed by the injection over the surface of commercially-prepared stable A-beta42 Oligomers (SynAging SAS, Vandceuvre-les-Nancy, France).

Results

[00451] ELISA testing found that the majority of hybridoma clones bound the cyclopeptide.

[00452] Next clones were tested by ELISA for their binding selectivity for cyclo- and linear- CGAEDVG (SEQ ID NO: 4) compounds. A number of clones preferentially bound cyclo(CGAEDVG) - conjugated -BSA (SEQ ID NO: 4) compared to linear CGAEDVG -conjugated -BSA (SEQ I D NO: 4).

[00453] Isotyping revealed that the majority of clones were IgG including lgG1 , lgG2a and lgG3 clones. Several IgM and IgA clones were also identified, but not pursued further.

[00454] A direct binding analysis using surface plasmon resonance was performed to screen for antibodies in tissue culture supernatants that bind to the cyclic peptide of SEQ ID NO: 4.

[00455] Clones were tested for their ability to bind cyclic peptide, linear peptide, A-beta42 monomer and A-beta42 oligomers prepared as described above. Binding assays were performed using SPR as described above (Direct binding assays). FIG.16 plots the results of the SPR binding of the IgG clones to linear and cyclic peptide (Panel A), and to A-beta monomer and A-beta oligomer (Panel B). A number of clones were selected based on the binding assays performed as shown in Table 5.

[00456] FIG. 17 plots the results of the direct binding assay and the ELISA results and shows that there is a correlation between the direct binding and ELISA results.

[00457] [00458] The selected clones were IgG mAb. Negative numbers in the primary screen are indicative of no binding (i.e. less than isotype control). Table 5

302

ELISA Prescreen

[00459] The ELISA prescreen of hybridoma supernatants identified clones which showed increased binding to the cyclic peptides compared to the linear peptide. A proportion of the clones were reactive to KLH-epitope linker peptide. These were excluded from further investigation. The majority of the clones were determined to be of the IgG isotype using the isotyping procedure described herein.

Direct Binding Measured by Surface Plasmon Resonance - Primary Screen [00460] Using surface plasmon resonance the tissue culture supernatants containing antibody clones were tested for direct binding to cyclic peptide, linear peptide, A-beta oligomer and A- beta monomer.

[00461] The results for the primary screen are shown in FIG. 16. Panel A shows binding to cyclic peptide and to linear peptide. Panel B shows binding to A-beta oligomer and A-beta monomer. A number of the clones have elevated reactivity to the cyclic peptide and all clones have minimal or no reactivity to linear peptide. There is a general selectivity for A-beta oligomer binding.

[00462] For select clones comparative binding profile is shown in FIG. 18. Each clone is assessed for direct binding using surface plasmon resonance against specific epitope in the context of cyclic peptide (structured), linear peptide (unstructured), A-beta monomer, and A-beta oligomer.

Example 8

Secondary Screen

Immunohistochemistry

[00463] Immunohistochemistry was performed on frozen human brain sections, with no fixation or antigen retrieval. In a humidified chamber, non-specific staining was blocked by incubation with serum-free protein blocking reagent (Dako Canada Inc. , Mississauga, ON, Canada) for 1 h. The following primary antibodies were used for immunostaining: mouse monoclonal isotype controls lgG1 , lgG2a, and lgG2b, and anti-amyloidp 6E10, all purchased from Biolegend, and selected purified clones reactive to the cyclopeptide. All antibodies were used at 1 μg/mL·. Sections were incubated at room temperature for 1 h, and washed 3 x 5 min in TBS-T. Anti-Mouse IgG Horseradish Peroxidase conjugated (1 : 1000, ECL) was applied to sections and incubated 45 min, then washed 3 x 5 min in TBS-T. DAB chromogen reagent (Vector Laboratories, Burlington ON, Canada) was applied and sections rinsed with distilled water when the desired level of target to background staining was achieved. Sections were counterstained with Mayer's haematoxylin, dehydrated and cover slips were applied. Slides were examined under a light microscope (Zeiss Axiovert 200M, Carl Zeiss Canada, Toronto ON, Canada) and representative images captured at 20 and 40X magnification using a Leica DC300 digital camera and software (Leica Microsystems Canada Inc. , Richmond Hill, ON). Images were optimized in Adobe Photoshop using Levels Auto Correction.

CSF and Brain Extracts

[00464] Human brain tissues were obtained from the University of Maryland Brain and

Tissue Bank upon approval from the UBC Clinical Research Ethics Board (C04-0595). CSFs were obtained from patients assessed at the UBC Hospital Clinic for Alzheimer's and Related Disorders. The study was approved by the UBC Clinical Research Ethics Board, and written consent from the participant or legal next of kin was obtained prior to collection of CSF samples. Clinical diagnosis of probable AD was based on NINCDS-ADRDA criteria. CSFs were collected in polypropylene tubes, processed, aliquoted into 100 μί polypropylene vials, and stored at -80°C within 1 hour after lumbar puncture.

[00465] Homogenization: Human brain tissue samples were weighed and subsequently submersed in a volume of fresh, ice cold TBS and EDTA-free protease inhibitor cocktail from Roche Diagnostics (Laval QC, Canada) such that the final concentration of brain tissue was 20% (w/v). Tissue was homogenized in this buffer using a mechanical probe homogenizer (3 x 30 sec pulses with 30 sec pauses in between, all performed on ice). TBS homogenized samples were then subjected to ultracentrifugation (70,000xg for 90 min). Supernatants were collected, aliquoted and stored at -80°C. The protein concentration of TBS homogenates was determined using a BCA protein assay (Pierce Biotechnology Inc, Rockford IL, USA).

[00466] CSF: CSF was pooled from 9 donors with AD and 9 donors without AD. Samples were analyzed by SPR using purified IgG at a concentration of 30 micrograms/ml for all antibodies. Mouse IgG was used as an antibody control, and all experiments were repeated at least 2 times. [00467] Positive binding in CSF and brain extracts was confirmed using antibody 6E10.

[00468] SPR Analysis: 4 brain extracts from AD patients and 4 brain extracts from age- matched controls were pooled and analyzed. Brain samples, homogenized in TBS, included frontal cortex Brodmann area 9. All experiments were performed using a Molecular Affinity Screening System (MASS-1 ) (Sierra Sensors GmbH, Hamburg, Germany), an analytical biosensor that employs high intensity laser light and high speed optical scanning to monitor binding interactions in real time as described in Example 6. Purified antibodies generated for cyclopeptides described herein were captured on separate flow cells of a protein A-derivatized sensor chip and diluted samples injected over the surfaces for 180 seconds, followed by 120 seconds of dissociation in buffer and surface regeneration. Binding responses were double-referenced by subtraction of mouse control IgG reference surface binding and assay buffer, and the different groups of samples compared.

Results

CSF Brain Extracts and Immunohistochemistry

[00469] Several clones were tested for their ability to bind A-beta in CSF, soluble brain extracts and tissue samples of cadaveric AD brains are shown in Table 6. Strength of positivity in Table 6 is shown by the number plus signs.

[00470] Table 6 and Table 7 provide data for selected clone's binding selectivity for oligomers over monomer measured as described herein by SPR.

[00471] IHC results are also summarized in Table 6 where "+/-" denotes staining similar to or distinct from isotype control but without clear plaque morphology. [00472] FIG. 19 shows an example of the lack of plaque staining on fresh frozen sections with clone 24-10A4 compared to the positive plaque staining seen with 6E10 antibody.

[00473] FIG. 20 shows, antibodies raised to the cyclopeptide comprising AEDV (SEQ ID

NO: 1 ) bound A-beta oligomer preferentially over monomer and also preferentially bound A-beta in brain extracts and/ or CSF of AD patients. [00474] As shown in Tables 6, 7 and FIGS. 19 and 20, antibodies raised to the cyclopeptide comprising AEDV (SEQ I D NO: 1 ) bound to A-beta in brain extracts and/or CSF, but did not bind strongly to monomers on SPR, and did not appreciably bind to plaque fibrils by IHC.

Table 6: Summary of binding characteristics

22 (9B1 ) +++ ++ +/-

24 (10A4) ++ ++ * Scoring is relative to other clones in the same sample category.

Table 7. A-beta Oligomer binding RU values subtracted for monomer binding

Example 9

Synthetic Oligomer Binding

Serial 2-fold dilutions (7.8 nM to 2000nM) of commercially-prepared synthetic amyloid beta oligomers (SynAging SAS, Vandceuvre-les-Nancy, were tested for binding to covalently immobilized antibodies. Results for control antibody mAb6E10 is shown in FIG. 21 A and for mouse control IgG control is shown in FIG. 21 B. FIG. 21 C shows results using an antibody raised against cyclo (CGAEDVG) (SEQ ID NO:4).

Example 10

Immunohistochemistry on Formalin Fixed Tissues

[00475] Human brain tissue was assessed using antibodies raised to CGAEDVG (SEQ ID NO:

4). The patient had been previously characterized and diagnosed with Alzheimer's disease with a tripartite approach: (i) Bielschowsky silver method to demonstrate senile plaques and neurofibrillary tangles, (ii) Congo red to demonstrate amyloid and (iii) tau immunohistochemistry to demonstrate tangles and to confirm the senile plaques are "neuritic". This tissue was used to test plaque reactivity of selected monoclonal antibody clones. The brain tissues were fixed in 10% buffered formalin for several days and paraffin processed in the Sakura VIP tissue processors. Tissue sections were probed with ^g/ml of antibody with and without microwave antigen retrieval (AR). The pan-amyloid beta reactive antibody 6E10 was included along with selected antibody clones as a positive control. Antibodies were diluted in Antibody Diluent (Ventana), color was developed with OptiView DAB (Ventana). The staining was performed on the Ventana Benchmark XT IHC stainer. Images were obtained with an Olympus BX45 microscope. Images were analyzed blind by a professional pathologist with expertise in neuropathology.

[00476] As shown in Table 8 below, using fixed tissue, the tested antibodies were negative for specific staining of senile plaque amyloid with or without antigen retrieval. 6E10 was used as the positive control. Table 8

Epitope Antibodies to test Convincing evidence of specific staining of senile plaque amyloid

Without AR Plus AR

302 19 Neg Neg

22 Neg Neg

24 Neg Neg

Positive control 6E10 Strongly Positive Strongly positive

Example 11

Inhibition of Oligomer Propagation

[00477] The biological functionality of antibodies was tested in vitro by examining their effects on propagation of Amyloid Beta (Αβ) aggregation using the Thioflavin T (ThT) binding assay. Αβ aggregation is induced by and propagated through nuclei of preformed small Αβ oligomers, and the complete process from monomeric Αβ to soluble oligomers to insoluble fibrils is accompanied by concomitantly increasing beta sheet formation. This can be monitored by ThT, a benzothiazole salt, whose excitation and emission maxima shifts from 385 to 450nm and from 445 to 482nm respectively when bound to beta sheet-rich structures and resulting in increased fluorescence Briefly, Αβ 1-42 (Bachem Americas Inc., Torrance, CA) was solubilized, sonicated, diluted in Tris-EDTA buffer (pH7.4) and added to wells of a black 96-well microtitre plate (Greiner Bio-One, Monroe, NC) to which equal volumes of cyclopeptide raised antibody or irrelevant mouse IgG antibody isotype controls were added, resulting in a 1 :5 molar ratio of Αβ1 -42 peptide to antibody. ThT was added and plates incubated at room temperature for 24 hours, with ThT fluorescence measurements (excitation at 440nm, emission at 486nm) recorded every hour using a Wallac Victor3v 1420 Multilabel Counter (PerkinElmer, Waltham, MA). Fluorescent readings from background buffer were subtracted from all wells, and readings from antibody only wells were further subtracted from the corresponding wells.

[00478] Αβ42 aggregation, as monitored by ThT fluorescence, demonstrated a sigmoidal shape characterized by an initial lag phase with minimal fluorescence, an exponential phase with a rapid increase in fluorescence and finally a plateau phase during which the Αβ molecular species are at equilibrium and during which there is no increase in fluorescence. Co-incubation of Αβ42 with an irrelevant mouse antibody did not have any significant effect on the aggregation process. In contrast, co-incubation of Αβ42 with the test antibodies completely inhibited all phases of the aggregation process. Results were obtained with antibody clone 24 (10A4; lgG3 isotype) . As the ThT aggregation assay mimics the in vivo biophysical / biochemical stages of Αβ propagation and aggregation from monomers, oligomers, protofibrils and fibrils that is pivotal in AD pathogenesis, the antibodies raised to cyclo CGAEDVG demonstrate the potential to completely abrogate this process. Example 12

70

RECTIFIED SHEET (RULE 91.1) [00479] Achieving the optimal profile for Alzheimer's immunotherapy: Rational generation of antibodies specific for toxic A-beta oligomers

[00480] Objective: Generate antibodies specific for toxic amyloid-β oligomers (ΑβΟ) [00481] Background: Current evidence suggests that propagating prion-like strains of ΑβΟ, as opposed to monomers and fibrils, are preferentially toxic to neurons and trigger tau pathology in Alzheimer's disease (AD). In addition, dose-limiting adverse effects have been associated with Αβ fibril recognition in clinical trials. These observations suggest that specific neutralization of toxic Αβθε may be desirable for safety and efficacy.

[00482] Design/Methods: Computational simulations were employed as described herein, using molecular dynamics with standardized force-fields to perturb atomic-level structures of Αβ fibrils deposited in the Protein Data Base. It was hypothesized that weakly-stable regions are likely to be exposed in nascent protofibrils or oligomers. Clustering analysis, curvature, exposure to solvent, solubility, dihedral angle distribution, and Ramachandran angle distributions were all used to characterize the conformational properties of predicted epitopes, which quantify differences in the antigenic profile when presented in the context of the oligomer vs the monomer or fibril. The candidate peptide epitopes were synthesized in a cyclic format that may mimic regional ΑβΟ conformation, conjugated to a carrier protein, and used to generate monoclonal antibodies in mice. Purified antibodies were screened by SPR and immunohistochemistry.

Results:

[00483] Sixty-six IgG clones against 5 predicted epitopes were selected for purification based on their ability to recognize the cognate structured peptide and synthetic ΑβΟ, with little or no binding to unstructured peptide, linker peptide, or Αβ monomers. Additional screening identified antibodies that preferentially bound to native soluble ΑβΟ in CSF and brain extracts of AD patients compared to controls. Immunohistochemical analysis of AD brain allowed for selection of antibody clones that do not react with plaque. [00484] Conclusion: Computationally identified ΑβΟ epitopes allowed for the generation of antibodies with the desired target profile of selective binding to native AD Αβθε with no significant cross-reactivity to monomers or fibrils.

Example 13

Toxicity inhibition assay

[00485] The inhibition of toxicity of A-beta42 oligomers by antibodies raised to the cyclopeptide can be tested in a rat primary cortical neuron assay. [00486] Antibody and control IgG are each adjusted to a concentration such as 2 mg/mL.

Various molar ratios of A-beta oligomer and antibody are tested along with a vehicle control, A-beta oligomer alone and a positive control such as the neuroprotective peptide humanin (HNG).

[00487] An exemplary set up is shown in Table 9.

[00488] Following preincubation for 10 minutes at room temperature, the volume is adjusted to 840 microlitres with culture medium. The solution is incubated for 5 min at 37C. The solution is then added directly to the primary cortical neurons and cells are incubated for 24h. Cell viability can be determined using the MTT assay.

Table 9

I ΑβΟ working solution: 2,2 mg/mL - 500 μΜ

CTRL vehicle: 1 ,68 μί of oligomer buffer + 127,3 μί PBS + 71 1 μί culture medium

CTRL ΑβΟ: 1 ,68 μί of ΑβΟ + 127, 3 μί PBS + 71 1 μΙ_ culture medium

1 ,68 μΙ_ of ΑβΟ + 8,4 μΙ_ HNG (100 nM final) + 127,3 μΙ_ PBS + 702,6 μΙ_ culture

CTRL HNG: medium

[00489] This test was conducted using other antibodies raised against other cyclopeptides comprising other epitopes predicted by the collective coordinates method described in Example 1. Inhibition of A-beta oligomer toxicity was observed for these other epitopes. Antibodies raised against cyclo(CGAEDVG) (SEQ ID NO: 4) will be tested.

Example 14

In vivo toxicity inhibition assay

[00490] The inhibition of toxicity of A-beta42 oligomers by antibodies raised to the cyclopeptide can be tested in vivo in mouse behavioral assays.

[00491] The antibody and an isotype control are each pre-mixed with A-beta42 oligomers at 2 or more different molar ratios prior to intracerebroventricular (ICV) injection into mice. Control groups include mice injected with vehicle alone, oligomers alone, antibody alone, and a positive control such as the neuroprotective peptide humanin. Alternatively, the antibodies can be administered systemically prior to, during, and/or after ICV injection of the oligomers. Starting approximately 4-7 days post ICV injection of oligomers, cognition is assessed in behavioral assays of learning and memory such as the mouse spatial recognition test (SRT), Y-Maze assay, Morris water maze model and novel object recognition model (NOR).

[00492] The mouse spatial recognition test (SRT) assesses topographical memory, a measure of hippocampal function (SynAging). The model uses a two-chamber apparatus, in which the chambers differ in shape, pattern and color (i.e. topographical difference). The chambers are connected by a clear Plexiglass corridor. Individual mice are first placed in the apparatus for a 5 min exploration phase where access to only one of the chambers is allowed. Mice are then returned to their home cage for 30 min and are placed back in the apparatus for a 5 min " choice" phase during which they have access to both chambers. Mice with normal cognitive function remember the previously explored chamber and spend more time in the novel chamber. A discrimination index (Dl) is calculated as follows: Dl = (TN - TF)/(TN + TF), in which TN is the amount of time spent in the novel chamber and TF is the amount of time spent in the familiar chamber. Toxic A-beta oligomers cause a decrease in Dl which can be partially rescued by the humanin positive control. Performance of this assay at different time points post ICV injection can be used to evaluate the potential of antibodies raised to the cyclopeptide to inhibit A-beta oligomer toxicity in vivo.

[00493] The Y-maze assay (SynAging) is a test of spatial working memory which is mainly mediated by the prefrontal cortex (working memory) and the hippocampus (spatial component). Mice are placed in a Y-shaped maze where they can explore 2 arms. Mice with intact short-term memory will alternate between the 2 arms in successive trials. Mice injected ICV with toxic A-beta oligomers are cognitively impaired and show random behavior with alternation close to a random value of 50% (versus -70% in normal animals). This impairment is partially or completely reversed by the cholinesterase inhibitor donepezil (Aricept) or humanin, respectively. This assay provides another in vivo assessment of the protective activity of test antibodies against A-beta oligomer toxicity.

[00494] The Morris water maze is another widely accepted cognition model, investigating spatial learning and long-term topographical memory, largely dependent on hippocampal function (SynAging). Mice are trained to find a platform hidden under an opaque water surface in multiple trials. Their learning performance in recalling the platform location is based on visual clues and video recorded. Their learning speed, which is the steadily reduced time from their release into the water until finding the platform, is measured over multiple days. Cognitively normal mice require less and less time to find the platform on successive days (learning). For analyzing long-term memory, the test is repeated multiple days after training: the platform is taken away and the number of crossings over the former platform location, or the time of the first crossing, are used as measures to evaluate long- term memory. Mice injected ICV with toxic A-beta oligomers show deficits in both learning and long- term memory and provide a model for evaluating the protective activity of test antibodies.

[00495] The Novel Object Recognition (NOR) model utilizes the normal behavior of rodents to investigate novel objects for a significantly longer time than known objects, largely dependent on perirhinal cortex function (SynAging). Mice or rats are allowed to explore two identical objects in the acquisition trial. Following a short inter-trial interval, one of the objects is replaced by a novel object. The animals are returned to the arena and the time spent actively exploring each object is recorded. Normal rodents recall the familiar object and will spend significantly more time exploring the novel object. In contrast, A-beta oligomer-treated rodents exhibit clear cognitive impairment and will spend a similar amount of time investigating both the 'familiar' and 'novel' object. This can be transiently reversed with known clinical cognitive enhancers (e.g. donepezil). The NOR assay can be performed multiple times in longitudinal studies to assess the potential cognitive benefit of test antibodies.

[00496] In addition to behavioral assays, brain tissue can be collected and analyzed for levels of synaptic markers (PSD95, SNAP25, synaptophysin) and inflammation markers (IL-1 -beta). Mice are sacrificed at -14 days post-ICV injection of oligomers and perfused with saline. Hippocampi are collected, snap frozen and stored at -80°C until analyzed. Protein concentrations of homogenized samples are determined by BCA. Concentration of synaptic markers are determined using ELISA kits (Cloud-Clone Corp, USA). Typically, synaptic markers are reduced by 25-30% in mice injected with A-beta oligomers and restored to 90-100% by the humanin positive control. Concentrations of the IL- 1 -beta inflammatory markers are increased approximately 3-fold in mice injected with A-beta oligomers and this increase is largely prevented by humanin. These assays provide another measure of the protective activity of test antibodies at the molecular level.

Example 15

CDR sequencing

[00497] 302-10A4.1 which was determined to have an lgG3 heavy chain and a kappa light chain was selected for CDR and variable regions of the heavy and light chains.

[00498] RT-PCR was carried out using 5' RACE and gene specific reverse primers which amplify the appropriate mouse immunoglobulin heavy chain (lgG1/lgG3/lgG2A) and light chain (kappa) variable region sequences.

[00499] The specific bands were excised and cloned into pCR-Blunt ll-TOPO vector for sequencing, and the constructs were transformed into E. coli.

[00500] At least 8 colonies of each chain were picked & PCR screened for the presence of amplified regions prior to sequencing. Selected PCR positive clones were sequenced.

[00501] The CDR sequences are in Table 10. The consensus DNA sequence and protein sequences of the variable portion of the heavy and light chain are provided in Table 1 1 . Table 10

Table 11

Consensus DNA sequence and translated protein sequences of the variable region. The complementarity determining regions (CDRs) are underlined according to IMTG/LIGM-DB.

Table 12 A-beta "Epitope" Sequences and select A-beta sequences with linker

AEDV (SEQ ID NO: 1 )

AEDVG (SEQ ID NO: 2)

AEDVGS (SEQ ID NO: 3)

CGAEDVG, cyclo(CGAEDVG) SEQ ID NO: 4

FAEDV (SEQ ID NO: 5)

EDVG (SEQ I D NO: 6)

FAED (SEQ ID NO: 7)

GAEDV (SEQ ID NO: 8)

GAEDVG (SEQ ID NO: 9)

GGAEDVG (SEQ ID NO: 1 0)

GAEDVGG (SEQ ID NO: 1 1 )

Cyc/o(C-PEG2-AEDVG); C-PEG2-AEDVG (SEQ ID NO: 23)

Cyclo(CGAEDV-PEG2); CGAEDV-PEG2(SEQ ID NO: 24)

Table 13

DAEFRHDSGYEVHHQKLVFFAEDVGSNKGAIIGLMVGGWIA (SEQ ID NO: 22)

[00502] While the present application has been described with reference to what are presently considered to be the preferred examples, it is to be understood that the application is not limited to the disclosed examples. To the contrary, the application is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.

[00503] All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Specifically, the sequences associated with each accession numbers provided herein including for example accession numbers and/or biomarker sequences (e.g. protein and/or nucleic acid) provided in the Tables or elsewhere, are incorporated by reference in its entirely.

[00504] The scope of the claims should not be limited by the preferred embodiments and examples, but should be given the broadest interpretation consistent with the description as a whole.

CITATIONS FOR REFERENCES REFERRED TO IN THE SPECIFICATION

[1 ] Gabriela A. N. Crespi, Stefan J. Hermans, Michael W. Parker, and Luke A. Miles. Molecular basis for mid-region amyloid-b capture by leading Alzheimer's disease immunotherapies SCIENTIFIC REPORTS | 5 : 9649, 2015\ DOT. 10.1038/srep09649

[2] Vincent J. Hilser and Ernesto Freire. Structure-based calculation of the equilibrium folding pathway of proteins, correlation with hydrogen exchange protection factors. J. Mol. Biol. ,262:756- 772, 1996. The COREX approach.

[3] Samuel I. A. Cohen, Sara Linse, Leila M. Luheshi, Erik Hellstrand, Duncan A. White, Luke Rajah, Daniel E. Otzen, Michele Vendruscolo, Christopher M. Dobson, and Tuomas P. J. Knowles. Proliferation of amyloid-p42 aggregates occurs through a secondary nucleation mechanism. Proc. Natl. l Acad. Sci. USA, 1 10(24):9758-9763, 2013.

[4] Pietro Sormanni, Francesco A. Aprile, and Michele Vendruscolo. The camsol method of rational design of protein mutants with enhanced solubility. Journal of Molecular Biology, 427(2):478-490, 2015.

[5] Deborah Blacker, MD, ScD; Marilyn S. Albert, PhD; Susan S. Bassett, PhD; Rodney C. P. Go, PhD; Lindy E. Harrell, MD, PhD; Marshal F. Folstein, MD Reliability and Validity of NINCDS-ADRDA Criteria for Alzheimer's Disease The National Institute of Mental Health Genetics Initiative. Arch Neurol. 1994;51 (12): 1 198-1204. doi: 10.1001/archneur.1994.00540240042014.

[6] Hamley, I.W. PEG-Peptide Conjugates 2014; 15, 1543-1559; dx.doi.org/10.1021/bm500246w

[7 ] Roberts, MJ et al Chemistry for peptide and protein PEGylation 64: 1 16-127.

[8] J.X. Lu, W.Qiang, W. M.Yau, C. D.Schwieters, S.C. Meredith, R.Tycko, MOLECULAR STRUCTURE OF BETA-AMYLOID FIBRILS IN ALZHEI MER'S DISEASE BRAIN TISSUE. CELL Vol. 154 p.1257 (2013)

[9] Y.Xiao, B. MA, D. McElheny, S. Parthasarathy, F.Long, M. Hoshi, R. Nussinov, Y. lshii, A BETA (1 -42) FIBRIL STRUCTURE ILLUM INATES SELF-RECOGNITION AND REPLICATION OF AMYLOID IN ALZHEI MER'S DISEASE. NAT. STRUCT. MOL. BIOL. Vol. 22 p.499 (2015).

[10] A. Petkova.W.Yau, R.Tycko EXPERIMENTAL CONSTRAINTS ON QUATERNARY STRUCTURE IN ALZHEI MER'S BETA-AMYLOID FIBRILS BIOCHEM ISTRY V. 45 498 2006.

[1 1 ] Yu YZ, Wang WB, Chao A, Chang Q, Liu S, Zhao M, et al. Strikingly reduced amyloid burden and improved behavioral performance in Alzheimer's disease mice immunized with recombinant chimeric vaccines by hexavalent foldable A _1 -15 fused to toxin-derived carrier proteins. J Alzheimer's Dis 2014;41 :243-60.

[12]Wang, HC; Yu, YZ; Liu, S; Zhao, M and Q Xu, Peripherally administered sera antibodies recognizing amyloid- _oligomers mitigate Alzheimer's disease-like pathology and cognitive decline in aged 3 χ Tg-AD mice, Vaccine 2016.