Antibody Capable of Binding Specifically to AB-Oligomer, and Use Thereof

MATSUBARA; Etsuro ;   et al.

Patent Application Summary

U.S. patent application number 12/851233 was filed with the patent office on 2011-04-28 for antibody capable of binding specifically to ab-oligomer, and use thereof. This patent application is currently assigned to Immunas Pharma, Inc.. Invention is credited to Etsuro MATSUBARA, Masao Shibata, Tatsuki Yokoseki.

Application Number20110097319 12/851233
Document ID /
Family ID40952248
Filed Date2011-04-28

United States Patent Application 20110097319
Kind Code A1
MATSUBARA; Etsuro ;   et al. April 28, 2011

Antibody Capable of Binding Specifically to AB-Oligomer, and Use Thereof

Abstract

The present inventors successfully produced monoclonal antibodies that are specific to only soluble A.beta. oligomers, but do not recognize soluble A.beta. monomers, which are physiological molecules. It was demonstrated that the antibodies are useful as diagnostic/therapeutic monoclonal antibodies for Alzheimer's disease.


Inventors: MATSUBARA; Etsuro; (Aomori, JP) ; Shibata; Masao; (Kanagawa, JP) ; Yokoseki; Tatsuki; (Kanagawa, JP)
Assignee: Immunas Pharma, Inc.
Kanagawa
JP

National Center for Geriatrics and Gerontology
Aichi
JP

Family ID: 40952248
Appl. No.: 12/851233
Filed: August 5, 2010

Related U.S. Patent Documents

Application Number Filing Date Patent Number
12533348 Jul 31, 2009
12851233
PCT/JP2009/052039 Feb 6, 2009
12533348
61085545 Aug 1, 2008

Current U.S. Class: 424/130.1 ; 435/7.1; 530/387.1; 530/387.3
Current CPC Class: C07K 2317/76 20130101; A61P 25/28 20180101; A61P 25/00 20180101; G01N 33/6896 20130101; A61K 2039/505 20130101; G01N 2800/2821 20130101; C07K 2317/565 20130101; A61P 17/00 20180101; G01N 2333/4709 20130101; A61K 39/3955 20130101; C07K 2317/56 20130101; C07K 2317/73 20130101; C07K 16/18 20130101
Class at Publication: 424/130.1 ; 530/387.1; 530/387.3; 435/7.1
International Class: A61K 39/395 20060101 A61K039/395; C07K 16/00 20060101 C07K016/00; A61P 25/28 20060101 A61P025/28; G01N 33/53 20060101 G01N033/53

Foreign Application Data

Date Code Application Number
Feb 8, 2008 JP 2008-028386
Aug 4, 2008 JP 2008-201058

Claims



1. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 1 and an L chain having the amino acid sequence of SEQ ID NO: 3.

2. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 21 and an L chain having the amino acid sequence of SEQ ID NO: 23.

3. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 41 and an L chain having the amino acid sequence of SEQ ID NO: 43.

4. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 61 and an L chain having the amino acid sequence of SEQ ID NO: 63.

5. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 81 and an L chain having the amino acid sequence of SEQ ID NO: 83.

6. An antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 101 and an L chain having the amino acid sequence of SEQ ID NO: 103.

7. An antibody of any one of (1) to (38) below: (1) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 9 as CDR1, the amino acid sequence of SEQ ID NO: 11 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3; (2) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 15 as CDR1, the amino acid sequence of SEQ ID NO: 17 as CDR2, and the amino acid sequence of SEQ ID NO: 19 as CDR3; (3) an antibody that comprises the H chain of (1) and the L chain of (2); (4) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 5 as VH; (5) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 7 as VL; (6) an antibody that comprises the H chain of (4) and the L chain of (5); (7) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 29 as CDR1, the amino acid sequence of SEQ ID NO: 31 as CDR2, and the amino acid sequence of SEQ ID NO: 33 as CDR3; (8) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 35 as CDR1, the amino acid sequence of SEQ ID NO: 37 as CDR2, and the amino acid sequence of SEQ ID NO: 39 as CDR3; (9) an antibody that comprises the H chain of (7) and the L chain of (8); (10) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 25 as VH; (11) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 27 as VL; (12) an antibody that comprises the H chain of (10) and the L chain of (11); (13) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 49 as CDR1, the amino acid sequence of SEQ ID NO: 51 as CDR2, and the amino acid sequence of SEQ ID NO: 53 as CDR3; (14) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 55 as CDR1, the amino acid sequence of SEQ ID NO: 57 as CDR2, and the amino acid sequence of SEQ ID NO: 59 as CDR3; (15) an antibody that comprises the H chain of (13) and the L chain of (14); (16) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 45 as VH; (17) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 47 as VL; (18) an antibody that comprises the H chain of (16) and the L chain of (17); (19) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 69 as CDR1, the amino acid sequence of SEQ ID NO: 71 as CDR2, and the amino acid sequence of SEQ ID NO: 73 as CDR3; (20) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 75 as CDR1, the amino acid sequence of SEQ ID NO: 77 as CDR2, and the amino acid sequence of SEQ ID NO: 79 as CDR3; (21) an antibody that comprises the H chain of (19) and the L chain of (20); (22) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 65 as VH; (23) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 67 as VL; (24) an antibody that comprises the H chain of (22) and the L chain of (23); (25) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 89 as CDR1, the amino acid sequence of SEQ ID NO: 91 as CDR2, and the amino acid sequence of SEQ ID NO: 93 as CDR3; (26) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 95 as CDR1, the amino acid sequence of SEQ ID NO: 97 as CDR2, and the amino acid sequence of SEQ ID NO: 99 as CDR3; (27) an antibody that comprises the H chain of (25) and the L chain of (26); (28) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 85 as VH; (29) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 87 as VL; (30) an antibody that comprises the H chain of (28) and the L chain of (29); (31) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 109 as CDR1, the amino acid sequence of SEQ ID NO: 111 as CDR2, and the amino acid sequence of SEQ ID NO: 113 as CDR3; (32) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 115 as CDR1, the amino acid sequence of SEQ ID NO: 117 as CDR2, and the amino acid sequence of SEQ ID NO: 119 as CDR3; (33) an antibody that comprises the H chain of (31) and the L chain of (32); (34) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 105 as VH; (35) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 107 as VL; (36) an antibody that comprises the H chain of (34) and the L chain of (35); (37) an antibody that comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of any one of (1) to (36), which has equivalent activity to the antibody of any one of (1) to (36); and (38) an antibody that binds to the epitope bound by the antibody of any one of (1) to (36).

8. The antibody of claim 7, wherein the antibody is a chimeric antibody or a humanized antibody.

9. A composition comprising the antibody of any one of claims 1 to 7 and a pharmaceutically acceptable carrier.

10. A method for preventing and/or treating cognitive impairment, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

11. A method for preventing and/or treating Alzheimer's disease, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

12. A method for suppressing the progression of Alzheimer's disease, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

13. A method for suppressing senile plaque formation, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

14. A method for suppressing A.beta. accumulation, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

15. A method for neutralizing neurotoxicity, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

16. A method for inhibiting A.beta. amyloid fibril formation, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

17. A method for neutralizing synaptic toxicity, which comprises administering the antibody of any one of claims 1 to 7 as an active ingredient.

18. A method for detecting an A.beta. oligomer, which comprises detecting an A.beta. oligomer contained in a sample collected from a subject using the antibody of any one of claims 1 to 7.

19. The method of claim 18, wherein the sample is blood or cerebrospinal fluid.

20. A method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises using the antibody of any one of claims 1 to 7 to detect an A.beta. oligomer in a sample collected from a subject.

21. A method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises: (a) contacting a sample collected from a subject with the antibody of any one of claims 1 to 7; and (b) measuring the amount of A.beta. oligomer in the sample, wherein the subject is determined to suffer from or be at a risk of developing Alzheimer's disease, when the amount measured in (b) is higher than that of a healthy individual.

22. A method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises: (a) contacting a sample collected from a subject with the antibody of any one of claims 1 to 7 and an antibody that binds to an A.beta. monomer; and (b) measuring the ratio of A.beta. oligomer to A.beta. monomer in the sample, wherein the subject is determined to suffer from or be at a risk of developing Alzheimer's disease, when the ratio measured in (b) is higher than that of a healthy individual.

23. The method of claim 20, wherein the sample is blood or cerebrospinal fluid.
Description



PRIORITY

[0001] The present application is a continuation of U.S. application Ser. No. 12/533,348, filed Jul. 31, 2009, now pending. U.S. application Ser. No. 12/533,348, is a nonprovisional of provisional U.S. Application No. 61/085,545, filed Aug. 1, 2008, now abandoned, and a continuation-in-part of International Application No. PCT/JP2009/052039, filed Feb. 6, 2009, now pending. The present application is also a continuation-in-part of International Application No. PCT/JP2009/052039, filed Feb. 6, 2009, now pending. International Application No. PCT/JP2009/052039 claims priority to Japanese Patent Application No. 2008-028386, filed Feb. 8, 2008, now abandoned, and Japanese Patent Application No. 2008-201058, filed Aug. 4, 2008, now abandoned. The entire contents of these applications are incorporated by reference herein.

FIELD OF THE INVENTION

[0002] The present invention relates to antibodies that specifically bind to A.beta. oligomers and uses thereof.

BACKGROUND OF THE INVENTION

[0003] Various evidence has shown that deterioration of memory arises from synaptic dysfunction triggered by soluble A.beta. oligomers (see Klein W L, Trends Neurosci. 24: 219-224, 2001; and Selkoe D J, Science 298: 789-791, 2002). Excessive accumulation and deposition of A.beta. oligomers may be the trigger for a series of pathological cascades that lead to Alzheimer's disease (AD). Therefore, therapeutic intervention targeting A.beta. oligomers may be effective for blocking these cascades. However, findings on core molecules of this amyloid cascade hypothesis which are responsible for neurodegeneration, particularly on neurodegeneration mediated by A.beta. oligomers, originate from in vitro experiments (see Hass C et al.: Nature Review 8: 101-12, 2007). This neurodegeneration has not been proven directly in vivo. The greatest defect of previously reported in vivo experiments is that they failed to demonstrate synaptic toxicity of endogenous A.beta. oligomers due to the lack of conformation-specific molecular tools (see Lee E B, et al.: J. Biol. Chem. 281: 4292-4299, 2006). There has been known no technique capable of proving the toxicity within the human brain, an aspect which is difficult to demonstrate even in Alzheimer's disease mouse models. Thus, the in vivo neurotoxicity of endogenous A.beta. has been often disregarded. It has been unknown why NFT formation and loss of nerve cells precede senile plaque formation in the human entorhinal cortex, and how A.beta.

SUMMARY OF THE INVENTION

[0004] The present invention was achieved in view of the above circumstances. An objective of the present invention is to provide antibodies that bind specifically to A.beta. oligomers, and uses thereof. More specifically, the present invention provides antibodies that bind specifically to A.beta. oligomers, methods for detecting A.beta. oligomers using the antibodies, methods for diagnosing Alzheimer's disease using the antibodies, and pharmaceutical agents comprising the antibodies.

[0005] The present inventors produced monoclonal antibodies that are specific to only soluble amyloid .beta. (A.beta.) oligomers and do not recognize soluble A.beta. monomers which are physiological molecules, and confirmed that the antibodies have the following:

(1) anti-neurotoxic activity; (2) activity to suppress A.beta. amyloid fibril formation; (3) specificity to recognize only A.beta. oligomers; (4) ability to capture A.beta. oligomers in AD brain; and (5) ability to prevent the development of Alzheimer's disease-like phenotypes (memory impairment, brain A.beta. accumulation) in APPswe transgenic mice (Tg2576).

[0006] Using an ultrafiltration/molecular sieve method, among the antibodies produced, monoclonal 1A9 and 2C3 were determined to specifically recognize oligomers of 30 kDa or more, mainly 100 kDa or more, but not monomers of approximately 4.5 kDa. The two antibodies were confirmed to have neurotoxicity-neutralizing activity by evaluating the neutralizing effect against A.beta. 1-42-induced neurotoxicity in PC12 cells differentiated into nerve cells. Thioflavin T assay and electron microscopy showed that the antibodies have activity to suppress A.beta. amyloid fibril formation. The ability of 1A9 and 2C3 to capture A.beta. oligomers in AD brain was confirmed by immunoprecipitation using the antibodies in the presence of SDS-stable 4-, 5-, 8-, and 12-mers. Furthermore, to determine the in vivo neurotoxicity in the human brain, the amount of polymers recognized by the antibodies was evaluated in the human entorhinal cortex mostly at Braak NFT Stages I to III. By particularly focusing on the 12-mer, which has been reported to have neurotoxicity in animal studies, it was confirmed that the polymer accumulation precedes the occurrence of cognitive impairment, and is increased with the progression of Braak NFT stage. This result shows for the first time that the 12-mer, which is specifically recognized by the antibodies, is a conformational assembly that causes in vivo neurotoxicity in the human brain. The present inventors also discovered that the oligomeric conformational structure recognized by the antibodies is present in cerebrospinal fluid (CSF), and is increased in AD patients. The present inventors used 1 A9 or 2C3 in passive immunotherapy by intravenous injection as with other neurological disorders. It was confirmed that Tg2576 mice are protected from memory impairment, senile plaque formation, synaptic dysfunction, and A.beta. accumulation by subchronic passive immunotherapy, without harmful side-effects. The results obtained by the present inventors demonstrated for the first time that monoclonal 1A9 and 2C3 are promising candidates for therapeutic antibodies for preventing Alzheimer's disease-like phenotypes in Tg2576 mice, which are expected to show their effect by conventional peripheral intravenous administration, and thus there is no need to consider brain transfer.

[0007] The present inventors also confirmed that passive immunotherapy using the 1A9 and 2C3 antibodies suppresses senile plaque amyloid formation and swollen dystrophic neurite formation. Furthermore, the present inventors discovered that a fraction of the 1A9 and 2C3 antibodies administered into the blood transfers into the brain.

[0008] As described above, the present inventors disclose herein that monoclonal 1A9 and 2C3, which are antibodies that specifically bind to A.beta. oligomers, fulfill all of the diagnostic/therapeutic antibody criteria, and are promising candidates for therapeutic antibodies for diagnosing/preventing Alzheimer's disease.

[0009] Furthermore, as with the 1A9 and 2C3 antibodies, the present inventors successfully obtained the 5A5, 5A9, 4F7, 4H5, 6E4, and 61-14 antibodies which bind specifically to A.beta. oligomers, but do not recognize A.beta. monomers. The present inventors discovered that these six types of antibodies have activity to neutralize A.beta.-induced neurotoxicity and to suppress A.beta. amyloid fibril formation.

[0010] The present inventors disclose that the above-mentioned 5A5, 5A9, 4F7, 4H5, 6E4, and 6H4 antibodies are promising candidates for therapeutic antibodies for diagnosing/preventing Alzheimer's disease.

[0011] More specifically, the present invention provides the following:

[1] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 1 and an L chain having the amino acid sequence of SEQ ID NO: 3; [2] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 21 and an L chain having the amino acid sequence of SEQ ID NO: 23; [3] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 41 and an L chain having the amino acid sequence of SEQ ID NO: 43; [4] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 61 and an L chain having the amino acid sequence of SEQ ID NO: 63; [5] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 81 and an L chain having the amino acid sequence of SEQ ID NO: 83; [6] an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 101 and an L chain having the amino acid sequence of SEQ ID NO: 103; [7] an antibody of any one of (1) to (38) below: (1) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 9 as CDR1, the amino acid sequence of SEQ ID NO: 11 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3; (2) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 15 as CDR1, the amino acid sequence of SEQ ID NO: 17 as CDR2, and the amino acid sequence of SEQ ID NO: 19 as CDR3; (3) an antibody that comprises the H chain of (1) and the L chain of (2); (4) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 5 as VH; (5) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 7 as VL; (6) an antibody that comprises the H chain of (4) and the L chain of (5); (7) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 29 as CDR1, the amino acid sequence of SEQ ID NO: 31 as CDR2, and the amino acid sequence of SEQ ID NO: 33 as CDR3; (8) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 35 as CDR1, the amino acid sequence of SEQ ID NO: 37 as CDR2, and the amino acid sequence of SEQ ID NO: 39 as CDR3; (9) an antibody that comprises the H chain of (7) and the L chain of (8); (10) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 25 as VH; (11) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 27 as VL; (12) an antibody that comprises the H chain of (10) and the L chain of (11); (13) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 49 as CDR1, the amino acid sequence of SEQ ID NO: 51 as CDR2, and the amino acid sequence of SEQ ID NO: 53 as CDR3; (14) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 55 as CDR1, the amino acid sequence of SEQ ID NO: 57 as CDR2, and the amino acid sequence of SEQ ID NO: 59 as CDR3; (15) an antibody that comprises the H chain of (13) and the L chain of (14); (16) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 45 as VH; (17) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 47 as VL; (18) an antibody that comprises the H chain of (16) and the L chain of (17); (19) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 69 as CDR1, the amino acid sequence of SEQ ID NO: 71 as CDR2, and the amino acid sequence of SEQ ID NO: 73 as CDR3; (20) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 75 as CDR1, the amino acid sequence of SEQ ID NO: 77 as CDR2, and the amino acid sequence of SEQ ID NO: 79 as CDR3; (21) an antibody that comprises the H chain of (19) and the L chain of (20); (22) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 65 as VH; (23) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 67 as VL; (24) an antibody that comprises the H chain of (22) and the L chain of (23); (25) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 89 as CDR1, the amino acid sequence of SEQ ID NO: 91 as CDR2, and the amino acid sequence of SEQ ID NO: 93 as CDR3; (26) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 95 as CDR1, the amino acid sequence of SEQ ID NO: 97 as CDR2, and the amino acid sequence of SEQ ID NO: 99 as CDR3; (27) an antibody that comprises the H chain of (25) and the L chain of (26); (28) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 85 as VH; (29) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 87 as VL; (30) an antibody that comprises the H chain of (28) and the L chain of (29); (31) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 109 as CDR1, the amino acid sequence of SEQ ID NO: 111 as CDR2, and the amino acid sequence of SEQ ID NO: 113 as CDR3; (32) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 115 as CDR1, the amino acid sequence of SEQ ID NO: 117 as CDR2, and the amino acid sequence of SEQ ID NO: 119 as CDR3; (33) an antibody that comprises the H chain of (31) and the L chain of (32); (34) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 105 as VH; (35) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 107 as VL; (36) an antibody that comprises the H chain of (34) and the L chain of (35); (37) an antibody that comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of any one of (1) to (36), which has equivalent activity to the antibody of any one of (1) to (36); and (38) an antibody that binds to the epitope bound by the antibody of any one of (1) to (36); [8] the antibody of [7], wherein the antibody is a chimeric antibody or a humanized antibody; [9] a composition comprising the antibody of any one of [1] to [8] and a pharmaceutically acceptable carrier; [10] an agent against cognitive impairment, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [11] a therapeutic agent for Alzheimer's disease, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [12] an agent for suppressing the progression of Alzheimer's disease, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [13] an agent for suppressing senile plaque formation, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [14] an agent for suppressing A.beta. accumulation, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [15] an anti-neurotoxic agent, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [16] an agent for inhibiting A.beta. amyloid fibril formation, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [17] an agent against synaptic toxicity, which comprises the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [18] a method for detecting an A.beta. oligomer, which comprises the step of detecting an A.beta. oligomer contained in a sample collected from a subject using the antibody of any one of [1] to [8]; [19] a method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises using the antibody of any one of [1] to [8] to detect an A.beta. oligomer in a sample collected from a subject; [20] a method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises the steps of: (a) contacting a sample collected from a subject with the antibody of any one of [1] to [8]; and (b) measuring the amount of A.beta. oligomer in the sample, wherein the subject is determined to suffer from or be at a risk of developing Alzheimer's disease, when the amount measured in step (b) is higher than that of a healthy individual; [21] a method of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprises the steps of: (a) contacting a sample collected from a subject with the antibody of any one of [1] to [8] and an antibody that binds to an A.beta. monomer; and (b) measuring the ratio of A.beta. oligomer to A.beta. monomer in the sample, wherein the subject is determined to suffer from or be at a risk of developing Alzheimer's disease, when the ratio measured in step (b) is higher than that of a healthy individual; [22] the method of any one of [18] to [21], wherein the sample is blood or cerebrospinal fluid; [23] a pharmaceutical agent for use in the method of any one of [18] to [21]; and [24] a kit for use in the method of any one of [18] to [21].

[0012] Furthermore, the present invention provides the following:

[25] a method for preventing and/or treating cognitive impairment, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [26] a method for preventing and/or treating Alzheimer's disease, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [27] a method for suppressing the progression of Alzheimer's disease, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [28] a method for suppressing senile plaque formation, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [29] a method for suppressing A.beta. accumulation, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [30] a method for neutralizing neurotoxicity, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [31] a method for inhibiting A.beta. amyloid fibril formation, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [32] a method for neutralizing synaptic toxicity, which comprises the step of administering the antibody of any one of [1] to [8] or the composition of [9] as an active ingredient; [33] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent against cognitive impairment; [34] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of a therapeutic agent for Alzheimer's disease; [35] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for suppressing the progression of Alzheimer's disease; [36] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for suppressing senile plaque formation; [37] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for suppressing A.beta. accumulation; [38] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for neutralizing (suppressing) neurotoxicity; [39] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for inhibiting A.beta. amyloid fibril formation; [40] use of the antibody of any one of [1] to [8] or the composition of [9] in the production of an agent for neutralizing (suppressing) synaptic toxicity; [41] the antibody of any one of [1] to [8] or the composition of [9] for use in preventing and/or treating cognitive impairment; [42] the antibody of any one of [1] to [8] or the composition of [9] for use in preventing and/or treating Alzheimer's disease; [43] the antibody of any one of [1] to [8] or the composition of [9] for use in suppressing the progression of Alzheimer's disease; [44] the antibody of any one of [1] to [8] or the composition of [9] for use in suppressing senile plaque formation; [45] the antibody of any one of [1] to [8] or the composition of [9] for use in suppressing A.beta. accumulation; [46] the antibody of any one of [1] to [8] or the composition of [9] for use in neutralizing (suppressing) neurotoxicity; [47] the antibody of any one of [1] to [8] or the composition of [9] for use in inhibiting A.beta. amyloid fibril formation; and [48] the antibody of any one of [1] to [8] or the composition of [9] for use in neutralizing (suppressing) synaptic toxicity.

BRIEF DESCRIPTION OF THE DRAWINGS

[0013] FIG. 1 presents photographs and a graph showing the results of production and characteristic determination of oligomer-specific antibodies. A: Electrophoresis of immunogens. The A.beta. 1-42 tetramer (black arrowhead) which is free of contamination of the A.beta. 1-42 monomer (outlined arrowhead) was isolated using SDS-PAGE. Lane 1: A.beta. 1-42 dissolved in 10 mM phosphate buffer; and Lane 2: A.beta. 1-42 dissolved in distilled deionized water. B: A.beta. amyloid, which is insoluble in a buffer but can be extracted using formic acid from the brain of Alzheimer's disease patients, was immunoprecipitated using the supernatant of a positive hybridoma cell culture, and the immune complex was selectively separated using protein-G agarose (Amersham). Nine clones were tested; lane 2 (asterisk) is 1A9 and lane 6 (double asterisk) is 2C3. C: Elution profile of SEC of a conditioned medium. Among the 24 SEC-collected fractions, fractions 8, 13, and 16 were subjected to 1A9 immunoprecipitation. A.beta. immunoreactivity was detected using 4G8. The black arrowhead indicates the trimer and the outlined arrowhead indicates the dimer. Asterisk (*) indicates the anti-mouse IgG light chain.

[0014] FIG. 2 presents photographs and a graph showing the antitoxic activity of 1A9 and 2C3. A to F: Representative images of NGF-treated PC12 (PC12N) cells, which were exposed to seed-free A.beta. 1-42 at 37.degree. C. for 48 hours in the presence or absence of the antibodies (left half of each panel). Representative calcein AM/PI staining where live cells were stained green and dead cells were stained red (right half of each panel). G: The viability of cells exposed to seed-free A.beta. 1-42 (25 .mu.M) with the following antibodies: non-specific IgG2b (filled square); 4G8 (open triangle); 1A9 (open square); and 2C3 (filled circle).

[0015] FIG. 3 presents photographs and a graph showing the size and morphological characteristics of the toxic A.beta. assemblies targeted by 1A9 and 2C3. A: The 540,000.times.g supernatant of A.beta. 1-42 (25 .mu.M) was subjected to a continuous molecular sieving process using ultrafiltration membranes having a molecular weight cutoff value of 3, 10, 30, and 100 kDa (Microcon). The four types of filtrates thus fractioned were named as follows: fraction 1 (<3 kDa), fraction 2 (3 to 10 kDa), fraction 3 (10 to 30 kDa), fraction 4 (30 to 100 kDa); and fraction 5 (>100 kDa) which was finally retained. The presence of A.beta. 1-42 in each of the above-mentioned fractions was detected by 4G8 immunoblotting. B: Representative images of NGF-treated PC12 (PC12N) cells treated with the five fractions at 37.degree. C. for 48 hours. The toxicity of each fraction was evaluated as described above for FIG. 2. C: The viability of cells treated with the 540,000.times.g supernatant of A.beta. 1-42 and the five fractions (fractions 1 to 5). Similar results were obtained from two independent experiments. The values are presented in percentage (mean.+-.SD) with respect to the control. D: Dot blot analysis of the five fractions (fractions 1 to 5). The blots were reacted with A11, 1A9, 2C3, and 4G8. E: AFM images of the five fractions. In fraction 5 (Fr. 5) that had the strongest toxicity, ring-shaped and bead-shaped structures were observed in addition to granular molecules.

[0016] FIG. 4 presents photographs and graphs showing the activity of 1A9 and 2C3 to suppress A.beta. amyloid fibril formation. A: Amyloid fibril formation of A.beta. 1-42 at various concentrations (10 .mu.M (open square), 25 .mu.M (filled diamond), and 50 .mu.M (open circle)) was monitored by ThT assay at 37.degree. C. for up to 72 hours. B: Coexisting antibody dose-dependent inhibition of amyloid fibril formation of A.beta. 1-42 was observed for 2C3 (open circle). In contrast, the 1A9 (open square), 4G8 (filled triangle), and non-specific IgG (filled square) antibodies did not inhibit fibril-forming assembly of seed-free A.beta. 1-42 (ThT-negative 540,000.times.g supernatant). C: Coexisting antibody dose-dependent inhibition of fibril-forming assembly of A.beta. 1-42 was observed for 2C3 (open circle), and nearly complete inhibition was observed also for 1A9 (open square) at 3 .mu.M. D: None of the test antibodies added after a 24-hour pre-incubation for A.beta. 1-42 amyloid fibril formation could dissolve nor disassemble the A.beta. 1-42 amyloid fibrils. E to G: EM images of A.beta. 1-42 in the absence (Panel E) and presence of 2C3 (Panel F) and 1A9 (Panel G).

[0017] FIG. 5 presents photographs and graphs on toxicity-related A.beta. 1-42 oligomers. A: Dot blot assay (upper half of Panel A): A.beta. 1-42 monomers (25 .mu.M) were incubated for a specified time (0 to 72 hours) at 37.degree. C., and immobilized onto a nitrocellulose membrane, and subjected to dot blot assay that uses A11, 1A9, 2C3, or 4G8. The emergence of immunoreactivity-positive structures for each antibody was tested. Immunoreactivity intensity analysis (lower half of Panel A): The results of dot blot assay were analyzed semiquantitatively using the Multi Gauge v 3.0 software (Fuji Film, Tokyo). To correlate the oligomer formation and amyloid fibril formation, the ThT fluorescence value (the right Y axis) was overlaid on the same time axis. B: The A.beta. 1-42 assembly after 0-, 2-, 4-, and 24-hour incubation at 37.degree. C., and the change in A.beta. 1-42 assembly after further 48-hour incubation. The A.beta. 1-42 assembly was detected by 4G8 immunoblotting. C: The toxic activity of the above-mentioned various A.beta. 1-42 assemblies. The viability of nerve cells was determined by the LIVE/DEAD assay as described for FIG. 2. D: The anti-neurotoxic activity of 1A9 and 2C3 was evaluated using various A.beta. assemblies (the A.beta. 1-42 assemblies formed at 37.degree. C. for 0 and 2 hours ("0h" and "2h"); and the ThT-positive supernatant collected after ultracentrifugation at 540,000.times.g for two hours ("2h sup")). Representative images of PC12N cells exposed to various A.beta. 1-42 assemblies in the absence or presence of the antibodies are shown in the left half of Panel D (a: "0h"; b: "2h"; c: "2h sup"; d: "2h sup"+IgG2b; e: "2h sup"+1A9; f: "2h sup"+2C3). The viability of cells exposed to various A.beta. 1-42 assemblies in the absence or presence of the antibodies is presented in percentage (mean.+-.SD) with respect to the control, and this is shown in the right half of Panel D. Compared to the "0h" A.beta. 1-42 assembly, the "2h" A.beta. 1-42 assembly lowered the neurotoxicity. "2h sup" recovered the neurotoxicity to a degree similar to that of the "0h" A.beta. 1-42 assembly. Non-specific IgG2b could not block the neurotoxicity induction of the "2h sup" A.beta. 1-42 assembly. Monoclonal 1A9 completely inhibited the "2h sup"-induced neurotoxicity, while the ability of 2C3 to inhibit the toxicity was slightly inferior. In the experiments using the two monoclonal antibodies (mAbs), the antitoxic activity of the mAbs was observed at a mAb:A.beta. mole ratio of 1:<25 to 50. This suggests that structurally different 1A9- and 2C3-recognized oligomeric assemblies exist at a relatively low concentration.

[0018] FIG. 6 presents photographs and graphs showing that soluble 1A9- and 2C3-recognized oligomers exist in the human brain. Antibodies against A.beta. oligomers can detect senile plaques and vascular amyloids in AD brain only after pretreatment with Protease K. A: 1A9 staining; B: 2C3 staining; and C: A11 staining. D: 4G8 immunoblotting of 1A9- or 2C3-immunoprecipitated A.beta. in buffer-soluble AD brain (lanes 1, 2, 4, and 5) and healthy control brain (lanes 3 and 6). Representative results for 1A9 and 2C3 are shown in the left and right half of the panel, respectively. E and F: Semiquantitative analysis (with actin control) of soluble 1A9-immunoreactive 12-mer (Panel E) and soluble 2C3-immunoreactive 12-mer (Panel F) in the human entorhinal cortex obtained from 50 autopsy cases of a healthy elderly population (Braak NFT Stage I or II: n=35; Braak NFT Stage III or IV: n=13; and Braak NFT Stage >IV, AD cases: n=2).

[0019] FIG. 7-1 present graphs showing that soluble 1A9- and 2C3-recognized oligomers exist in human CSF. Pooled whole cerebrospinal fluid (CSF) (AD=10 and NC=10) (Panels A and B) and pooled lipoprotein-depleted CSF (AD=10, and NC=10) (Panels C and D) were subjected to size exclusion chromatography (SEC). In Panels A and B, the collected fractions were analyzed for the distribution of A.beta. 40 and A.beta. 42 monomers by BNT77-BA27 and BNT77-BC05 ELISAs. Panels C and D show the presence of A.beta. 40 and A.beta. 42 oligomers captured by 1A9/2C3 mixed antibodies.

[0020] FIG. 7-2 is the continuation of FIG. 7-1. The amount of 1A9-recognized oligomeric assembly (1A9-BC05 and 1A9-BA27 ELISAs) and the amount of 2C3-recognized assembly (2C3-BC05 and 2C3-BA27 ELISAs) were measured for 12 AD cases (open circle) and 13 NC cases (filled circle) (Panels E and G). The oligomer/monomer ratio is shown in Panels F (1A9) and H (2C3).

[0021] FIG. 8 presents graphs showing that the onset of memory impairment in Tg2576 mice can be prevented by passive immunization treatment. 13-month-old Tg2576 mice were divided into the following three groups to perform learning/behavior tests: PBS-administered group: n=10; 1A9-administered group: n=13; and 2C3-administered group: n=11. All of the measured values were indicated as mean.+-.SE. (A) Y-maze test. Spontaneous alteration behavior was monitored in each group during an eight-minute session of the Y-maze task. The results of one-way ANOVA were as follows: F(1, 52)=3.09, p<0.05; * p<0.05 in the comparison with PBS-administered Tg2576 mice. (B) Novel object recognition test. The retention session was performed 24 hours after training. The exploratory preference in a ten-minute session in the novel object recognition test was determined in each group. The results of two-way ANOVA were as follows: training/retention, F(1, 64)=31.53, p<0.01; animal group, F(2, 64) .about.7.49, p<0.01; repeated training/retention by the animal group, F(2, 64)=10.12, p<0.01; ** p<0.01 in the comparison with the corresponding untrained mice, ## p<0.01 in the comparison with PBS-administered Tg2576 mice. (C) The swimming path length during a 60-second session of water maze test was measured for each group. The results of two-way ANOVA were as follows: trial, F(9, 320)=20.46, p<0.01; animal group, F(2, 320)=12.59, p<0.01; repeated trial by the animal group, F(18, 320)=1.78, p<0.05; p<0.05, ** p<0.01 in the comparison with PBS-administered Tg2576 mice. Fear-conditioned learning test: Context-dependent (D) and clue-dependent freezing times (E) were determined. The results of two-way ANOVA were as follows: context-dependent test, F(2, 32)=5.94, p<0.01; clue-dependent test, F(2, 32)=7.33, p<0.01; * p<0.05 and ** p<0.01 in the comparison with PBS-administered Tg2576 mice.

[0022] FIG. 9 presents graphs and a photograph showing that the brain A.beta. accumulation in Tg2576 can be prevented by passive immunotherapy. The hippocampus and cerebral cortex of three groups of 13-month-old Tg2576 mice (PBS-administered group, n=10; 1A9-administered group, n=13; and 2C3-administered group, n=11) were extracted in three continuous steps to prepare the buffer-soluble, SDS-soluble, and formic acid (FA)-extractable fractions. Each of the fractions was subjected to A.beta.-specific ELISAs (WAKO kit: BNT77-BA27 for A.beta. x-40; BNT77-BC05 for A.beta. x-42). The accumulation of A.beta. 40 (SDS and FA) and A.beta. 42 (SDS) was found to be significantly suppressed only in the 1A9-treated group. The accumulation-suppressing effect for the A11-positive oligomer (4-mer) was confirmed in the SDS-soluble cerebral cortex fractions from the two antibody-treated groups.

[0023] FIG. 10 presents photographs and graphs on A.beta. oligomers in the plasma and brain of Tg2576. A and B: As a result of ELISA analysis, no significant difference in the amount of A.beta. x-40 and A.beta. x-42 in the plasma was observed between the PBS-administered group and the immunotherapy group. C: Similarly, no difference in the A.beta. 40/A.beta. 42 ratio was observed among the three groups tested. D: As a result of dot blot analysis using pooled brain homogenates, no difference in the amount of physiological saline-soluble A11-positive oligomer was observed among the three groups tested. Hippocampus (left panel) and cerebral cortex (right panel). PBS-administered group, n=10; 1A9-administered group, n=13; and 2C3-administered group, n=11. E: According to immunoblot analysis using the anti-oligomer A11 antibody, the immunoreactivity of the A.beta. tetramer in the SDS-extracted cerebral cortex fraction (right panel) was decreased in the 1A9- and 2C3-administered groups compared to the PBS-administered group. On the other hand, this was not observed in the hippocampus (left panel). F: Blood (albumin-depleted plasma, upper part of Panel F;

albumin/lipoprotein-depleted plasma, lower part of Panel F) was pooled from each of the groups, and subjected to A11 dot blot analysis. As a result, the A11 immunoreactivity was found to be increased in the 1A9- and 2C3-administered groups compared to the PBS-administered group (Panel F). The proportion of the lipoprotein-bound form of 2C3-recognized oligomers was higher than that of 1A9-recognized oligomers (lower part of Panel F). Furthermore, the A11 immunoblotting also showed positive signals at approximately 200 kDa, and the immunoreactivity was clearly increased in the 1A9- and 2C3-administered groups compared to the PBS-administered group (Panel G). From these results, it is conceivable that the therapeutic effect selective only to target A.beta. oligomer molecules was obtained in the antibody-administered groups without affecting physiological molecules.

[0024] FIG. 11 presents photographs and graphs showing that senile plaque amyloid formation (A: A.beta.-specific antibody staining; and B: thioflavin-S-positive analysis) and swollen dystrophic neurite formation (C: synaptophysin-positive analysis) were suppressed in the Tg2576 mouse brain by passive immunization treatment.

[0025] FIG. 12 presents photographs showing the suppression of synaptic degeneration by passive immunization treatment with 1A9 and 2C3. Immunostaining of synaptophysin (left panels) and drebrin (right panels) in presynaptic and postsynaptic dot-like peripheral cells. Top: PBS administration; middle: 1A9 administration; and bottom: 2C3 administration.

[0026] FIG. 13 presents photographs showing the brain transfer of the antibodies by passive immunization treatment. The distribution of administered antibodies in the Tg2576 mouse brain is shown. Staining with anti-A.beta. antibodies (left panels) and IgG (center panels). 1A9 administration (A), 2C3 administration (B), and PBS administration (C).

[0027] FIG. 14 presents photographs showing, by dot blot analysis, that the monoclonal antibodies 5A5, 5A9, 4F7, 4H5, 6E4, and 6H4 are specific to A.beta. oligomers (3 to 96 hours), but do not recognize A.beta. monomers (0 hour).

[0028] FIG. 15 presents graphs showing the A.beta. oligomer-selective binding ability of the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4). The vertical axis indicates the absorbance at a wavelength of 450 nm, and the horizontal axis indicates the concentration of A.beta. oligomer or A.beta. monomer used as an inhibitor. In each graph, the dashed line indicates the antibody-binding activity when the A.beta. oligomer was used as the inhibitor, and the solid line indicates the antibody-binding activity when the A.beta. monomer was used as the inhibitor.

[0029] FIG. 16 presents graphs showing the neutralizing activity of the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) against A.beta.-induced neurotoxicity. The horizontal axis indicates the amount of antibody added, and the vertical axis shows the cytotoxicity relative to that under the antibody-free condition as the standard (see the equation in the figure). Control IgG (3F1), which is an antibody that does not bind to A.beta. 42, was used for comparison.

[0030] FIG. 17 presents graphs showing the suppressing activity of the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) against A.beta. amyloid fibril formation. The antibodies were added at three different concentrations to a A.beta. 1-42 solution (12.5 .mu.M). After incubation at 37.degree. C. for 24 hours, the level of A.beta. amyloid fibril formation was measured by the ThT fluorescence intensity method. The horizontal axis indicates the amount of antibody added, and the vertical axis shows the level of amyloid fibril formation by the antibody addition that is relative to the level of amyloid fibril formation without antibody addition as the standard.

DETAILED DESCRIPTION OF THE INVENTION

[0031] The present invention will be described more specifically below.

[0032] As described above, the present inventors succeeded in obtaining antibodies that bind specifically to A.beta. oligomers but not to A.beta. monomers. That is, the present invention provides antibodies that bind to A.beta. oligomers but not to A.beta. monomers. The antibodies are preferably isolated or purified.

[0033] The terms "isolated" and "purified" used for substances (antibodies and such) of the present invention indicate that the substances do not substantially include at least one other substance that may be contained in the natural source. Therefore, "isolated antibodies" and "purified antibodies" refer to antibodies that do not substantially include cell materials such as hydrocarbons, lipids, or other contaminant proteins from the cell or tissue source from which the antibodies (proteins) are derived. When the antibodies are chemically synthesized, the terms refer to antibodies that do not substantially include chemical precursor substances or other chemical substances. In a preferred embodiment, the antibodies of the present invention are isolated or purified.

[0034] "Antibodies" refers to glycoproteins that have the same structural characteristics. Antibodies show binding specificity towards specific antigens. Herein, "antigens" refers to proteins that have the ability to bind to the corresponding antibodies, and induce antigen-antibody reactions in vivo.

[0035] A.beta. proteins, which are the major constituents of amyloids, are peptides consisting of 40 to 42 amino acids, and are known to be produced from precursor proteins called amyloid precursor proteins (APPs) by the action of proteases. Besides amyloid fibrils collected in ultracentrifuged sediment fractions, the amyloid molecules produced from APPs include oligomeric non-fibrous assemblies in addition to soluble monomers. "A.beta. oligomers" of the present invention refer to non-fibrous assemblies. The "A.beta. oligomers" of the present invention include, for example, A.beta. 40 (A.beta. 1-40) oligomers and A.beta.42 (A.beta. 1-42) oligomers. For example, "A.beta.42 oligomers" of the present invention are molecules showing a molecular weight of 45 to 160 kDa in SDS-PAGE, and 22.5 to 1,035 kDa in Blue Native PAGE. Using molecular sieves, the molecules are collected mainly in the >100 kDa retention solution. When observed under an atomic force microscope, the molecules show mixed morphologies of granular, bead-shaped, and ring-shaped molecules having a height of 1.5 to 3.1 nm. By the gel filtration method, the molecules can be eluted in the void volume fraction 8 with a molecular weight of 680 kDa or more, and in fraction 15 with a molecular weight of 17 to 44 kDa.

[0036] There is no limitation on the origin and form of the antibodies used in the present invention as long as they bind to A.beta. oligomers but not to A.beta. monomers.

[0037] "Antibodies" of the present invention include both monoclonal and polyclonal antibodies. The antibodies of the present invention also include any type of antibodies such as non-human animal antibodies, humanized antibodies, chimeric antibodies, human antibodies, the later-described minibodies, amino acid sequence-modified antibodies, modified antibodies conjugated to other molecules (for example, polymers such as polyethylene glycol), and sugar chain-modified antibodies.

[0038] Herein, the term "monoclonal antibodies" refers to antibodies that are obtained from a substantially homogeneous population of antibodies. That is, the individual antibodies constituting the population are identical with the exception of possible natural mutants that may be present in a trace amount. Monoclonal antibodies are highly specific and recognize a single antigenic site. Each of the monoclonal antibodies recognizes a single determinant of the antigen, in contrast to conventional (polyclonal) antibody preparations that typically contain different antibodies against different antigenic determinants (epitopes).

[0039] In addition to the above-mentioned specificity, monoclonal antibodies have the advantage that they can be synthesized from a hybridoma culture that is not contaminated with other immunoglobulins. Therefore, "monoclonal" indicates the characteristics of antibodies that can be obtained from a substantially homogeneous antibody population. This term does not indicate the requirement for any specific method for antibody production.

[0040] Basically, monoclonal antibodies can be produced by using known techniques. For example, they may be produced by the hybridoma method first described by Kohler and Milstein (Nature 256: 495-7, 1975), or by the recombinant DNA method (Cabilly et al., Proc. Natl. Acad. Sci. USA 81:3273-7, 1984), but the methods are not limited thereto. For example, when using the hybridoma method, an A.beta. oligomer (for example, the A.beta. tetramer described in the Examples) is used as a sensitizing antigen, and immunization is carried out according to a conventional immunization method. The obtained immune cells are fused with known parent cells by a conventional cell fusion method, and monoclonal antibody-producing cells can be screened and isolated using a conventional screening method.

[0041] The monoclonal antibodies of the present invention can be produced as follows. Synthetic A.beta. 1-42 (Peptide Institute, Inc., Osaka) is dissolved in distilled deionized water or a 10 mM phosphate buffer solution, and this is incubated at 37.degree. C. for 18 hours. Then, the peptides are separated by 4-12% SDS-PAGE, and visualized by CBB staining, and the portion of the A.beta. 1-42 tetramer alone which is not contaminated with the A.beta. 1-42 monomer is cut out and used as an antigen. On the other hand, a preparation containing a large amount of the A.beta. 1-40 oligomer is prepared by mixing (i) a modified A.beta. 1-40 prepared by chemically linking 6-carboxytetramethylrhodamine (6-TAMRA) (SIGMA) to the N terminus of a synthetic A.beta. 1-40 peptide using a conventional method with (ii) synthetic A.beta. 1-40 (Peptide Institute, Inc., Osaka) at a ratio of 5:100, 10:100, 20:100, 30:100, 40:100, 50:100, 60:100, 70:100, or 80:100, preferably 90:100, or more preferably 100:100, and carrying out polymerization reaction for three hours, preferably six hours, or more preferably 20 hours. Next, Balb-c mice are immunized with 2.5 .mu.g of either the A.beta. 1-42 tetramer or A.beta. 1-40 oligomer emulsified using complete Freund's adjuvant by injecting the antigen into their foot pad. Subsequently, booster immunizations are carried out six times. Hybridomas are produced from the inguinal lymph node by fusion with Sp2/O--Ag14 cells using Polyethylene Glycol 1500.

[0042] The animals immunized with sensitizing antigens are not particularly limited, but are preferably selected considering the compatibility with parent cells used for cell fusion. Generally, rodents, lagomorphs, or primates are used. Rodents include, for example, mice, rats, and hamsters. Lagomorphs include, for example, rabbits. Primates include, for example, Catarrhini (old-world) monkeys such as Macaca fascicularis, Macaca mulatta, hamadryas, and chimpanzees.

[0043] Animals are immunized with sensitizing antigens according to known methods. For example, as a standard method, immunization is performed by intraperitoneal or subcutaneous injection of a sensitizing antigen into mammals.

[0044] An example of the parent cells fused with the aforementioned immunocytes is the Sp2/O--Ag14 cell, which will be described below in the Examples. However, various other known cell lines can be used.

[0045] Cell fusion between the aforementioned immunocyte and a myeloma cell can be carried out basically according to known methods including the method by Kohler and Milstein (Kohler G. and Milstein C., Methods Enzymol. (1981) 73, 3-46).

[0046] Hybridomas obtained in this manner are selected by culturing them in a conventional selection culture medium such as a HAT culture medium, which contains hypoxanthine, aminopterin, and thymidine. Culturing in the above-mentioned HAT culture medium is generally continued for several days to several weeks for an adequate time for killing cells other than the desired hybridomas (non-fused cells). Next, a conventional limiting dilution method is performed for screening and singly-cloning of a hybridoma that produces the desired antibody.

[0047] Thereafter, the obtained hybridoma is transplanted into the abdominal cavity of a mouse, and ascitic fluid containing the desired monoclonal antibodies is extracted. For example, the antibodies can be purified from the ascitic fluid by conventional protein separation and/or purification methods such as a selected combination of column chromatography including, but not limited to, affinity chromatography, filtration, ultrafiltration, salt precipitation, dialysis, SDS polyacrylamide gel electrophoresis, and isoelectric focusing (Antibodies: A Laboratory manual, Harlow and David, Lane (edit.), Cold Spring Harbor Laboratory, 1988).

[0048] Protein A columns and Protein G columns can be used for affinity columns. Examples of the Protein A columns used include Hyper D, POROS, and Sepharose F.F. (Pharmacia).

[0049] Chromatography (excluding affinity chromatography) includes ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse-phase chromatography, and adsorption chromatography ("Strategies for Protein Purification and Characterization: A Laboratory Course Manual", Daniel R Marshak et al., Cold Spring Harbor Laboratory Press, 1996). When chromatography is carried out, liquid-phase chromatography methods such as HPLC and FPLC can be used.

[0050] Monoclonal antibody-producing hybridomas prepared in this manner can be subcultured in a conventional culture medium, and they can be stored for a long time in liquid nitrogen.

[0051] Any mammal can be immunized using an immunogen for antibody production. However, when preparing monoclonal antibodies by producing hybridomas, the compatibility with parent cells used in cell fusion for hybridoma production is preferably considered.

[0052] Generally, rodents, lagomorphs, or primates are used for the immunization. Rodents include, for example, mice, rats, and hamsters. Lagomorphs include, for example, rabbits. Primates include, for example, Catarrhini (old-world) monkeys such as Macaca fascicularis, Macaca mulatta, hamadryas, and chimpanzees.

[0053] The use of transgenic animals that have a human antibody gene repertoire is known in the art (Ishida I, et al., Cloning and Stem Cells 4: 91-102, 2002). As with other animals, to obtain human monoclonal antibodies, the transgenic animals are immunized, then antibody-producing cells are collected from the animals and fused with myeloma cells to produce hybridomas, and anti-protein human antibodies can be prepared from these hybridomas (see International Publication Nos. WO92/03918, WO94/02602, WO94/25585, WO96/33735, and WO96/34096).

[0054] Alternatively, lymphocytes immortalized with oncogenes may be used for monoclonal antibody production. For example, human lymphocytes infected with EB virus or such is immunized in vitro with immunogens. Next, the immunized lymphocytes are fused with human-derived myeloma cells (U266, etc) capable of unlimited division, and thus hybridomas that produce the desired human antibodies are obtained (Japanese Patent Application Kokai Publication No. (JP-A) S63-17688 (unexamined, published Japanese patent application)).

[0055] Once monoclonal antibodies can be obtained by any of the aforementioned methods, the antibodies may also be prepared using genetic engineering methods (see, for example, Borrebaeck C A K and Larrick J W, Therapeutic Monoclonal Antibodies, MacMillan Publishers, UK, 1990). For example, recombinant antibodies may be prepared by cloning DNAs that encode the desired antibodies from antigen-producing cells such as hybridomas or immunized lymphocytes that produce the antibodies, then inserting the cloned DNAs into appropriate vectors, and transfecting the vectors into suitable host cells. Such recombinant antibodies are also included in the present invention.

[0056] Examples of the monoclonal antibodies of the present invention include the 1A9 monoclonal antibody, 2C3 monoclonal antibody, 5A5 monoclonal antibody, 5A9 monoclonal antibody, 4F7 monoclonal antibody, 4H5 monoclonal antibody, 6E4 monoclonal antibody, and 6H4 monoclonal antibody. Preferably, the monoclonal antibodies include the antibodies of (i) to (vi) below:

(i) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 1 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 3; (ii) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 21 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 23; (iii) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 41 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 43; (iv) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 61 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 63; (v) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 81 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 83; (vi) an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 101 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 103.

[0057] In an embodiment, the antibodies of the present invention include minibodies. A minibody contains an antibody fragment lacking a portion of a whole antibody, and is not particularly limited as long as it has the ability to bind to an antigen. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv. Examples of minibodies include Fab, Fab', F(ab')2, Fv, scFv (single chain Fv), diabody, and sc(Fv)2 (single chain (Fv)2).

[0058] To obtain polyclonal antibodies against the proteins of the present invention, blood is removed from a mammal sensitized with an antigen after the serum level of the desired antibody is confirmed to be increased. Serum is separated from blood by a known method. When a polyclonal antibody is used, serum containing the polyclonal antibody may be utilized. Alternatively, if necessary, a fraction containing the polyclonal antibody may be isolated from serum and then used. For example, immunoglobulin G or M can be prepared by obtaining a fraction that specifically recognizes a protein of the present invention using an affinity column coupled with the protein, and then purifying this fraction using a Protein A or Protein G column.

[0059] In the present invention, the antibody that binds to an A.beta. oligomer is an antibody binding to an A.beta. oligomer that binds 1A9, 2C3, 5A5, 5A9, 4F7, 4H5, 6E4, or 6H4. Preferably, the antibody is any one of the antibodies of (A) to (F) below:

(A) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 1 and an L chain having the amino acid sequence of SEQ ID NO: 3; (B) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 21 and an L chain having the amino acid sequence of SEQ ID NO: 23; (C) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 41 and an L chain having the amino acid sequence of SEQ ID NO: 43; (D) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 61 and an L chain having the amino acid sequence of SEQ ID NO: 63; (E) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 81 and an L chain having the amino acid sequence of SEQ ID NO: 83; and (F) an antibody binding to an A.beta. oligomer that binds to an antibody comprising an H chain having the amino acid sequence of SEQ ID NO: 101 and an L chain having the amino acid sequence of SEQ ID NO: 103.

[0060] Furthermore, the present invention provides A.beta. oligomers to which the antibodies of the present invention bind. Preferably, the antibodies include, for example, the 1A9 monoclonal antibody, 2C3 monoclonal antibody, 5A5 monoclonal antibody, 5A9 monoclonal antibody, 4F7 monoclonal antibody, 4H5 monoclonal antibody, 6E4 monoclonal antibody, and 6H4 monoclonal antibody. Such A.beta. oligomers can be used as antigens for preparing antibodies, or vaccines.

[0061] In a preferred embodiment, the antibodies of the present invention include, for example, the antibody of any one of (1) to (38) below:

(1) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 9 as CDR1, the amino acid sequence of SEQ ID NO: 11 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3; (2) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 15 as CDR1, the amino acid sequence of SEQ ID NO: 17 as CDR2, and the amino acid sequence of SEQ ID NO: 19 as CDR3; (3) an antibody that comprises the H chain of (1) and the L chain of (2); (4) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 5 as VH; (5) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 7 as VL; (6) an antibody that comprises the H chain of (4) and the L chain of (5); (7) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 29 as CDR1, the amino acid sequence of SEQ ID NO: 31 as CDR2, and the amino acid sequence of SEQ ID NO: 33 as CDR3; (8) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 35 as CDR1, the amino acid sequence of SEQ ID NO: 37 as CDR2, and the amino acid sequence of SEQ ID NO: 39 as CDR3; (9) an antibody that comprises the H chain of (7) and the L chain of (8); (10) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 25 as VH; (11) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 27 as VL; (12) an antibody that comprises the H chain of (10) and the L chain of (11); (13) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 49 as CDR1, the amino acid sequence of SEQ ID NO: 51 as CDR2, and the amino acid sequence of SEQ ID NO: 53 as CDR3; (14) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 55 as CDR1, the amino acid sequence of SEQ ID NO: 57 as CDR2, and the amino acid sequence of SEQ ID NO: 59 as CDR3; (15) an antibody that comprises the H chain of (13) and the L chain of (14); (16) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 45 as VH; (17) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 47 as VL; (18) an antibody that comprises the H chain of (16) and the L chain of (17); (19) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 69 as CDR1, the amino acid sequence of SEQ ID NO: 71 as CDR2, and the amino acid sequence of SEQ ID NO: 73 as CDR3; (20) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 75 as CDR1, the amino acid sequence of SEQ ID NO: 77 as CDR2, and the amino acid sequence of SEQ ID NO: 79 as CDR3; (21) an antibody that comprises the H chain of (19) and the L chain of (20); (22) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 65 as VH; (23) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 67 as VL; (24) an antibody that comprises the H chain of (22) and the L chain of (23); (25) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 89 as CDR1, the amino acid sequence of SEQ ID NO: 91 as CDR2, and the amino acid sequence of SEQ ID NO: 93 as CDR3; (26) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 95 as CDR1, the amino acid sequence of SEQ ID NO: 97 as CDR2, and the amino acid sequence of SEQ ID NO: 99 as CDR3; (27) an antibody that comprises the H chain of (25) and the L chain of (26); (28) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 85 as VH; (29) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 87 as VL; (30) an antibody that comprises the H chain of (28) and the L chain of (29); (31) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 109 as CDR1, the amino acid sequence of SEQ ID NO: 111 as CDR2, and the amino acid sequence of SEQ ID NO: 113 as CDR3; (32) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 115 as CDR1, the amino acid sequence of SEQ ID NO: 117 as CDR2, and the amino acid sequence of SEQ ID NO: 119 as CDR3; (33) an antibody that comprises the H chain of (31) and the L chain of (32); (34) an antibody that comprises an H chain having the amino acid sequence of SEQ ID NO: 105 as VH; (35) an antibody that comprises an L chain having the amino acid sequence of SEQ ID NO: 107 as VL; (36) an antibody that comprises the H chain of (34) and the L chain of (35); (37) an antibody that comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of any one of (1) to (36), which has equivalent activity as the antibody of any one of (1) to (36); and (38) an antibody that binds to the epitope bound by the antibody of any one of (1) to (36).

[0062] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 9 (sequence of the 5A5 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 11 (sequence of the 5A5 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 13 (sequence of the 5A5 antibody H-chain CDR3) as CDR3" of (1) is a VH having the amino acid sequence of SEQ ID NO: 5 (sequence of the 5A5 antibody VH).

[0063] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 15 (sequence of the 5A5 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 17 (sequence of the 5A5 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 19 (sequence of the 5A5 antibody L-chain CDR3) as CDR3" of (2) is a VL having the amino acid sequence of SEQ ID NO: 7 (sequence of the 5A5 antibody VL).

[0064] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 29 (sequence of the 5A9 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 31 (sequence of the 5A9 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 33 (sequence of the 5A9 antibody H-chain CDR3) as CDR3" of (7) is a VH having the amino acid sequence of SEQ ID NO: 25 (sequence of the 5A9 antibody VH).

[0065] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 35 (sequence of the 5A9 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 37 (sequence of the 5A9 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 39 (sequence of the 5A9 antibody L-chain CDR3) as CDR3" of (8) is a VL having the amino acid sequence of SEQ ID NO: 27 (sequence of the 5A9 antibody VL).

[0066] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 49 (sequence of the 4F7 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 51 (sequence of the 4F7 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 53 (sequence of the 4F7 antibody H-chain CDR3) as CDR3" of (13) is a VH having the amino acid sequence of SEQ ID NO: 45 (sequence of the 4F7 antibody VH).

[0067] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 55 (sequence of the 4F7 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 57 (sequence of the 4F7 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 59 (sequence of the 4F7 antibody L-chain CDR3) as CDR3" of (14) is a VL having the amino acid sequence of SEQ ID NO: 47 (sequence of the 4F7 antibody VL).

[0068] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 69 (sequence of the 4H5 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 71 (sequence of the 4H5 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 73 (sequence of the 4H5 antibody H-chain CDR3) as CDR3" of (19) is a VH having the amino acid sequence of SEQ ID NO: 65 (sequence of the 4H5 antibody VH).

[0069] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 75 (sequence of the 4H5 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 77 (sequence of the 4H5 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 79 (sequence of the 4H5 antibody L-chain CDR3) as CDR3" of (20) is a VL having the amino acid sequence of SEQ ID NO: 67 (sequence of the 4H5 antibody VL).

[0070] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 89 (sequence of the 6E4 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 91 (sequence of the 6E4 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 93 (sequence of the 6E4 antibody H-chain CDR3) as CDR3" of (25) is a VH having the amino acid sequence of SEQ ID NO: 85 (sequence of the 6E4 antibody VH).

[0071] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 95 (sequence of the 6E4 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 97 (sequence of the 6E4 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 99 (sequence of the 6E4 antibody L-chain CDR3) as CDR3" of (26) is a VL having the amino acid sequence of SEQ ID NO: 87 (sequence of the 6E4 antibody VL).

[0072] An example of the VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 109 (sequence of the 6H4 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 111 (sequence of the 6H4 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 113 (sequence of the 6H4 antibody H-chain CDR3) as CDR3" of (31) is a VH having the amino acid sequence of SEQ ID NO: 105 (sequence of the 6H4 antibody VH).

[0073] An example of the VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 115 (sequence of the 6H4 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 117 (sequence of the 6H4 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 119 (sequence of the 6H4 antibody L-chain CDR3) as CDR3" of (32) is a VL having the amino acid sequence of SEQ ID NO: 107 (sequence of the 6H4 antibody VL).

[0074] For the 5A5 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 1 and SEQ ID NO: 2, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 3 and SEQ ID NO: 4, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 5 and SEQ ID NO: 6, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 7 and SEQ ID NO: 8, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 9 and SEQ ID NO: 10, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 11 and SEQ ID NO: 12, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 13 and SEQ ID NO: 14, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 15 and SEQ ID NO: 16, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 17 and SEQ ID NO: 18, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 19 and SEQ ID NO: 20, respectively.

[0075] For the 5A9 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 21 and SEQ ID NO: 22, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 23 and SEQ ID NO: 24, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 25 and SEQ ID NO: 26, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 27 and SEQ ID NO: 28, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 29 and SEQ ID NO: 30, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 31 and SEQ ID NO: 32, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 33 and SEQ ID NO: 34, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 35 and SEQ ID NO: 36, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 37 and SEQ ID NO: 38, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 39 and SEQ ID NO: 40, respectively.

[0076] For the 4F7 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 41 and SEQ ID NO: 42, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 43 and SEQ ID NO: 44, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 45 and SEQ ID NO: 46, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 47 and SEQ ID NO: 48, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 49 and SEQ ID NO: 50, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 51 and SEQ ID NO: 52, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 53 and SEQ ID NO: 54, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 55 and SEQ ID NO: 56, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 57 and SEQ ID NO: 58, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 59 and SEQ ID NO: 60, respectively.

[0077] For the 4H5 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 61 and SEQ ID NO: 62, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 63 and SEQ ID NO: 64, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 65 and SEQ ID NO: 66, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 67 and SEQ ID NO: 68, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 69 and SEQ ID NO: 70, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 71 and SEQ ID NO: 72, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 73 and SEQ ID NO: 74, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 75 and SEQ ID NO: 76, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 77 and SEQ ID NO: 78, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 79 and SEQ ID NO: 80, respectively.

[0078] For the 6E4 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 81 and SEQ ID NO: 82, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 83 and SEQ ID NO: 84, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 85 and SEQ ID NO: 86, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 87 and SEQ ID NO: 88, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 89 and SEQ ID NO: 90, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 91 and SEQ ID NO: 92, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 93 and SEQ ID NO: 94, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 95 and SEQ ID NO: 96, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 97 and SEQ ID NO: 98, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 99 and SEQ ID NO: 100, respectively.

[0079] For the 6H4 antibody of the present invention, the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 101 and SEQ ID NO: 102, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 103 and SEQ ID NO: 104, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 105 and SEQ ID NO: 106, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 107 and SEQ ID NO: 108, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 109 and SEQ ID NO: 110, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 111 and SEQ ID NO: 112, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR3 are shown in SEQ ID NO: 113 and SEQ ID NO: 114, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR1 are shown in SEQ ID NO: 115 and SEQ ID NO: 116, respectively; the amino acid sequence and the nucleotide sequence of the L-chain CDR2 are shown in SEQ ID NO: 117 and SEQ ID NO: 118, respectively; and the amino acid sequence and the nucleotide sequence of the L-chain CDR3 are shown in SEQ ID NO: 119 and SEQ ID NO: 120, respectively.

[0080] The above-mentioned antibodies of (1) to (38) include not only monovalent antibodies but also multivalent antibodies with two or more valencies. The multivalent antibodies of the present invention include multivalent antibodies whose antigen binding sites are all the same and multivalent antibodies whose antigen binding sites are partially or completely different.

[0081] In a preferred embodiment, the above-mentioned antibody of (37) is an antibody with no modified CDRs. For example, the "antibody that comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of (1), which has equivalent activity as the antibody of (1)" of the above-mentioned antibody of (37) is preferably "an antibody that has equivalent activity as the antibody of (1), and comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of (1), and comprises an H chain having the amino acid sequence of SEQ ID NO: 9 as CDR1, the amino acid sequence of SEQ ID NO: 11 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3". Another preferred antibody of the above-mentioned antibody of (37) can be expressed in a similar manner.

[0082] Herein, "equivalent activity" means that the antibody of interest has biological or biochemical activity similar to that of an antibody of the present invention. Examples of the "activity" of the present invention include the activity to bind specifically to A.beta. oligomers but not to A.beta. monomers, anti-neurotoxic activity, activity to suppress A.beta. amyloid fibril formation, anti-synaptic toxicity activity, and anti-memory impairment activity.

[0083] Methods for preparing a polypeptide having activity equivalent to that of a certain polypeptide that are well known to those skilled in the art include methods for introducing mutations into a polypeptide. For example, one skilled in the art can prepare an antibody having activity equivalent to that of an antibody of the present invention by introducing appropriate mutations into the antibody using site-directed mutagenesis (Hashimoto-Gotoh, T. et al. (1995) Gene 152, 271-275; Zoller, M J, and Smith, M. (1983) Methods Enzymol. 100, 468-500; Kramer, W. et al. (1984) Nucleic Acids Res. 12, 9441-9456; Kramer W, and Fritz H J (1987) Methods. Enzymol. 154, 350-367; Kunkel, TA (1985) Proc. Natl. Acad. Sci. USA. 82, 488-492; Kunkel (1988) Methods Enzymol. 85, 2763-2766) and such. Amino acid mutations may also occur naturally. The antibodies of the present invention also include an antibody that comprises an amino acid sequence with one or more amino acid mutations in the amino acid sequence of an antibody of the present invention, and which has activity equivalent to that of the antibody of the present invention. The number of mutated amino acids in such mutants may be generally 50 amino acids or less, preferably 30 amino acids or less, and more preferably ten amino acids or less (for example, five amino acids or less).

[0084] Amino acid residues are preferably mutated into other amino acids that conserve the properties of the amino acid side chains. For example, amino acids are categorized as follows depending on the side chain properties: hydrophobic amino acids (A, I, L, M, F, P, W, Y, and V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, and T), amino acids with aliphatic side chains (G, A, V, L, I, and P), amino acids with hydroxyl-containing side chains (S, T, and Y), amino acids with sulfur atom-containing side chains (C and M), amino acids with carboxylic acid- and amide-containing side chains (D, N, E, and Q), amino acids with basic side chains (R, K, and H), and amino acids with aromatic ring-containing side chains (H, F, Y, and W) (amino acids are represented by one-letter codes in parentheses).

[0085] A polypeptide having an amino acid sequence, in which one or more amino acid residues are modified (deleted, added, and/or substituted with other amino acids) in a certain amino acid sequence, is known to retain its original biological activity (function).

[0086] In addition to the above-mentioned modifications, the antibodies of the present invention may be conjugated to other substances as long as the activity is maintained. Examples of the substances include peptides, lipids, sugars and sugar chains, acetyl groups, and natural and synthetic polymers. These modifications may be performed to confer additional functions to the antibodies, or to stabilize the antibodies.

[0087] Antibodies in which several amino acid residues have been added to the amino acid sequence of an antibody of the present invention include fusion proteins containing the antibody. In the fusion proteins, the antibody is fused with another peptide or protein. Methods for producing a fusion protein can be carried out by ligating a polynucleotide encoding an antibody of the present invention in frame with a polynucleotide encoding another peptide or polypeptide, and inserting this into an expression vector, and expressing the fusion construct in a host. Techniques known to those skilled in the art can be used for this purpose. The peptides or polypeptides fused with an antibody of the present invention include, for example, known peptides such as FLAG (Hopp, T. P. et al., BioTechnology (1988) 6, 1204-1210), 6.times.His consisting of six histidine (His) residues, 10.times.His, Influenza hemagglutinin (HA), human c-myc fragments, VSV-GP fragments, p18HIV fragments, T7-tag, HSV-tag, E-tag, SV40T antigen fragments, lck tag, .alpha.-tubulin fragments, B-tag, and Protein C fragments; glutathione-S-transferase (GST); immunoglobulin constant regions; .beta.-galactosidase; and maltose-binding protein (MBP), etc. Commercially available polynucleotides encoding these peptides or polypeptides can be fused with polynucleotides encoding the antibodies of the present invention, and the fusion polypeptides can be produced by expressing the fusion polynucleotides thus prepared.

[0088] The antibodies of the present invention may differ in the amino acid sequence, molecular weight, presence or absence of sugar chains, structure and such, depending on the cell or host producing the antibodies or the purification method. However, as long as the obtained antibody has an activity equivalent to an antibody of the present invention, it is included in the present invention.

[0089] Antibodies that bind to an epitope to which an antibody of any one of (1) to (36) above binds can be obtained by methods known to those skilled in the art. For example, the antibodies can be obtained by (i) determining the epitope bound by the antibody of any one of (1) to (36) using a conventional method, and producing the antibodies using a polypeptide comprising an amino acid sequence included in the epitope as an immunogen; or (ii) determining the epitopes of antibodies produced by a conventional method, and selecting antibodies whose epitope is the same as that of the antibody of any one of (1) to (36).

[0090] The above-mentioned antibodies of (1) to (38) also include any type of antibodies such as the above-described minibodies, antibodies with modified amino acid sequences such as humanized antibodies and chimeric antibodies, non-human animal antibodies, human antibodies, modified antibodies conjugated to other molecules (for example, polymers such as polyethylene glycol), and sugar chain-modified antibodies.

[0091] In a preferred embodiment, the antibodies of the present invention are modified antibodies such as chimeric antibodies and humanized antibodies. Examples of preferred antibodies include (i) a chimeric antibody whose variable region is derived from the 2C3 antibody, 1A9 antibody, 5A5 antibody, 5A9 antibody, 4F7 antibody, 4H5 antibody, 6E4 antibody, or 6H4 antibody, and whose constant region is derived from a human immunoglobulin; and (ii) a humanized antibody whose CDR is derived from the 2C3 antibody, 1A9 antibody, 5A5 antibody, 5A9 antibody, 4F7 antibody, 4H5 antibody, 6E4 antibody, or 6H4 antibody, and whose FR is derived from a human immunoglobulin, and whose constant region is derived from a human immunoglobulin. These modified antibodies can be produced using known methods.

[0092] Since the antigenicity of a chimeric antibody or a humanized antibody in the human body is reduced, such an antibody is useful for administration to humans for therapeutic purposes or such.

[0093] Chimeric antibodies are produced by combining sequences derived from different animals. Examples of chimeric antibodies include antibodies comprising the heavy-chain and light-chain variable regions of a mouse antibody and the heavy-chain and light-chain constant regions of a human antibody. The production of chimeric antibodies can be carried out using known methods (see, for example, Jones et al. Nature 321:522-5, 1986; Riechmann et al., Nature 332:323-7, 1988; and Presta, Curr. Opin. Struct. Biol. 2:593-6, 1992). For example, first, genes encoding the variable regions or CDRs of the antibody of interest are prepared from the RNAs of antibody-producing cells by polymerase chain reaction (PCR) or such (see, for example, Larrick et al., "Methods: a Companion to Methods in Enzymology", Vol. 2: 106, 1991; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies" in Monoclonal Antibodies: Production, Engineering and Clinical Application; Ritter et al. (eds.), page 166, Cambridge University Press, 1995, and Ward et al., "Genetic Manipulation and Expression of Antibodies" in Monoclonal Antibodies: Principles and Applications; and Birch et al. (eds.), page 137, Wiley-Liss, Inc., 1995). The prepared genes encoding the variable regions are linked to genes encoding the constant regions or framework regions. The genes encoding the constant regions or framework regions may be determined in a manner similar to that for the CDR-encoding genes, or alternatively, they can be prepared based on the sequence information of known antibodies. DNA sequences encoding chimeric products and CDR-grafted products may be synthesized completely or partially using oligonucleotide synthesis techniques. For example, the oligonucleotide synthesis described by Jones et al. (Nature 321:522-5, 1986) may be performed. Furthermore, in some cases, site-directed mutagenesis and polymerase chain reaction techniques may be appropriately used. Techniques for oligonucleotide-specific mutagenesis of known variable regions described by Verhoeyen et al. (Science 239: 1534-6, 1988) and Riechmann et al. (Nature 332: 323-7, 1988) may be used for modifying the variable region sequences, for example, to enhance the binding ability of chimeric antibodies. Furthermore, if necessary, enzymatic fill-in of gapped oligonucleotides using T4 DNA polymerase may be performed, for example, as described by Queen et al. (Proc. Natl. Acad. Sci. USA 86: 10029-33, 1989; and WO 90/07861).

[0094] For example, CDR-grafting techniques are known in the art ("Immunoglobulin genes", Academic Press (London), pp 260-74, 1989; and Michael A et al., Proc. Natl. Acad. Sci. USA 91: 969-73, 1994). Using the techniques, the CDRs of a certain antibody are replaced with the CDRs of another antibody. Through such replacement, the binding specificity of the former antibody is changed to that of the latter antibody. Among such chimeric antibodies, those in which the framework amino acids are derived from a human antibody are called "humanized antibodies (CDR-grafted antibodies)". When using antibodies to treat humans, human antibodies or humanized antibodies are preferably utilized.

[0095] Generally, chimeric antibodies comprise the variable regions of a non-human mammal-derived antibody and the constant regions derived from a human antibody. On the other hand, humanized antibodies comprise the complementarity-determining regions of a non-human mammal-derived antibody and the framework regions and constant regions derived from a human antibody.

[0096] After producing the chimeric antibodies or humanized antibodies, amino acids in the variable regions (for example, FRs) or the constant regions may be substituted with other amino acids.

[0097] The origin of the variable regions of the chimeric antibodies or the CDRs of the humanized antibodies is not particularly limited.

[0098] Human antibody-derived C-regions are used for the C-regions of the chimeric antibodies and humanized antibodies. For example, C.gamma.1, C.gamma.2, C.gamma.3, C.gamma.4, C.mu., C.delta., C.alpha.1, C.alpha.2, and C.epsilon. can be used for the H-chain C-regions, and C.kappa. and C.lamda. can be used for the L-chain C-regions. Their sequences are known. Furthermore, the human antibody C regions can be modified to improve the stability of the antibodies or their production.

[0099] The binding activity of the antibodies of the present invention to the antigens (A.beta. oligomers) can be measured using, for example, an absorbance measurement method, an enzyme-linked immunosorbent assay (ELISA) method, an enzyme immunoassay (EIA) method, a radioimmunoassay (RIA) method, and/or a fluoroimmunoassay method. In ELISA, an antibody is immobilized on a plate, and an antigen for the antibody is added to the plate, then a sample containing the desired antibody, such as the culture supernatant of antibody-producing cells or a purified antibody is added. Next, a secondary antibody which recognizes the primary antibody and is tagged with an enzyme such as alkaline phosphatase is added to the plate, and this is preincubated. After washing, an enzyme substrate such as p-nitrophenyl phosphate is added to the plate, and the absorbance is measured to evaluate the antigen-binding ability of the sample of interest. The evaluation can be performed using BIAcore (Pharmacia).

[0100] Furthermore, the present invention provides compositions comprising the above-mentioned antibody of the present invention and a pharmaceutically acceptable carrier.

[0101] As described below, the present invention strongly suggests that monoclonal 1A9 and 2C3 antibody are promising candidates for therapeutic antibodies for preventing Alzheimer-like phenotypes. Memory deterioration has been shown to be related to synaptic dysfunction caused by soluble A.beta. oligomers (Klein W L, 2001, Trends Neurosci; and Selkoe D J, 2002, Science). Excessive accumulation and deposition of A.beta. oligomers may trigger the complicated downstream cascades that cause Alzheimer's disease. If this is the case, therapeutic intervention using a composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier could be effective for blocking the pathologic cascades, and thus this could enable the treatment of Alzheimer's disease.

[0102] The "treatment" of the present invention does not necessarily have complete therapeutic or preventive effects against organs or tissues exhibiting symptoms of disorders or diseases, but may have partial effects.

[0103] "Treatment of Alzheimer's disease" in the present invention means amelioration of at least one symptom that may be caused by Alzheimer's disease, and examples include amelioration or suppression of cognitive impairment, amelioration or suppression of senile plaque formation, amelioration or suppression of synaptic dysfunction, and reduction or suppression of A.beta. accumulation in brain tissues, blood, or such. Herein, "cognitive impairment" includes, for example, memory impairment including long term/short term memory impairment, object recognition memory impairment, spatial memory impairment, and associative and emotional memory impairment.

[0104] The present invention provides pharmaceutical compositions or pharmaceutical agents which comprise as an active ingredient the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier.

[0105] In the present invention, the phrase "comprising as an active ingredient the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier" means comprising the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier as a major ingredient, but does not limit its content rate.

[0106] Examples of the above-mentioned pharmaceutical compositions include agents against cognitive impairment, Alzheimer's disease agents, agents for suppressing the progression of Alzheimer's disease, agents for suppressing senile plaque formation, agents for suppressing A.beta. accumulation, anti-neurotoxic agents (agents for neutralizing neurotoxicity), agents for inhibiting A.beta. amyloid fibril formation, and anti-synaptic toxicity agents (agents for neutralizing synaptic toxicity).

[0107] The above-mentioned pharmaceutical composition of the present invention can be expressed, for example, as "methods for suppressing Alzheimer's disease" which comprise the step of administering to a subject (individual) the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier. In other embodiments, examples include methods for suppressing cognitive impairment, methods for suppressing the progression of Alzheimer's disease, methods for suppressing senile plaque formation, methods for suppressing A.beta. accumulation, methods for neutralizing (suppressing) neurotoxic activity, methods for inhibiting A.beta. amyloid fibril formation, and methods for neutralizing (suppressing) synaptic toxicity. In further embodiments, examples include methods for preventing and/or treating cognitive impairment, and methods for preventing and/or treating Alzheimer's disease.

[0108] The present invention also provides use of a composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier in the production of the above-mentioned pharmaceutical composition.

[0109] Furthermore, the present invention relates to the following compositions.

[0110] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in preventing and/or treating cognitive impairment.

[0111] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in preventing and/or treating Alzheimer's disease.

[0112] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in suppressing the progression of Alzheimer's disease.

[0113] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in suppressing senile plaque formation.

[0114] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in suppressing A.beta. accumulation.

[0115] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in neutralizing (suppressing) neurotoxic activity.

[0116] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in inhibiting A.beta. amyloid fibril formation.

[0117] A composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier for use in neutralizing (suppressing) synaptic toxicity.

[0118] The above-mentioned pharmaceutical agents of the present invention can be administered to humans or other animals. In the present invention, non-human animals to which the pharmaceutical agents are administered include mice, rats, guinea pigs, rabbits, chickens, cats, dogs, sheep, pigs, cattle, monkeys, baboons, and chimpanzees. These animals preferably exhibit at least one symptom selected from, for example, cognitive impairment, senile plaque formation, synaptic dysfunction, A.beta. accumulation in brain tissues or blood, etc.

[0119] Antibodies contained in the pharmaceutical compositions of the present invention are not particularly limited as long as they are included in the above-mentioned antibodies of the present invention, and examples include the antibodies described herein.

[0120] When using the above-mentioned antibodies of the present invention for pharmaceutical compositions, they may be formulated by methods known to those skilled in the art. For example, as necessary, they can be prepared in the form of injectable sterile solutions or suspensions using water or another pharmaceutically acceptable liquid, and can be administered parenterally. For example, the antibodies to be included in the pharmaceutical compositions can be combined with acceptable carriers or media, specifically, sterile water, physiological saline, vegetable oils, emulsifiers, suspensions, surfactants, stabilizers, flavoring agents, excipients, solvents, preservatives, binders, or such, and mixed into a unit dose form required for generally accepted pharmaceutical practice. The phrase "pharmaceutically acceptable" indicates that the substance is inactive, and contains conventional substances used as diluents or vehicles for pharmaceuticals. Suitable excipients and their formulations are described, for example, in Remington's Pharmaceutical Sciences, 16.sup.th ed. (1980) Mack Publishing Co., ed. Oslo et al.

[0121] Physiological saline and other isotonic solutions containing glucose or adjuvants (for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride) can be used as aqueous solutions for injection. They can be used together with appropriate solubilizers such as alcohols, more specifically, ethanol and polyalcohols (propylene glycol, polyethylene glycol, and such), and non-ionic surfactants (Polysorbate 80.TM., HCO-50, and such).

[0122] Sesame oil or soybean oil can be used as an oleaginous liquid, and benzyl benzoate or benzyl alcohol can be used in combination as a solubilizer. Buffers (for example, phosphate buffer and sodium acetate buffer), soothing agents (for example, procaine hydrochloride), stabilizers (for example, benzyl alcohol and phenol), and antioxidants can be used for the formulations. Prepared injection solutions can be filled into appropriate ampules.

[0123] The administration is preferably parenteral administration, and specific examples include administration by injection, transnasal administration, transpulmonary administration, and transdermal administration. Examples of administration by injection include systemic and local administration by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, and such.

[0124] The pharmaceutical compositions contain a pharmaceutically effective amount of the active component (the above-mentioned antibody of the present invention). "Pharmaceutically effective amount (of a compound)" refers to an amount sufficient for treating and/or preventing disorders in which the antigens for the above-mentioned antibodies of the present invention play an important role. For example, "a pharmaceutically acceptable amount" may be an amount required for reducing A.beta. accumulation, neutralizing A.beta.-induced toxicity, reducing A.beta. fibril formation, or such, thereby treating or preventing conditions caused by Alzheimer's disease, when the compound is administered to individuals (patients). The reduction or neutralization may be, for example, a reduction or neutralization of at least approximately 5%, 10%, 20%, 30%, 40%, 50%, 75%, 80%, 90%, 95%, 99%, or 100%.

[0125] Assessment for determining such a pharmaceutically effective amount of the above-mentioned antibodies of the present invention may be carried out using a standard clinical protocol including histopathological diagnosis.

[0126] A suitable administration method may be selected depending on the age and symptoms of the patient. The dosage of an antibody-containing pharmaceutical composition may be selected, for example, within the range of 0.0001 mg to 1000 mg per kilogram body weight for each administration. Alternatively, for example, the dosage for each patient may be selected within the range of 0.001 to 100,000 mg/body; however, the dosage is not necessarily limited to these ranges. Although the dosage and administration methods vary depending on the patient's body weight, age, symptoms, and such, one skilled in the art can appropriately select them. In the later-described animal experiments, the dosage was selected based on the high-dose intravenous immunoglobulin therapy (400 mg/kg) covered by health insurance for humans.

[0127] Furthermore, the present invention provides methods for detecting A.beta. oligomers (examples include A.beta. 40 (A.beta. 1-40) and A.beta. 42 (A.beta. 1-42) oligomers) in samples. Examples of "samples" of the present invention include samples collected from subjects. Specifically, the present methods include the step of detecting A.beta. oligomers contained in a sample collected from a subject using an antibody of the present invention. A.beta. oligomers in a sample can be detected using, for example, sandwich solid-phase enzyme immunoassay methods that use chemiluminescence (chemiluminescence ELISA), immunoprecipitation methods that use the obtained antibodies, immunoblotting, flow cytometry, mass spectrometry, and immunohistochemical analysis.

[0128] When A.beta. oligomers are detected in a sample collected from a subject by the above-mentioned measurement methods, the subject may be an Alzheimer's disease patient. For example, when the amount of A.beta. oligomers in a sample collected from a subject is compared with that from a healthy individual, and if the amount of A.beta. oligomers is greater in the subject than in the healthy individual, the subject is determined to suffer from or be at a risk of developing Alzheimer's disease. Whether or not a subject suffers from or is at a risk of developing Alzheimer's disease is diagnosed usually by physicians (including individuals under instructions from physicians; same herein below). Data on the amount of A.beta. oligomers in samples collected from a subject and a healthy individual, which are obtained by the present methods of diagnosis, will be useful for diagnosis by physicians. Therefore, the present methods of diagnosis can be expressed as methods of collecting and presenting data useful for diagnosis by physicians.

[0129] Specifically, the present invention provides methods for diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, wherein the methods comprise detecting A.beta. oligomers in a sample collected from the subject using an antibody of the present invention.

[0130] Furthermore, the present invention provides methods of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease, which comprise the steps of:

(a) contacting a sample collected from a subject with an antibody of the present invention and an antibody that binds to an A.beta. monomer; and (b) measuring the ratio of A.beta. oligomer to A.beta. monomer in the sample, wherein the subject is determined to suffer from or be at a risk of developing Alzheimer's disease, if the ratio measured in step (b) is higher than that of a healthy individual.

[0131] First, in the present methods, a sample collected from a subject is contacted with an antibody of the present invention and an antibody that binds to an A.beta. monomer. Herein, "contact" may be carried out, for example, by adding each of the above-mentioned antibodies to a sample collected from a subject, which is placed in a test tube. In this case, the antibody is added suitably in the form of a solution, a solid obtained by freeze-drying, or such. When adding the antibody as an aqueous solution, the solution may purely contain the antibody alone, or may contain, for example, surfactants, excipients, coloring agents, flavors, preservatives, stabilizers, buffers, suspending agents, tonicity agents, binding agents, disintegrants, lubricants, fluidity promoters, or corrigents. The concentration at which the antibody is added is not particularly limited. For example, as with human immunoglobulin formulations, 500-mg, 1000-mg, and 2500-mg freeze-dried formulations and such may be suitably used.

[0132] Next, the ratio of A.beta. oligomer to A.beta. monomer (herein, this is also referred to as "O/M index") in the aforementioned sample is measured. To measure this ratio, the following method is suitably used. For example, as described below in the Examples, the measurement can be carried out using a method of comparing the oligomer and monomer ELISA values obtained from the same sample.

[0133] Then, this ratio is compared with the ratio for a healthy individual. When the ratio is higher in the subject than in the healthy individual, the subject is determined to suffer from or be at a risk of developing Alzheimer's disease.

[0134] The methods of diagnosis of the present invention can be performed both in vitro and in vivo, but they are preferably performed in vitro.

[0135] Preferably, the "sample" of the present invention is not particularly limited as long as it is a tissue derived from a subject. Examples include the brain (brain parenchyma, and such), organs, and body fluids (blood, cerebrospinal fluid, and such) of a subject. In the present invention, the sample is preferably blood (more preferably, plasma) or cerebrospinal fluid.

[0136] Furthermore, the present invention provides pharmaceutical agents for use in the above-mentioned methods of measuring A.beta. oligomers in a sample, or methods of diagnosing whether or not a subject suffers from or is at a risk of developing Alzheimer's disease.

[0137] In the present invention, the pharmaceutical compositions comprising an antibody may be included in products and kits containing materials useful for treating pathological conditions of a subject. The products may comprise any labeled container for a compound. Suitable containers include bottles, vials, and test tubes. The containers may be formed from a variety of materials such as glass and plastic. The label on the container surface should indicate that the composition is used to treat or prevent one or more conditions of the disease. The label may also indicate descriptions for administration, and such.

[0138] In addition to the above-mentioned container, a kit containing a pharmaceutical composition comprising an antibody may optionally include a second container that stores a pharmaceutically acceptable diluent. The kit may further include other materials desirable from a commercial and user's standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with descriptions for use.

[0139] If necessary, the pharmaceutical compositions may be provided in a pack or dispenser device that may contain one or more unit dosage forms comprising an active ingredient. The pack may comprise metal or plastic foil, and, for example, it is a blister pack. The pack or dispenser device may be accompanied by instructions for administration.

[0140] In the above-mentioned pharmaceutical agents and kits, besides the antibody of the present invention that is an active ingredient, sterile water, physiological saline, vegetable oils, surfactants, lipids, solubilizing agents, buffers, protein stabilizers (BSA, gelatin, etc.), preservatives, blocking solutions, reaction solutions, reaction quenching solutions, reagents for treating samples, and such, may be mixed as necessary.

[0141] The present inventors showed that the antibodies of the present invention are effective for preventing Alzheimer's disease. That is, the present invention provides methods for suppressing the progression of Alzheimer's disease, wherein the methods comprise the step of administering to an individual affected with Alzheimer's disease, a composition comprising the above-mentioned antibody of the present invention and a pharmaceutically acceptable carrier.

[0142] The antibodies provided by the present invention are expected to greatly contribute to the establishment of preventive/therapeutic methods selective to molecules responsible for evoking pathological conditions of Alzheimer's disease, and the establishment of early diagnostic markers for Alzheimer's disease. The present inventors obtained evidence showing that, even in antibody therapy targeting pathological conditions in the brain, peripheral intravenous administration is sufficient and there is no need to consider brain transfer. Thus, the present invention is expected to rapidly accelerate the progress of antibody drugs for Alzheimer's disease.

[0143] All prior art references cited herein are incorporated by reference into this description.

EXAMPLES

[0144] Hereinbelow, the present invention is specifically described with reference to the Examples, but it is not to be construed as being limited thereto.

Methods

Preparation of Antigens (1A9 and 2C3)

[0145] Synthetic A.beta. 1-42 (Peptide Institute, Inc., Osaka) was dissolved in distilled water or 10 mM phosphate buffer, and incubated at 37.degree. C. for 18 hours. Then, the peptides were separated by SDS-PAGE (4-12% NuPAGE Tris-Glycine gel), and after visualization by CBB staining, just the A.beta. 1-42 tetramer was excised without contamination of the A.beta. 1-42 monomer.

Preparation of antigens (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4)

[0146] A fluorescent dye, 6-carboxytetramethylrhodamine (6-TAMRA) (SIGMA) was chemically linked to the N terminus of a synthetic A.beta. 1-40 peptide (Peptide Institute, Inc.) to produce a modified A.beta.. An oligomer-rich sample (A.beta. 1-40 oligomer) was prepared by copolymerizing the modified A.beta. and synthetic A.beta. 1-40 peptide. It is preferable to adjust the conditions so that the fluorescence intensity determined by ThT assay, which is described below, is one-fourth or less the fluorescence intensity in the absence of modified A.beta.. More specifically, it is preferred that 100 .mu.M each of the modified A.beta. and synthetic A.beta. 1-40 peptide are mixed, and polymerized for 20 hours.

Preparation of Antibody-Producing Hybridomas

[0147] Balb/c mice were immunized by injecting the antigen prepared by the method described above into their foot pads. Then, booster immunization was carried out six times. Hybridomas were prepared from inguinal lymph nodes by fusion with Sp2/O--Ag14 cells using Polyethylene Glycol 1500.

Antibody Isotyping

[0148] Isotyping of purified immunoglobulins was carried out using a Serotec (Oxford, UK) mouse monoclonal antibody isotyping kit.

Dot Blot Analysis (Primary Screening)

[0149] The initial screening was carried out by dot blot analysis using a nitrocellulose membrane onto which 2.5 .mu.l of A.beta. 1-42 (2.5 .mu.g/dot) pre-incubated for 18 hours was immobilized. Non-specific binding sites on the membrane were blocked with a phosphate buffer containing 5% low-fat milk, 1% BSA, and 0.05% Tween-20, and then the membrane was incubated with a culture supernatant. A.beta. oligomer-binding antibodies in the culture supernatant were detected by horseradish peroxidase-labeled goat anti-mouse F(ab').sub.2 (1:3000; Amersham), and visualized using an enhanced chemiluminescence (ECL) kit and LAS3000 mini (Fujitsu, Tokyo, Japan). Among 400 clones, 16 clones positive in the dot blotting, including 1A9 and 2C3, were subjected to the secondary screening described below.

Immunoprecipitation and Immunoblot Analysis (Secondary Screening)

[0150] Immunoprecipitation experiments (Ghiso J, et al., Biochem J, 1993) were conducted using an A.beta. oligomer-rich amyloid fraction (Matsubara E, et al., Neurobiol Aging, 2004) for the secondary screening to assess whether the 16 clones selected in the primary screening can capture A.beta. oligomers in AD brain. A buffer-insoluble, formic acid-soluble fraction prepared from AD brain was incubated with a culture supernatant and Protein G-Sepharose. The immunoprecipitated A.beta. oligomers were separated using an NuPAGE 4-12% Bis-Tris-Glycine gel, and transferred onto a nitrocellulose membrane or Immobilon P (Millipore) using 10 mM 3-cyclohexylamino-1-propane sulfonic acid (pH 11) containing 10% methanol at 400 mA for one hour. Non-specific binding sites on the membrane were blocked with a phosphate buffer containing 5% low-fat milk, 1% BSA, and 0.05% Tween-20 at room temperature for three hours. The immunoprecipitated A.beta. oligomers were detected by immunoblotting using the 4G8 (1:1000) or 6E10 (1:1000) monoclonal antibody as described above. Two clones, 1A9 and 2C3, were selected from the 16 clones as candidates for therapeutic antibodies for Alzheimer's disease.

Antibodies

[0151] The 6E 10 and 4G8 monoclonal antibodies (Covance Immuno-Technologies, Dedham, Mass.) recognize the epitopes at amino acid positions 1-16 and 17-24 of the human A.beta. sequence, respectively. Polyclonal A11 which specifically recognizes A.beta. oligomers was purchased from Biosource (Camarillo, Calif.). Alex Fluor(AF)488- or 594-conjugated goat anti-mouse IgG and Alex Fluor(AF)488-conjugated goat anti-rat IgG were purchased from Molecular Probes (Eugene, Oreg.). Anti-mouse IgG2b was purchased from Sigma (St. Louis, Mo.). An anti-synaptophysin antibody was purchased from Santa Cruz (Santa Cruz, Calif.), and an anti-drebrin antibody was purchased from MBL (Nagoya, Japan).

Size Exclusion Chromatography (SEC)

[0152] SEC was carried out to assess 1A9 and 2C3 for their size specificity. As previously reported (Matsubara E., et al., Neurobiol Aging, 25: 833-841, 2004), this method can selectively separate A.beta. monomers and A.beta. oligomers, or lipoprotein-bound A.beta. and lipoprotein-free A.beta.. The present inventors concentrated the culture supernatant from APP/PS1-overexpessing HEK293 cells about ten-fold using a Microcon 3 kDa molecular weight cut-off filter (Millipore Corp.). Then, this concentrate was fractionated into 28 one-milliliter fractions using a Superose 12 size exclusion column (1 cm.times.30 cm; Pharmacia Biotech., Uppsala, Sweden; flow rate of 0.5 ml/min) pre-equilibrated with a phosphate buffer. Half of each fraction was subjected to immunoprecipitation using 1A9 or 2C3. A.beta. contained in the resulting immunoprecipitates was detected by immunoblotting using 4G8.

[0153] Cerebrospinal fluid (CSF) pooled from ten cases of Alzheimer's disease patients or age-matched healthy individuals, and lipoprotein-depleted CSF from the pools were also fractionated under the same conditions as described above. A.beta. in the collected fractions was detected by ELISA analysis. To detect the lipid, the total cholesterol was enzymatically quantified using a standard kit (Wako, Osaka, Japan). Under the experimental conditions of the present inventors, the CSF lipoproteins were eluted at fractions 7 to 14, while fractions 15 to 28 contained cholesterol-free proteins.

Preparation of Seed-Free A.beta. Solution

[0154] Synthetic A.beta. 1-42 was dissolved at 250 .mu.M in 0.02% ammonia water. Then, in order to prepare a seed-free A.beta. solution, insoluble peptides, which may function as a seed, were precipitated by ultracentrifugation using an Optima TL ultracentrifuge (Beckman, USA) at 540,000.times.g for three hours. The resulting supernatant was collected, aliquoted, and stored at -80.degree. C. until use. Samples were prepared by thawing the A.beta. stock solutions immediately before use, and diluting them ten-fold with Tris-buffered saline (TBS; 150 mM NaCl and 10 mM Tris-HCl (pH 7.4)). The resulting 25 .mu.M solutions were used in the experiments described below. Synthetic A.beta. 1-40 (HCL form; Peptide Institute, Inc., Osaka) was prepared at 50 .mu.M.

A.beta. Incubation and ThT Assay (Yamamoto N, et al., J Biol Chem, 282: 2646-2655, 2007)

[0155] An A.beta. solution (25 .mu.M) was incubated in the presence of a predetermined concentration of an antibody at 37.degree. C. for two or 24 hours. The ThT fluorescence intensity of the incubation mixture was determined using a fluorescence spectrophotometer (RF-5300PC; Shimadzu Co., Kyoto, Japan). The optimal fluorescence intensity was determined for A.beta. amyloid fibrils at excitation and emission wavelengths of 446 and 490 nm, respectively, using 1.0 ml of a reaction mixture containing 5 .mu.M ThT and 50 mM glycine-NaOH (pH 8.5). The fluorescence intensity was determined immediately after preparation of the mixture.

[0156] Furthermore, the activity of the 4F7, 4H5, 5A5, 5A9, 6E4, and 6H4 antibodies to suppress A.beta. amyloid fibril formation was assessed by the following procedure. An A.beta. 1-42 solution diluted to 12.5 .mu.M with cell culture medium was incubated in the presence or absence of each antibody at 37.degree. C. for 24 hours. The amount of foamed amyloid fibrils was determined by the above-described ThT fluorescence intensity assay method.

A.beta.-Induced Neurotoxicity Assay (Yamamoto N, et al., J Biol Chem, 282: 2646-2655, 2007)

[0157] Rat pheochromocytoma PC 12 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) (Invitrogen, Carlsbad, Calif.) containing 10% heat-inactivated horse serum (Invitrogen) and 5% fetal bovine serum (FBS) (Invitrogen). In order to induce the differentiation into nerve cells, PC12 cells were plated at a density of 20,000 cells/cm.sup.2 in culture dishes coated with poly-L-lysine (10 mg/ml), and cultured for six days in DMEM supplemented with 100 ng/ml nerve growth factor (NGF; Alornone Labs, Jerusalem, Israel) (PC12N). PC12N was exposed to 25 .mu.M seed-free A.beta. 1-42 or pre-incubated A.beta. 1-42 in the presence or absence of antibody at 4.degree. C. for 48 hours. The neurotoxicity induced by A.beta. 1-42 was assessed by Live/Dead dual-color fluorescence assay according to the supplier's instructions (Molecular Probes, Eugene, Oreg.).

[0158] Furthermore, the activity of the 4F7, 4H5, 5A5, 5A9, 6E4, and 6H4 antibodies to neutralize A.beta.-induced neurotoxicity was assessed by the method described below. First, human neuroblastoma cells (SH-SY5Y) were cultured for 24 hours in DMEM containing 10% FBS, at a density of 150,000 cells/well in 24-well plates. Then, the medium was replaced with serum-free culture medium containing A.beta. 1-42 (12.5 .mu.M) in the presence or absence of antibody, and the cells were cultured for another 24 hours. To determine the cytotoxicity induced by A.beta. 1-42, the level of dead cell-derived LDH released into the medium was measured by a CytoTox96 kit (Promega).

Ultrafiltration and Molecular Sieve

[0159] In order to determine the size-dependent characteristics of neurotoxic A.beta. oligomers, the four types of filtrates (<3 kDa, 3 to 10 kDa, 10 to 30 kDa, 30 to 100 kDa) and the retention solution (>100 kDa) were prepared from a 25 .mu.M A.beta. oligomer solution by sequential ultrafiltration using Microcon 3 kDa, 10 kDa, 30 kDa, and 100 kDa cut-off membranes. Each of the fractions was subjected to the A.beta.-induced neurotoxicity assay described above. PC12N was exposed to each fraction to identify the toxic fraction as described above. The distribution of the three-dimensional structures recognized by A11, 1A9, 2C3, and 4G8 was also identified by the dot blot analysis described above. The morphological characterization of the neurotoxic oligomers was performed by examining each fraction using an atomic force microscope.

Electron Microscopy (EM) and Atomic Force Microscopy (AFM)

[0160] Samples were diluted with distilled water and sprayed over carbon-coated grids to conduct electron microscopy. The grids were negatively-stained with 1% phosphotungstic acid and observed under a Hitachi H-7000 electron microscope (Tokyo, Japan) with an acceleration voltage of 77 kV. AFM assessment was carried out as recently reported. Drops of the samples were placed onto freshly cleaved mica. The mica was allowed to stand for 30 minutes and then washed with water, and the liquid samples were analyzed using Nanoscope IIIa (Digital Instruments, Santa Barbara, Calif., USA) set to the tapping mode (Tero, R, et al., Langmuir 20, 7526-7531, 2004). The cantilever used was OMCL-TR400PSA (Olympus, Japan).

Subject Tissues and Extraction

[0161] The present study was conducted based on autopsy cases (n=50; 26 male and 24 female cases) from the Tokyo Metropolitan Brain Bank for Aging Research of the Tokyo Metropolitan Institute of Gerontology (Itabashi, Tokyo, Japan). This research project was approved by the institutional ethical committees of the Faculty of Medicine, the University of Tokyo; the Tokyo Metropolitan Geriatric Hospital of the Tokyo Metropolitan Institute of Gerontology; and the National Center of Geriatrics and Gerontology. Details of subjects and sample collection have been reported (Katsuno T, Neurology, 64: 687-692, 2005). However, that study analyzed insoluble brain fractions, whereas in this research project (Katsuno T, Neurology, 64: 687-692, 2005), the present inventors analyzed soluble brain fractions, which remain uncharacterized in previous studies. Frozen tissue samples (the anterior portion of entorhinal cortex) were homogenized in nine volumes of Tris-buffered saline (TS) containing a protease inhibitor cocktail. The homogenates were ultracentrifuged at 265,000.times.g for 20 minutes. One-third aliquots (0.5 ml) of the resulting supernatants were subjected to immunoblot analysis.

Immunohistochemistry

[0162] The left brain hemispheres of Tg2576 mice were sliced into 30-.mu.m-thick sagittal sections using a cryotome (RM 2145; Leica, Wetzlar, Germany), and stained with thioflavin S as previously described (Wyss-Coray et al., 2001). The formation of swollen dystrophic neurites was observed using an anti-synaptophysin antibody (Chemicon, Temecula, Calif.). The number of thioflavin S-positive plaques and synaptophysin-positive swollen dystrophic neurites was counted by observing four or five sections from the left brain hemisphere of each mouse at 40-fold magnification. To observe A.beta. deposition, serial sections briefly pre-treated with formic acid or Protease K were stained using an A.beta. immunostaining kit (Sigma, St. Louis, Mo.), and immuno-positive signals were visualized using an ABC elite kit (Vector Laboratories). Images of the cerebral cortex and hippocampus were recorded using a digital camera connected with a microscope, and analyzed using a simple PCI software (Compix Imaging System, Lake Oswego, Oreg.). The brain translocation of antibodies was observed using a confocal laser microscope (Carl Zeiss LSM510). The number of thioflavin S-positive plaques and synaptophysin-positive swollen dystrophic neurites was determined in a double blind manner.

Passive Immunotherapy and Behavioral Analysis

[0163] Three-month-old female non-transgenic (non-Tg) mice, and Tg2576 mice having and overexpressing the Swedish-type mutant human APP gene with dual mutations (K670N and M671L) derived from familial AD were purchased from Taconics (Germantown, N.Y., USA). These mice were reared until 13 months old in the animal facility of the present inventors. To determine whether the Alzheimer-like phenotype is prevented by passive immunotherapy, 1A9 or 2C3 (0.4 mg/kg/week), or PBS was administered into the caudal vein of four-month-old Tg2576, and the administration was continued until 13 months. The memory function was assessed at month 13 as previously described (Mouri A, FASEB J, 21: 2135-2148, 2007), based on the following four behavioral paradigms:

(1) Y-maze test for short-term memory; (2) novel object recognition test; (3) Morris water maze test; and (4) contextual fear conditioning test. Three days after the behavioral tests, the mice were sacrificed for biochemical and histological assessments. The experimental results were analyzed by one-way ANOVA and two-way ANOVA. Post-hoc analysis was carried out using Fisher test.

Separation and Removal of Lipoprotein

[0164] CSF was collected from 12 AD patients and 13 NC individuals. Then, lipoproteins were removed from 600 .mu.l each of the CSF by preparative continuous density gradient ultracentrifugation according to a protocol reported previously (Matsubara E, et al., Ann Neurol, 45: 537-541, 1999). The density of CSF was adjusted to 1.25 g/ml with KBr. The CSF was ultracentrifuged at 100,000 rpm and 16.degree. C. for eight hours using a Hitachi RP100AT centrifuge. Lipoproteins floating at a density of 1.25 g/ml and lipoprotein-depleted CSF (LPD-CSF) were subjected to ultrafiltration using a 3 kDa cut-off membrane (Microcon 3; Arnicon, Inc), and then frozen and stored, or stored at 4.degree. C., until use.

[0165] Lipoproteins were also removed by affinity chromatography using PHML-LIPOSORB (Calbiochem, La Jolla, Calif.). Each sample (plasma or brain) and PHML-LIPOSORB (Calbiochem, La Jolla, Calif.) were combined at a ratio of 1.5:1, and mixed for 60 seconds. Then, the mixture was centrifuged at 3,000 rpm for ten minutes. The resulting supernatants (lipoprotein-free samples) were subjected to ELISA using 6E10 for the oligomers. The lipoprotein-bound samples were eluted from PHML-LIPOSORB using 20 mM sodium deoxycholate. The removal of specific lipoproteins was confirmed by agarose electrophoresis using 1% gel (Beckmann), followed by staining with FAST-RED 7B (Wako, Osaka, Japan).

Quantification of Human A.beta.

[0166] Whole plasma and LPDP A.beta. species were specifically quantified by sandwich ELISA as previously described (Matsubara E, et al., Ann Neurol, 537-541, 1999; Matsubara E, et al., Neurobiol Aging, 25: 833-841, 2004). To analyze brain A.beta. species, soluble A.beta. species in 100 .mu.l of buffer were directly subjected to ELISA, while insoluble A.beta. samples extracted with 70% formic acid were neutralized with 1 M Tris-HCl (pH 8.0) and diluted 1.000-fold prior to ELISA. The values obtained by the assay were normalized using the brain wet weight, and ultimately presented in pmol/g. Normalization among plates was done by including the three standard plasma samples in all three plates.

A.beta. Oligomer-Specific ELISA

[0167] Chemiluminescence-based sandwich solid-phase enzyme immunoassay (chemiluminescent ELISA) was used to specifically detect oligomeric A.beta. but not monomeric A.beta.. Microplates were coated with monoclonal 1A9 (IgG2b isotype) or 2C3 (IgG2b isotype), or a mixture of 1A9 and 2C3. 100 .mu.l of a sample (brain or cerebrospinal fluid) was added and incubated continuously for 24 hours at 4.degree. C. Then, horseradish peroxidase-conjugated BA27 Fab' fragment (anti-A.beta. 1-40 specific to A.beta. 40; Wako pure chemical, Osaka, Japan) or horseradish peroxidase-conjugated BCOS Fab' fragment (anti-A.beta. 35-43 specific to A.beta. 42; Wako pure chemical, Osaka, Japan) was added and incubated at 4.degree. C. for 24 hours. The chemiluminescence generated using SuperSignal ELISA Pico Chemiluminescent Substrate (Pierce, Rockford, Ill., USA) was quantified by a Veritas Microplate Luminometer (Promega).

[0168] To assess the in vivo efficacy of the peripheral administration of monoclonal 1A9 and 2C3, plasma and organ samples were collected from administered mice and analyzed for A.beta. oligomers by ELISA using HRP-labeled 6E10 (Senetek PLC, Napa, Calif., USA) specific to the human oligomers. High-sensitivity detection was achieved by using the above-described chemiluminescent system. To avoid interference by lipoprotein-bound A.beta. monomers, the present inventors pre-treated plasma and organ samples using PHML-LIPOSORB in the same way as described above. The resulting lipoprotein-depleted samples were used for the assay.

Inhibition ELISA

[0169] A.beta. oligomers used in this assay were prepared by diluting synthetic A.beta. 1-40 (HCl form) to a concentration of 0.1 mg/ml with PBS and incubating this at 37.degree. C. for one hour. Meanwhile, A.beta. monomers were prepared by diluting synthetic A.beta. 1-40 (TFA form) to a concentration of 0.1 mg/ml with PBS. A.beta. oligomers were immobilized onto 96-well immunoplates at 400 ng/well, and then the plates were blocked with BSA. Next, the A.beta. monomers or oligomers stepwise-diluted in the range of 100 pg/ml to 100 .mu.g/ml were reacted with the 4F7, 4H5, 5A5, 5A9, 6E4, or 6H4 antibodies, or the control anti-A.beta. antibodies (4G8 and 6E10). After incubation for two hours, the mixtures were added to the above-described 96-well immunoplates, and incubated at room temperature for ten minutes. The binding of immobilized A.beta. oligomers to each of the antibodies was detected by measuring the absorbance at 450 nm in the color development reaction using an HRP-labeled anti-mouse IgG antibody and a TMB solution.

Example 1

Preparation of A.beta. Oligomer-Specific Monoclonal Antibodies (1A9 and 2C3)

[0170] A.beta. oligomers and monomers co-exist in a solution. Thus, it is essential to remove A.beta. monomers for preparation of antigens to produce A.beta. oligomer-specific antibodies. As shown in FIG. 1A, the present inventors succeeded in isolating SDS-stable A.beta. tetramers without contamination of A.beta. monomers by SDS-PAGE. After in vivo immunization with the isolated A.beta. tetramers, positive hybridoma clones were selected by two-step screening using dot blot analysis followed by immunoprecipitation. Among 400 clones subjected to dot blot analysis, 16 clones were determined to be positive (positivity rate=4%). To assess the specificity of the isolated positive clones to the oligomers, a phosphate buffer-insoluble and formic acid (FA)-soluble amyloid fraction derived from AD brain (Matsubara E et al., Neurobiol Aging, 25: 833-841, 2004) was analyzed by immunoprecipitation using the cell culture supernatants of the positive hybridomas (FIG. 1B). The A.beta. dimer, a smaller amount of the trimer, and a high-molecular-weight smear characteristic to aggregated A.beta. molecular species were detected by immunoblot analysis using anti-A.beta. monoclonal 4G8. A very small amount of A.beta. monomers dissociated in the presence of SDS was also detected. To further confirm the existence of three-dimensional structures recognized by native 1 A9 and 2C3 (i.e., oligomers), the present inventors detected the oligomers in conditioned medium (CM) of human embryonic kidney (HEK) 293 cells transfected with mutant PS1 cDNA (Nakaya Y et al., J Biol Chem, 280: 19070-19077, 2005). The present inventors fractionated HEK293 CM by SEC, and then identified the oligomers. As reported previously (Matsubara E et al., Neurobiol Aging, 25: 833-841, 2004; Yamamoto N, et al., J Biol Chem, 282: 2646-2655, 2007), this method can effectively separate the oligomers (fractions 8 to 13) from monomers (fractions 14 to 20). When immunoprecipitated with monoclonal 1A9, SDS-stable A.beta. dimers secreted into CM were precipitated in fraction 8 (>680 kDa); SDS-stable A.beta. dimers and trimers were precipitated in fraction 13 (17 to 44 kDa); and a very small amount of the dimers was precipitated in fraction 16 (FIG. 1C). Similar results were obtained when immunoprecipitation was carried out using 2C3 (data not shown). These data demonstrate that monoclonal 1A9 and 2C3 are exactly specific to An oligomers but do not recognize An monomers.

Example 2

The anti-Neurotoxic Activity of Monoclonal 1 A9 and 2C3

[0171] To assess whether monoclonal 1A9 and 2C3 can prevent A.beta.-induced neurotoxicity, NGF-differentiated PC12 cells (PC12N) were incubated with 25 .mu.M seed-free A.beta. 1-42 (ThT-negative 540,000.times.g supernatant) in the presence or absence of the monoclonal antibodies (mAbs) at 37.degree. C. for 48 hours. The viability of nerve cells was determined by LIVE/DEAD assay (FIG. 2). Nerve cell death was detected at a significantly high level (50%) in the presence of A.beta. 1-42 (FIGS. 2B and 2G), as compared to the control assay (FIG. 2A). Non-specific IgG2b (FIGS. 2C and 2G) could not inhibit the A.beta. 1-42-induced neurotoxicity under the same conditions. The commercially available A.beta.-specific monoclonal antibody 4G8 (IgG2b isotype; FIGS. 2D and 2G) had a tendency to enhance the toxicity. Monoclonal 2C3 (IgG2b isotype; FIGS. 2F and 2G) neutralized the neurotoxicity of A.beta. 1-42 almost completely in a concentration-dependent manner. Thus, the de novo-formed neurotoxic three-dimensional structure recognized by 2C3 was speculated to take an oligomer form. Meanwhile, the anti-neurotoxic activity of 1A9 (IgG2b isotype; FIGS. 2E and 2G) falls between the anti-neurotoxic activity of 2C3 and non-specific IgG2b. This suggests that the three-dimensional structure recognized by 1A9 is structurally different from the 2C3-recognized oligomers.

Example 3

[0172] Currently, the determination of the precise size and conformation of neurotoxic A.beta. 1-42 oligomers is one of the most urgent issues and which is subjected to intense competition. The present inventors succeeded in isolating soluble neurotoxic A.beta. 1-42 molecular species and fractionating the species into the following five fractions by ultrafiltration and molecular sieving (UC/MS) (FIG. 3A):

fraction 1, filtrate of <3 kDa (lane 1); fraction 2, filtrate of 3 to 10 kDa (lane 2); fraction 3, filtrate of 10 to 30 kDa (lane 3); fraction 4, filtrate of 30 to 100 kDa (lane 4); and fraction 5, retention solution of >100 kDa (lane 5).

[0173] The immunoblot analysis using monoclonal 4G8 (FIG. 3A) revealed that:

fraction 1 does not contain A.beta. (lane 1); fraction 2 contains A.beta. monomers (lane 2); fraction 3 contains A.beta. monomers and a small amount of A.beta. dimers (lane 3); fraction 4 contains A.beta. monomers to pentamers (lane 4); and fraction 5 contains A.beta. monomers to pentamers, and molecules of 45 to 160 kDa (lane 5).

[0174] These data suggest that 2% SDS depolymerizes high-molecular-weight (HMW) A.beta. oligomers into A.beta. monomers and low-molecular-weight (LMW) A.beta. oligomers. To assess the size distribution of toxic A.beta. 1-42, the present inventors measured the biological activity of each fraction incubated with PC12N at 37.degree. C. for 48 hours. As shown in FIGS. 3B and 3C, it was demonstrated that: fraction 1 was non-toxic, and fraction 2 had a very weak toxicity, suggesting that A.beta. monomers and dimers are unlikely to be toxic. Fractions 3 to 5 were significantly toxic (one-way ANOVA; p<0.0001), suggesting that the size of neurotoxic oligomers theoretically corresponds to the size of trimers or higher-molecular-weight polymers. The dot blot analysis using the oligomer-specific A11 antibody demonstrated that the above-mentioned three neurotoxic fractions (3 to 5) were positive for A11, supporting the evidence that the neurotoxic molecules are oligomeric (FIG. 3D). The 2C3-recognized oligomers were detected in fractions 4 and 5 (FIG. 3D). Thus, 2C3 was demonstrated to actually react with neurotoxic A.beta. oligomers (>30 kDa). Furthermore, the majority of the 2C3-recognized oligomers was detected in fraction 5 (>100 kDa) that was the most toxic, and thus the 2C3-recognized oligomers having a molecular weight over 100 kDa were considered to show a strong neurotoxicity (FIG. 3D). Meanwhile, only an extremely small amount of the 1A9-recognized oligomers was distributed in fraction 5 that was the most toxic. This is consistent with the result that the neutralization of neurotoxicity by 1A9 was insufficient (FIGS. 2E and 2G). By contrast, monoclonal 4G8 having no anti-neurotoxic activity detected the A.beta. species distributed in all of the fractions (FIG. 3D). This suggests the possibility that non-toxic and toxic oligomers of the same size co-exist.

[0175] To further assess the toxicity-structure correlation, each fraction was subjected to atomic force microscopy (AFM). The presence of globular particle morphology consistent with the fraction size was detected in the three neurotoxic fractions. FIG. 3E shows the atomic force microscopic images of non-toxic fraction 2 (Fr. 2), toxic fractions 3 (Fr. 3) and 4 (Fr. 4), and the most toxic fraction 5 (Fr. 5). The formation of many granular polymer molecules was clearly observed in the toxic fractions. In particular, fraction 5 was revealed to contain heterogeneous toxic molecules including bead-shaped and ring-shaped molecules in addition to various large and small granular molecules.

Example 4

The Activity of 1A9 and 2C3 to Suppress A.beta. Amyloid Fibril Formation

[0176] Next, the present inventors assessed the activity of 1A9 and 2C3 to suppress A.beta. amyloid fibril formation. The formation of A.beta. 1-42 amyloid fibrils (at 0, 10, 25, and 50 .mu.M) was assessed by measuring the ThT fluorescence for 72 hours at 37.degree. C. Under the conditions used by the present inventors, seed-free A.beta. 1-42 (ThT-negative supernatant fraction obtained by ultracentrifugation at 540,000.times.g) was polymerized into amyloid fibrils by nucleation-dependent polymerization (FIG. 4A). To assess the activity of 1A9 and 2C3 to suppress A.beta. amyloid fibril formation, the present inventors incubated 25 .mu.M seed-free A.beta. 1-42 in the presence or absence of the antibodies at 37.degree. C. for 48 hours. As shown in FIG. 4B, the ThT fluorescence intensity was altered in a 2C3 concentration-dependent manner, while none of monoclonal 1A9 and 4G8 and non-specific IgG2b altered the florescence intensity. Meanwhile, when A.beta. was polymerized by incubation for two hours, 1A9, as well as 2C3, showed the activity to almost completely suppress the fibril formation (FIG. 4C). Since the activity to suppress A.beta. amyloid fibril formation was detected even when the molar ratio of 2C3 to A.beta. was low, 2C3 was inferred to have the activity to inhibit the polymerization nucleus formed de novo or the seed function at an early stage of A.beta. 1-42 amyloid fibril formation. Similar results were obtained by morphological observation. As shown in FIG. 4E (A.beta. 42 alone) and FIG. 4F (A.beta. 42+2C3, 25:3), electron microscopy (EM) demonstrated that the formation of A.beta. amyloid fibrils was partially inhibited in the presence of monoclonal 2C3, while only a weak inhibitory effect was produced in the presence of 1A9 (FIG. 4G). Meanwhile, none of the test antibodies exhibited the effect of lysing or depolymerizing A.beta. 1-42 amyloid fibrils that were formed by incubation for 24 hours (FIG. 4D).

Example 5

Toxicity-Related Oligomers Targeted by 1A9 and 2C3

[0177] To elucidate the structural and kinetic connection between the A.beta. 1-42 oligomerization and amyloid fibril formation, the polymerization time course was analyzed by dot blotting using A11, 1A9, 2C3, and 4G8. As shown in FIG. 5A, the majority of A11 antibody-reactive oligomers was formed during the lag time phase of polymerization (0 to 8 hours), and the ThT fluorescence intensity was relatively weak. During the next fibril extension phase (8 to 24 hours), the level of A11-immunoreactive oligomers reached a plateau, and then was constant (about 20% of the peak level) until 72 hours (plateau phase). It has been demonstrated that, since the anti-oligomer A11 antibody does not recognize amyloid fibrils, the A.beta. oligomer formation can be specifically observed using the antibody (Kayed R, et al., Science 300, 486-489, 2003). Hence, the present results suggest that the A.beta. oligomer formation precedes amyloid fibril formation, and there is an oligomerization state that does not directly enter the amyloid fibril formation pathway. The 2C3-recognized oligomers were kinetically similar to the A11-recognized oligomers, but not the 1A9-recognized oligomers. The 1A9-recognized oligomers were detected only after four hours, and then the immunoreactivity to 1A9 increased twofold over time. This suggests that the 1A9-recognized oligomers are slowly formed. Meanwhile, it was revealed that the 2C3-recognized oligomers are transiently increased during the lag time phase (0 to 8 hours), and then exist at a very low level (less than 5%) in a oligomerized state from 8 to 72 hours. The above-described data obtained by the present inventors suggest the possibility that the A11-, 1A9-, and 2C3-recognized oligomers have structurally and immunologically different conformations or stability, and the 2C3-recognized oligomers are relatively unstable as compared to the 1A9-recognized oligomers.

[0178] To characterize the de novo toxic polymerization state, PC12N were exposed at 37.degree. C. for 48 hours to seed-free A.beta. 1-42 (0 hour), or A.beta. 1-42 pre-incubated for two, four, or 24 hours (FIG. 5B), and the neurotoxic activity was assayed. As shown in FIG. 5B, the immunoblotting analysis using 4G8 revealed that the monomers, dimers, and timers exist even at the 0 hour time point. The pre-incubation of two or four hours resulted in a high-molecular-weight (HMW) smear pattern of 45 to 160 kDa, in addition to the monomers to pentamers. At the time point of 24 hours, the HMW smear was dramatically reduced, and there were two types of major components: a high-molecular-weight species that could not enter the gel and thus remained in the well, and a small amount of the monomers. The HMW smear disappeared after further incubation at 37.degree. C. for 48 hours. As shown in FIGS. 3A and 5C, the molecular sieve experiment revealed that seed-free A.beta. 1-42 is converted into molecular species of 100 kDa or more, and exhibits the strongest toxicity. By SDS-PAGE, it was demonstrated that the toxic molecules include molecular species showing a high-molecular-weight (HMW) smear pattern of 45 to 160 kDa, in addition to the monomer to pentamer species, and the toxic polymers can be easily depolymerized into low-molecular-weight species in the presence of SDS. However, when seed-free A.beta. 1-42 was pre-incubated for two and four hours, the neurotoxic activity of de novo-formed A.beta. oligomers was reduced by about 12.5% and 26%, respectively (FIG. 5C). This result suggests that the level of de novo-formed A.beta. oligomers in the early period of A.beta. polymerization is a determining factor for neurotoxicity, and that the formation reaches a peak in the period of zero to two hours, and then the level of formed A.beta. oligomers reduces over time. Alternatively, there is a possibility that nuclei for the de novo polymerization of A.beta. amyloid fibrils, or amyloid fibrils themselves have the neurotoxicity-neutralizing activity. The present inventors incubated A.beta. 1-42 for two hours, and then removed insoluble A.beta. polymerization nuclei and amyloid fibrils by ultracentrifugation for three hours at 540,000.times.g. The supernatant and pellet obtained by ultracentrifugation at 540,000.times.g exhibited similar levels of thioflavin T signals, suggesting that the 540,000.times.g supernatant contains soluble ThT-positive A.beta. polymers (The ThT binding indicates structural changes to form a .beta. sheet-rich structure, but not fibril formation). The neurotoxicity was restored and enhanced when PC12N was exposed to the soluble polymers (FIG. 5D). This suggests that insoluble A.beta. 1-42 itself has the anti-toxic activity. Under the conditions described above, monoclonal 1A9 completely neutralized the neurotoxicity induced by soluble A.beta. oligomers enriched in P sheet structures, and this neutralizing activity was greater than that of 2C3. Meanwhile, non-specific IgG2b has no effect on the viability of cultured PC12N. Accordingly, it is speculated that neurotoxic 1A9-recognized polymers are basically soluble toxic oligomers that have been slightly stabilized due to some structural change, while neurotoxic 2C3-recognized polymers are basically short-lived oligomeric intermediates that are very unstable due to drastic structural changes during the early stage of polymerization process.

Example 6

Monoclonal 1A9 and 2C3 Recognize A.beta. Oligomers in the Brain Parenchyma

[0179] The present inventors demonstrated the specificity and biological activity of 1A9 and 2C3. Furthermore, the inventors detected 1A9 and 2C3 polymers in the brain by immunohistochemistry. The present inventors performed conventional immunohistochemistry methods to enhance immune reaction by formaldehyde fixation, and formic acid, SDS, or microwave treatment of brain sections. The two antibodies exhibited no immunoreactivity to AD brain by any one of the enhancement methods. Thus, the present inventors pre-treated the sections with Protease K, which is known to improve immunostaining (Wrzolek Mass., et al., Am J Pathol, 141: 343-355, 1992). As a result, many senile plaques were stained with 1A9 (FIG. 6A), 2C3 (FIG. 6B), and A11 (FIG. 6C). Together with the finding from the in vitro experiments by the present inventors that A.beta. amyloid fibrils neutralize the A.beta. oligomer-induced neurotoxicity, the result described above suggests that a senile plaque serves as a defensive reservoir to isolate and store A.beta. oligomers, and thus the interior of the reservoir is hardly accessible for antibodies. Indeed, immunoprecipitation using 1A9 and 2C3 demonstrated that amyloid fractions composed of senile plaques contain A.beta. oligomers recognized by the two antibodies. Thus, the hypothesis of the present inventors was proven to be consistent with the in vivo finding (see FIG. 1B).

[0180] To further assess the existence of "soluble" 1A9- and 2C3-recognized polymers in the brain, the present inventors carried out immunoprecipitation experiments using the two antibodies. Brain homogenates were prepared using Tris-buffered saline (TBS) to avoid chemical modification during the extraction of soluble oligomers. The oligomers having a molecular weight of 4mer, 5mer, 8mer, and 12mer were immunoprecipitated with 1A9 from TBS samples of the cerebral cortex from AD brain (FIG. 6D, lane 2), while the level of the oligomers in the control healthy brain was below the detection limit (lane 3). While the intensity of 4mer, 5mer, and 8mer was comparable between 1A9 (lane 2) and the monoclonal 4G8/6E10 mixture (lane 1), 1A9 appeared to recover a larger amount of 12mer than 4G8/6E10. The immunoprecipitation with 2C3 showed a comparable result (FIG. 6, lanes 4 to 6). Next, the present inventors identified the molecules responsible for the in vivo neurotoxicity in the human entorhinal cortex. It is well known that neurofibrillary tangle (NFT) and nerve cell loss precede the formation of senile plaques in lesions in general elderly populations. The present inventors hypothesized that the lack of functional reservoirs such as senile plaques for 1A9- and 2C3-recognized polymers is harmful for entorhinal cortex neurons, and is a possible cause of memory disturbance. The level of 12mer in the buffer-soluble fractions of previously reported 50 autopsy cases was determined by immunoblotting using monoclonal 1A9 and 2C3. The 50 cases include two AD cases, 35 cases at Braak NFT stages I to II, and 13 cases at NFT stages III to IV (Katsuno et al., Neurology, 64: 687-692, 2005). As shown in FIGS. 6E (1A9) and 6F (2C3), the immunological activity of 1A9- or 2C3-immunoreactive 12mer relative to actin was significantly higher in AD patients as compared to the healthy control group (Braak NFT stages I to II) and mild cognitive impairment group (Braak stages III to IV). Interestingly, the 12mer was accumulated in the entorhinal cortex of the healthy control group (Braak NFT stages I to II) and mild cognitive impairment group (Braak stages III to IV) at a level of about 40% and 60% (the level of AD cases is 100%), respectively (FIGS. 6E and 6F). This result indicates that the accumulation of 12mer precedes the onset of cognitive impairment, and is increased as the Braak NFT stage advances, suggesting that the 1A9- and 2C3-immunoreactive 12mer are polymers responsible for the in vivo neurotoxicity.

Example 7

Monoclonal 1A9 and 2C3 Recognize A.beta. Oligomers in the Cerebrospinal Fluid

[0181] The A.beta. polymers (soluble 1A9- and 2C3-immunoreactive 12mer) responsible for the in vivo neurotoxicity were found in the brain parenchyma. Thus, the present inventors speculated that CSF also contains the polymers. To verify this, the present inventors fractionated pooled CSFs from ten AD patients and ten age-matched healthy individuals as a control by SEC, and assayed the fractions by A.beta. oligomer-specific sandwich ELISA using monoclonal BC05 or BA27 in the capturing and detection systems. The BC05/BC05 oligomer ELISA detected soluble A.beta. 1-42 in fraction 13, while the BA27/BA27 ELISA detected soluble A.beta. 1-40 in fractions 7 to 14 (data not shown). However, in each ELISA, the absorbance (O.D. at 450 nm) was low for sensitive detection of a small amount of A.beta. oligomers in CSF. The detection of A.beta. monomers in the same fractions by BNT77 ELISA showed that lipoprotein-bound A.beta. monomers (fractions 7 to 14) and lipoprotein-free A.beta. monomers (fractions 15 to 17) coexist with A.beta. oligomers in the fractions (FIGS. 7-1A and 7-1B) (Matsubara E, et al., Neurobiol Aging, 25: 833-841, 2004). The level of lipoprotein-bound A.beta. monomer in AD was comparable to that of the healthy control, while the level of lipoprotein-free A.beta.40 monomer (FIG. 7-1A) and A.beta.42 monomer (FIG. 7-1B) in AD was lower as compared to the age-matched healthy control. The present inventors also found that lipoprotein-bound A.beta. monomers, in addition to the oligomers, can be detected when ELISA is designed to use HRP-labeled BC05 or BA27 as a capture antibody. This problem remained unnoticed in the prior art document (Lee E B, et al., J Biol Chem, 281: 4292-4299, 2006), which describes assay methods (for example, 6E10/6E10 ELISA) that are similar to the methods described herein. Since the oligomers and lipoprotein-bound A.beta. monomers are eluted at a comparable retention time in SEC, it is impossible to distinguish them by oligomer ELISA using the same antibody in capturing and detection. Thus, it was revealed that CSF containing lipoproteins is unsuitable for a test sample when A.beta. oligomers are analyzed using A.beta. oligomer-nonselective antibodies.

[0182] To overcome the weaknesses of the prior art methods, the present inventors improved the detection antibodies and samples used in ELISA. Lipoproteins were pre-depleted from CSF, and the resulting lipoprotein-depleted CSF (LPD-CSF) was used as an assay sample. A.beta. oligomer-specific 1A9 and 2C3 were used as detection antibodies for ELISA. Furthermore, chemiluminescence ELISA was developed to enhance the sensitivity. Pooled LPD-CSF (FIGS. 7-1C to D) was fractionated by SEC, and each fraction was analyzed for A.beta. oligomer distribution by luminescence ELISA using 1A9 or 2C3 as a detection antibody. As shown in FIGS. 7-1C to D, A.beta. oligomers were detected in SEC fractions 12 to 15 (relatively large A.beta. with a molecular weight ranging within 18 to 108 kDa, which corresponds to the size of 4mer to 24mer). The level of 1A9- and 2C3-recognized oligomers was elevated in all of the AD patient-derived fractions in which the oligomers were detectable. To assess the usefulness of the A.beta. oligomers as therapeutic markers, the level of A.beta. oligomers in LPD-CSF from AD patients was compared to that from the age-matched healthy control, although a limited number of cases were analyzed. As shown in FIG. 7-2G, 2C3-recognized oligomers composed of A.beta. x-42 were significantly increased in the AD patient group as compared to the normal control group (nonparametric analysis; p=0.0103). By contrast, for 2C3-recognized oligomers composed of A.beta. x-42, there was no significant difference between the two groups. Meanwhile, the level of 1A9-recognized oligomers composed of A.beta. x-42 was higher in AD than in the control, although the difference was not statistically significant. For 1A9-recognized oligomers composed of A.beta. x-40, there was no significant difference between the two groups (FIG. 7-2E). The structural change from A.beta. monomer to oligomer occurs in the earliest period of the process of A.beta. polymerization. The ratio between A.beta. oligomer and monomer (O/M index) can be used as a clinical indicator reflecting the pathological conditions of AD. As shown in FIGS. 7-2F and 7-2H, the O/M indices for A.beta.42 and A.beta.40 were significantly increased in the AD patient group as compared to the healthy control group (1A9, P=0.0137 for A.beta.42 and P=0.0429 for A.beta.40; 2C3, P=0.0012 for A.beta.2 and P=0.0051 for A.beta.40). The results described above show that the 1A9- and 2C3-positive three-dimensional structures are present as A.beta. oligomers in LPD-CSF, and increased in AD patients. In addition, the results obtained by the present inventors demonstrated that the structural conversion of lipoprotein-free soluble A.beta. to the oligomeric intermediates occurs in CSF of AD patients, and the oligomers can be detected as useful biological markers for diagnosis of sporadic AD.

Example 8

Passive Immunotherapy Using Monoclonal 1A9 and 2C3 Prevents the Onset of Memory Disturbance in Tg2576

[0183] To assess the in vivo preventive/therapeutic effect of passive immunotherapy based on the administration of 1A9 (n=13) or 2C3 (n=11), the present inventors administered 1A9 or 2C3 (0.4 mg/kg/week), or PBS to Tg2576 mice via the caudal vein during the 4 to 13 month period. The memory function was assessed at 13 months old in terms of the following four types of learning/behavioral paradigms:

(1) short-term memory in Y-maze test (FIG. 8A); (2) object recognition memory in novel object recognition test (FIG. 8B); (3) spacial memory in water maze test (FIG. 8C); and (4) associative emotional memory in contextual fear conditioning test (FIG. 8D).

[0184] As compared to 1A9- and 2C3-administered Tg2576 mice, PBS-administered Tg2576 mice showed significant learning and behavioral impairments (FIG. 8A to 8D). Unlike the memory function of PBS-administered Tg2576 mice (n=10), the memory function of 1A9 and 2C3-administered Tg2576 mice was indistinguishable from that of age-matched non-administered wild type cohort mice, which was previously determined. Therefore, 1A9 and 2C3-administered Tg2576 mice were shown to retain both short- and long-term memory, which were impaired in the PBS administration group. That is, the present inventors obtained evidence supporting the view that the onset of memory disturbance, in particular AD, can be prevented by conducting passive immunotherapy targeting A.beta. oligomers before the onset. Furthermore, the result described above presents the first in vivo evidence that directly indicates that A.beta. oligomers are responsible for the onset of memory disturbance.

Example 9

Monoclonal 1A9 Prevents A.beta. Accumulation in the Brain of Tg2576

[0185] Tg2576 mice administered with PBS (n=10) and Tg2576 mice treated with passive immunotherapy during the 4 to 13 month period (1A9 administration group, n=13; 2C3 administration group, n=11) were dissected after the learning/behavioral experiments. The amount of A.beta. accumulated in the brain (cerebral cortex vs. hippocampus) was determined in the following three fractions (150 mg/extract) prepared by serial extraction: soluble fraction in Tris buffer containing protease inhibitors; 2% SDS-soluble amyloid fraction; and 2% SDS-insoluble and 70% formic acid-soluble amyloid fraction. It is considered that non-accumulative, physiological A.beta. molecules are contained in the Tris buffer fraction, while 2% SDS-soluble A.beta. includes A.beta. in diffuse senile plaques before amyloid fibril formation, immunocytochemically undetectable A.beta., and conformationally altered, accumulative soluble oligomeric A.beta.. A.beta. was selectively quantified by A.beta.40 and A.beta.42 end-specific ELISA (BNT77/BA27 specific for A.beta.40, BNT77/BC05 specific for A.beta.42, WAKO kit). There was no marked difference among the three groups in the A.beta. concentration in the Tris buffer fraction where the major components were non-accumulative, physiological A.beta. molecules (FIGS. 9A and 9C, A.beta. x-40; FIGS. 9B and 9D, A.beta. x-42). Regarding soluble A.beta. accumulated in the brain (SDS fraction), a significant suppressive effect on the accumulation of A.beta. x-40 and A.beta. x-42 in the cerebral cortex was observed only in the 1A9 administration group (FIG. 9E, A.beta. x-40; FIG. 9F, A.beta. x-42). No accumulation-suppressive effect was observed in the hippocampus (FIG. 9G, A.beta. x-40; FIG. 9H, A.beta. x-42). Meanwhile, regarding insoluble A.beta. accumulated in the brain (FA fraction), a significant suppressive effect on the accumulation of A.beta. x-40 in the cerebral cortex was observed only in the 1A9 administration group (FIG. 9I, A.beta. x-40; FIG. 9J, A.beta. x-42). No accumulation-suppressive effect was observed in the hippocampus (FIG. 9K, A.beta. x-40; FIG. 9L, A.beta. x-42). The A11 immunoblot analysis of the SDS-soluble fractions showed a suppressive effect on the accumulation of A11-positive oligomer (4mer) in the cerebral cortex in the two antibody treatment groups (FIG. 9M).

Example 10

Plasma A.beta. Oligomers are Increased by Passive Immunotherapy with 1A9 and 2C3

[0186] There was no significant difference in the plasma A.beta. concentration among the following three groups: Tg2576 mice administered with PBS (n=10), and Tg2576 mice treated with passive immunotherapy during the 4 to 13 month period (1A9 administration group, n=13; 2C3 administration group, n=11) (FIG. 10A, A.beta. x-40; FIG. 10B, A.beta. x-42). There was also no significant difference in the A.beta.40/42 ratio (FIG. 10C).

[0187] In order to elucidate the mechanism underlying the preventive effect of passive immunotherapy with 1A9 and 2C3 (IVIg) against the AD-like phenotype in Tg2576 mice, the present inventors assessed the level of physiological saline-soluble and -insoluble A.beta. oligomers in pooled brain homogenates, and the level of A.beta. oligomers in the peripheral blood and plasma. There was no difference in the amount of physiological saline-soluble A.beta. oligomers in the pooled brain homogenates among the treatment groups (FIG. 10D). Meanwhile, the amount of insoluble A.beta. oligomers was shown to be reduced in the 1A9 and 2C3 treatment groups (FIG. 10E). Furthermore, pooled plasma from each group (albumin-depleted plasma, upper part of Panel F; albumin/lipoprotein-depleted plasma, lower part of Panel F) was assayed for A.beta. oligomers by A11 dot blotting. The result shows that the oligomers were present in the plasma from PBS-administered Tg2576 mice (FIG. 10F). A11-positive oligomers in plasma were clearly increased in the passive immunotherapy groups as compared to the PBS administration group (FIG. 10F). The proportion of 2C3-recognized oligomers in a lipoprotein-bound form was greater than that of 1A9-recognized oligomers (lower part of Panel F). Furthermore, plasma A.beta. oligomers were detected by A11 immunoprecipitation. The result shows that the oligomers of about 200 kDa were increased in Tg2576 mice treated with passive immunotherapy as compared to the PBS administration group (FIG. 10G). The increase in plasma A.beta. oligomers in the passive immunotherapy groups can be considered to directly reflect enhanced cerebral clearance. Thus, the present inventors obtained evidence that direct target molecules for intravenous passive immunotherapy are also present in blood in addition to brain, and that oligomer-selective cerebral clearance can be enhanced through peripheral sites of action. That is, the present inventors showed the clinical usefulness of the intravenous passive immunotherapy.

Example 11

Formation of Senile Amyloid Plaques and Swollen Dystrophic Neurites can be Suppressed by Passive Immunotherapy Using 1A9 and 2C3

[0188] Immunohistochemical A.beta. deposition was suppressed in the passive immunotherapy groups (FIG. 11A). The formation of thioflavin S-positive senile amyloid plaques was significantly suppressed in both the cerebral cortex and hippocampus (FIG. 11B, upper part), and the reduction was also clearly demonstrated by histochemistry (FIG. 11B, lower part). The formation of synaptophysin-positive swollen dystrophic neurites was also significantly suppressed in the passive immunotherapy groups (FIG. 11C).

Example 12

Immunostaining Analysis Using Anti-Synaptophysin and Anti-Drebrin Antibodies

[0189] 1A9 and 2C3 suppressed the presynaptic and postsynaptic degeneration in the cerebral neocortex (FIG. 12).

Example 13

The Antibodies Translocate to the Brain

[0190] The existence/localization of deposited A.beta. and cerebral mouse IgG was assessed using a confocal laser microscope. The result shows that mouse IgG is localized almost independently of deposited A.beta. within the areas containing diffuse senile plaques. Mouse IgG was observed only in the passive immunotherapy groups (1A9, FIG. 13A; 2C3, FIG. 13B), but not in the PBS administration group (FIG. 13C). Thus, a fraction of the antibodies administered into the blood was considered to translocate to the brain. This result shows that the preventive effect on memory disturbance was produced not only through the direct neutralization of the toxicity of soluble A.beta. polymers by the antibodies translocated to the brain, but also through the clearance of soluble A.beta. polymers in the form of a complex with the antibodies into the blood. Thus, the therapeutic effect was considered to be based on multiple action mechanisms.

Example 14

Preparation of A.beta. Oligomer-Specific Monoclonal Antibodies (5A5, 5A9, 4F7, 4H5, 6E4, and 6H4) and Dot Blot Analysis

[0191] 33 clones prepared by the above-described method that uses the A.beta. 1-40 oligomer as an antigen were assessed by dot blot analysis. The result showed that the six types of monoclonal antibodies specifically recognize A.beta. oligomers. As shown below, the isotype of the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) was determined:

4F7: .kappa. for the L chain, and IgG2a for the H chain; 4H5: .kappa. for the L chain, and IgG2a for the H chain; 5A5: .kappa. for the L chain, and IgG2b for the H chain; 5A9: .kappa. for the L chain, and IgG2b for the H chain; 6E4: .kappa. for the L chain, and IgG1 for the H chain; and 6H4: .kappa. for the L chain, and IgG2b for the H chain. Furthermore, the immuno-dot blot analysis showed that, as with 2C3 described above, the 4F7, 4H5, 5A5, 5A9, 6E4, and 6H4 antibodies specifically bind to A.beta. oligomers but do not recognize A.beta. monomers (see FIG. 14).

Example 15

Inhibition ELISA

[0192] To assess the A.beta. oligomer-selective binding activity of the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4), each antibody was mixed with stepwise-diluted A.beta. oligomers or monomers ("inhibitors"), and the pre-mixed solutions were added to A.beta. oligomer-immobilized 96-well immunoplates, and then incubated (see the "Methods" section). The commercially available 4G8 and 6E10 antibodies were used as control antibodies that nonselectively bind to A.beta. oligomers and monomers. When an antibody selectively binds to A.beta. oligomers, the antibody pre-mixed with A.beta. monomers does not bind to the A.beta. monomers in the solution, and therefore can bind to immobilized A.beta. oligomers. On the other hand, the antibody pre-mixed with A.beta. oligomers binds to the A.beta. oligomers in the solution, and therefore the amount of antibody bound to immobilized A.beta. oligomers is reduced with the increase in inhibitor concentration. The results for the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) showed concentration-dependent reduction in the amount of bound antibody when A.beta. oligomers were used. In contrast, no such strong reduction in binding was detected when A.beta. monomers were used (see FIG. 15). Meanwhile, for 4G8 and 6E10, the concentration-dependent reduction in the amount of bound antibody was observed when A.beta. monomers and oligomers were used (see FIG. 15). These results suggest that the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) selectively bind to A.beta. oligomers.

Example 16

The Activity of the Six Types of Antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) to Neutralize A.beta.-Induced Neurotoxicity

[0193] To assess whether the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) have an activity of neutralizing A.beta.-induced neurotoxicity, human neuroblastoma cells (SH-SY5Y) were cultured in a medium containing A.beta. 1-42 (12.5 .mu.M) in the presence or absence of the antibodies for 24 hours, and the change in A.beta. 1-42-induced cytotoxicity was monitored. As a result, the cytotoxicity was enhanced by addition of control IgG (3F1). Although the cytotoxicity was also increased by addition of the 4F7 and 4H5 antibodies, the increase was smaller than that observed for 3F1 (see FIG. 16). The remaining four types of antibodies (5A5, 5A9, 6E4, and 6H4) were found to markedly reduce the cytotoxicity (see FIG. 16). The results described above demonstrate that the four types of antibodies (5A5, 5A9, 6E4, and 6H4) have a strong activity of neutralizing A.beta.-induced neurotoxicity. Since 4F7 and 4H5 lowered the cytotoxicity as compared to control IgG, these antibodies are also inferred to have an activity of neutralizing A.beta.-induced neurotoxicity.

Example 17

The Activity of the Six Types of Antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 61-14) to Suppress A.beta. Amyloid Fibril Formation

[0194] To assess whether the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) have an activity of suppressing A.beta. amyloid fibril formation, the formation of A.beta. amyloid fibrils was detected by the ThT fluorescence intensity assay method in a solution (medium) whose composition was the same as that used in the experiment for A.beta.-induced neurotoxicity (see the "Methods" section). 6E4 and 6H4 were found to suppress the fibril formation in an antibody concentration-dependent manner (see FIG. 17). The other four antibodies (4F7, 4H5, 5A5, and 5A9) were also inferred to have an activity of suppressing the fibril formation, since the antibodies exhibited the tendency of suppressing the fibril formation as compared to control IgG.

Discussion

[0195] The data obtained by the present inventors show that monoclonal 1A9 and 2C3 specifically recognize the "neurotoxic epitope" and "polymerization epitope" of soluble A.beta. polymers that are responsible for the toxic activity and neurofibril formation activity. Since monoclonal 1A9 and 2C3 do not react with soluble A.beta. monomers, which are physiological molecules, it can be concluded that a three-dimensional structure having the epitope that is recognized by 1A9 or 2C3 is specific to soluble oligomeric polymers. The experiments using ultrafiltration and molecular sieve revealed that the size of 1A9- and 2C3-immunoreactive oligomers is greater than 100 kDa (>20mer). The result of morphological observation by AFM demonstrated that the toxic polymers are morphologically heterogeneous (granular, bead-shaped, and ring-shaped).

[0196] To demonstrate that the toxic polymers are actually bioactive molecules that exhibit in vivo synaptic toxicity, the present inventors commenced treatment of young Tg2576 mice before the onset of memory disturbance with anti-A.beta. oligomer passive immunotherapy targeting 1A9- and 2C3-recognized toxic polymers. For the first time, the present inventors presented evidence supporting that age-dependent memory deterioration that naturally develops in Tg2576 mice can be prevented by passive immunotherapy using anti-A.beta. oligomer-specific antibodies (1A9 and 2C3). Herein, short-term memory disturbance assessed by the Y-maze test is similar to the A.beta. accumulation-associated memory disturbance observed in mild cognitive impairment (MCI) and early AD. The Y-maze test showed excellent and almost normal results in Tg2576 mice administered with 1A9 and 2C3, respectively. When assessed by the novel object recognition task, Morris water maze, and contextual fear conditioning task, the long-term memory was maintained nearly normal by the anti-A.beta. oligomer antibodies.

[0197] A selective increase in A11-positive oligomers in blood was observed in the antibody-treated mouse groups as compared to the PBS treatment group, which is consistent with the ability of the antibodies to prevent the onset of memory disturbance (the memory maintenance ability). The 1A9 antibody treatment also exhibited the effect of suppressing cerebral A.beta. accumulation. The 2C3 antibody treatment demonstrated a higher blood level of A11-positive oligomers as compared to the 1A9 antibody treatment. However, the cerebral A.beta. accumulation-suppressing effect of the 2C3 antibody treatment was unclear. Accordingly, 1A9-recognized oligomers were considered to have greater contribution to the cerebral A.beta. accumulation than 2C3-recognized oligomers. The involvement of the polymers in cerebral A.beta. accumulation can be explained based on the following assumption: neurotoxic 1A9 polymers are soluble toxic oligomers that are somewhat conformationally, while neurotoxic 2C3 polymers are very unstable, short-lived oligomeric intermediates that appear at an early stage of the polymerization process, the conformation of which is easily changed.

[0198] The present inventors disclose herein the in vivo preventive effect of anti-oligomer antibodies on Alzheimer's disease, and this is the first evidence that directly demonstrates that toxic A.beta. oligomers formed in vivo can inhibit the functions of nerve cells, thereby inducing the symptoms of Alzheimer's disease.

[0199] The data obtained by the present inventors is also the first evidence supporting the view that A.beta. exhibits in vivo neurotoxicity in the human brain. It is well known that the human entorhinal cortex is an area that is easily affected with AD. In this area, NFT formation and nerve cell loss precede the formation of senile plaques. Thus, the entorhinal cortex is an exceptional area to which the commonly accepted amyloid cascade hypothesis cannot be applied. However, this inconsistency has been neglected and remained unstudied for a long time.

[0200] The present inventors proposed and examined the hypothesis that previously unidentifiable, invisible A.beta. oligomers are harmful for nerve cells in the entorhinal cortex and cause memory disturbance. To examine this hypothesis, the present inventors performed semi-quantitative analysis of 1A9- and 2C3-immunoreactive 12mer in the entorhinal cortex of elderly individuals who were mostly at Braak NFT stages I to III. The 1A9- and 2C3-immunoreactive 12mer were already present in the entorhinal cortex of healthy individuals at Braak NFT stages I to II, and increased with the advancement of Braak NFT stage. The 12mer was found to be significantly increased in AD. Thus, the appearance of 1A9- and 2C3-immunoreactive 12mer was demonstrated to precede the onset of cognitive impairment in the human brain. On the other hand, by biochemical and immunohistochemical techniques, it was demonstrated that senile plaques contain 1A9- and 2C3-immunoreactive A.beta. oligomers. In addition, insolubilized amyloid fibrils themselves were revealed to have an activity of neutralizing the neurotoxicity. These findings suggest that, under conditions where A.beta. oligomers are present without senile plaque formation, A.beta. oligomers exert in vivo toxicity and thus can be a cause of memory disturbance.

[0201] As described above, the data of the present inventors show for the first time evidence that directly demonstrates in vivo the memory disturbance resulting from synaptic dysfunction caused by endogenous A.beta. oligomers. Although active immunotherapy (Janus D, 2000, Nature; Morgan D, 2000, Nature) and passive immunotherapy (Bard F, 2222, Nat med; DeMattos R B, PNAS, 2001) have been used previously, the mechanism by which learning disability and memory disturbance can be prevented has remained a matter of conjecture. One widely proposed possibility is that the antibodies reach the brain through the blood-brain barrier and directly neutralize in vivo soluble A.beta. oligomers that cause memory impairment. The second possibility, the "sink theory", is that the antibodies act peripherally to deplete the peripheral blood A.beta. pool and thus activate A.beta. clearance from the brain. DeMattos et al. have reported that a peripherally administered anti-A.beta. antibody rapidly transports not only cerebral A.beta. monomers but also A.beta. dimers into plasma, and also cerebral A.beta. into CSF (DeMattos R B et al., PNAS, 98; 8850-8855, 2001). The present inventors also revealed that A.beta. oligomers are present in human CSF and increased in AD patients. Thus, the present inventors demonstrated that the A.beta. oligomers can be used as diagnostic markers for AD. Furthermore, the present inventors presented the first evidence supporting the view that A.beta. oligomers are present in the plasma of Tg2576 mice, and, in passive immunotherapy by which A.beta. oligomers are specifically captured and neutralized through intravenous injection, intracerebral antibody delivery is not required and the clearance of A.beta. oligomers from the brain to blood can be enhanced at the peripheral sites of action, i.e., blood vessels. In addition, the present inventors presented the first evidence that passive immunotherapy can suppress senile amyloid plaque formation, and indirectly suppress nerve cell damage (swollen dystrophic neurite formation) through senile amyloid plaque suppression. These results confirm that the A.beta. oligomer is the molecular basis for the onset of Alzheimer's disease, and selective control using oligomer-specific antibodies enables the control of Alzheimer's disease from a prophylactic viewpoint, in addition to a therapeutic viewpoint. Furthermore, a fraction of the administered antibodies was proven to translocate into the brain. This suggests that the effect of suppressing memory disturbance is exerted by a combination of multiple actions such as direct neutralization of soluble A.beta. oligomers in the brain, transport of antibody-A.beta. oligomer immune complexes into blood by the neonatal Fc receptor (Deane R, 2005, J Neurosci), and the "sink" action described above.

[0202] The establishment of accurate pre-onset diagnosis to identify cases at a high risk of developing AD is essential to design preventive/therapeutic strategies. The significant increase in the CSF O/M ratio in AD, which is reported herein, is expected to be one of the leading candidates for pre-onset diagnostic markers.

INDUSTRIAL APPLICABILITY

[0203] The antibodies provided by the present invention can be used, for example, in intravenous injection-based preventive passive immunotherapy for Alzheimer's disease, and as biological markers for pre-onset diagnosis, disease monitoring, drug efficacy monitoring/assessment for the disease, and such.

[0204] Furthermore, the antibodies of the present invention are expected to greatly contribute to the establishment of preventive/therapeutic methods for Alzheimer's disease that are selective to molecules responsible for evoking the pathological conditions of the disease, and the establishment of early diagnostic markers. The present inventors obtained evidence supporting that antibody therapies, even when they target intracerebral pathological conditions, can be satisfactorily achieved by peripheral intravenous administration, without the need to consider intracerebral transfer of the antibodies. In addition, the present inventors obtained evidence demonstrating that a fraction of administered antibodies translocates to the brain and produces a direct effect even in peripheral intravenous administration therapy, again without the need to consider intracerebral transfer of the antibodies. Thus, the present invention is expected to rapidly accelerate the progress of antibody therapeutics for Alzheimer's disease.

Sequence CWU 1

1

1201452PRTMus musculus 1Asp Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Arg Lys Leu Ser Cys Ala Thr Ser Gly Phe Thr Phe Ser Ser Phe 20 25 30Gly Met His Trp Val Arg Gln Ala Pro Glu Lys Gly Leu Glu Trp Val 35 40 45Ala Tyr Ile Ser Ser Gly Ser Ser Ala Ile Tyr Tyr Ala Asp Thr Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Pro Lys Asn Thr Leu Phe65 70 75 80Leu Gln Met Thr Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95Ala Arg Ser Gly Asp Thr Met Asp Tyr Trp Gly Gln Gly Thr Ser Val 100 105 110Thr Val Ser Ser Ala Lys Thr Thr Pro Pro Ser Val Tyr Pro Leu Ala 115 120 125Pro Gly Cys Gly Asp Thr Thr Gly Ser Ser Val Thr Leu Gly Cys Leu 130 135 140Val Lys Gly Tyr Phe Pro Glu Ser Val Thr Val Thr Trp Asn Ser Gly145 150 155 160Ser Leu Ser Ser Ser Val His Thr Phe Pro Ala Leu Leu Gln Ser Gly 165 170 175Leu Tyr Thr Met Ser Ser Ser Val Thr Val Pro Ser Ser Thr Trp Pro 180 185 190Ser Gln Thr Val Thr Cys Ser Val Ala His Pro Ala Ser Ser Thr Thr 195 200 205Val Asp Lys Lys Leu Glu Pro Ser Gly Pro Ile Ser Thr Ile Asn Pro 210 215 220Cys Pro Pro Cys Lys Glu Cys His Lys Cys Pro Ala Pro Asn Leu Glu225 230 235 240Gly Gly Pro Ser Val Phe Ile Phe Pro Pro Asn Ile Lys Asp Val Leu 245 250 255Met Ile Ser Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Val Ser 260 265 270Glu Asp Asp Pro Asp Val Gln Ile Ser Trp Phe Val Asn Asn Val Glu 275 280 285Val His Thr Ala Gln Thr Gln Thr His Arg Glu Asp Tyr Asn Ser Thr 290 295 300Ile Arg Val Val Ser Thr Leu Pro Ile Gln His Gln Asp Trp Met Ser305 310 315 320Gly Lys Glu Phe Lys Cys Lys Val Asn Asn Lys Asp Leu Pro Ser Pro 325 330 335Ile Glu Arg Thr Ile Ser Lys Ile Lys Gly Leu Val Arg Ala Pro Gln 340 345 350Val Tyr Ile Leu Pro Pro Pro Ala Glu Gln Leu Ser Arg Lys Asp Val 355 360 365Ser Leu Thr Cys Leu Val Val Gly Phe Asn Pro Gly Asp Ile Ser Val 370 375 380Glu Trp Thr Ser Asn Gly His Thr Glu Glu Asn Tyr Lys Asp Thr Ala385 390 395 400Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe Ile Tyr Ser Lys Leu Asn 405 410 415Met Lys Thr Ser Lys Trp Glu Lys Thr Asp Ser Phe Ser Cys Asn Val 420 425 430Arg His Glu Gly Leu Lys Asn Tyr Tyr Leu Lys Lys Thr Ile Ser Arg 435 440 445Ser Pro Gly Lys 45021359DNAMus musculus 2gatgtgcagc tggtggagtc tgggggaggc ttagtgcagc ctggagggtc ccggaaactc 60tcctgtgcaa cctctggatt cactttcagt agctttggaa tgcactgggt tcgtcaggct 120ccagagaagg gactggagtg ggtcgcatac attagtagtg gcagtagtgc catctactat 180gcagacacag tgaagggccg attcaccatc tccagagaca atcccaagaa caccctgttc 240ctgcaaatga ccagtctaag gtctgaggac acggccatgt attactgtgc aagatctggg 300gatactatgg actactgggg tcaaggaacc tcagtcaccg tctcctcagc caaaacaaca 360cccccatcag tctatccact ggcccctggg tgtggagata caactggttc ctccgtgact 420ctgggatgcc tggtcaaggg ctacttccct gagtcagtga ctgtgacttg gaactctgga 480tccctgtcca gcagtgtgca caccttccca gctctcctgc agtctggact ctacactatg 540agcagctcag tgactgtccc ctccagcacc tggccaagtc agaccgtcac ctgcagcgtt 600gctcacccag ccagcagcac cacggtggac aaaaaacttg agcccagcgg gcccatttca 660acaatcaacc cctgtcctcc atgcaaggag tgtcacaaat gcccagctcc taacctcgag 720ggtggaccat ccgtcttcat cttccctcca aatatcaagg atgtactcat gatctccctg 780acacccaagg tcacgtgtgt ggtggtggat gtgagcgagg atgacccaga cgtccagatc 840agctggtttg tgaacaacgt ggaagtacac acagctcaga cacaaaccca tagagaggat 900tacaacagta ctatccgggt ggtcagcacc ctccccatcc agcaccagga ctggatgagt 960ggcaaggagt tcaaatgcaa ggtcaacaac aaagacctcc catcacccat cgagagaacc 1020atctcaaaaa ttaaagggct agtcagagct ccacaagtat acatcttgcc gccaccagca 1080gagcagttgt ccaggaaaga tgtcagtctc acttgcctgg tcgtgggctt caaccctgga 1140gacatcagtg tggagtggac cagcaatggg catacagagg agaactacaa ggacaccgca 1200ccagtcctgg actctgacgg ttcttacttc atatatagca agctcaatat gaaaacaagc 1260aagtgggaga aaacagattc cttctcatgc aacgtgagac acgagggtct gaaaaattac 1320tacctgaaga agaccatctc ccggtctccg ggtaaatga 13593219PRTMus musculus 3Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Asn Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Ser Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Val 85 90 95Ser His Val Pro Pro Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys 100 105 110Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu 115 120 125Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe 130 135 140Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg145 150 155 160Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser 165 170 175Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu 180 185 190Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser 195 200 205Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 2154660DNAMus musculus 4gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gaacattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagcgtgg aggctgagga tctgggagtt tattactgct ttcaagtttc acatgttcct 300ccgacgttcg gtggaggcac caagctggaa atcaaacggg ctgatgctgc accaactgta 360tccatcttcc caccatccag tgagcagtta acatctggag gtgcctcagt cgtgtgcttc 420ttgaacaact tctaccccaa agacatcaat gtcaagtgga agattgatgg cagtgaacga 480caaaatggcg tcctgaacag ttggactgat caggacagca aagacagcac ctacagcatg 540agcagtaccc tcacgttgac caaggacgag tatgaacgac ataacagcta tacctgtgag 600gccactcaca agacatcaac ttcacccatt gtcaagagct tcaacaggaa tgagtgttag 6605116PRTMus musculus 5Asp Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Arg Lys Leu Ser Cys Ala Thr Ser Gly Phe Thr Phe Ser Ser Phe 20 25 30Gly Met His Trp Val Arg Gln Ala Pro Glu Lys Gly Leu Glu Trp Val 35 40 45Ala Tyr Ile Ser Ser Gly Ser Ser Ala Ile Tyr Tyr Ala Asp Thr Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Pro Lys Asn Thr Leu Phe65 70 75 80Leu Gln Met Thr Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95Ala Arg Ser Gly Asp Thr Met Asp Tyr Trp Gly Gln Gly Thr Ser Val 100 105 110Thr Val Ser Ser 1156348DNAMus musculus 6gatgtgcagc tggtggagtc tgggggaggc ttagtgcagc ctggagggtc ccggaaactc 60tcctgtgcaa cctctggatt cactttcagt agctttggaa tgcactgggt tcgtcaggct 120ccagagaagg gactggagtg ggtcgcatac attagtagtg gcagtagtgc catctactat 180gcagacacag tgaagggccg attcaccatc tccagagaca atcccaagaa caccctgttc 240ctgcaaatga ccagtctaag gtctgaggac acggccatgt attactgtgc aagatctggg 300gatactatgg actactgggg tcaaggaacc tcagtcaccg tctcctca 3487112PRTMus musculus 7Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Asn Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Ser Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Val 85 90 95Ser His Val Pro Pro Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys 100 105 1108336DNAMus musculus 8gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gaacattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagcgtgg aggctgagga tctgggagtt tattactgct ttcaagtttc acatgttcct 300ccgacgttcg gtggaggcac caagctggaa atcaaa 33695PRTMus musculus 9Ser Phe Gly Met His1 51015DNAMus musculus 10agctttggaa tgcac 151110PRTMus musculus 11Tyr Ile Ser Ser Gly Ser Ser Ala Ile Tyr1 5 101230DNAMus musculus 12tacattagta gtggcagtag tgccatctac 30137PRTMus musculus 13Ser Gly Asp Thr Met Asp Tyr1 51421DNAMus musculus 14tctggggata ctatggacta c 211516PRTMus musculus 15Arg Ser Ser Gln Asn Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu1 5 10 151648DNAMus musculus 16agatctagtc agaacattgt acatagtaat ggaaacacct atttagaa 48177PRTMus musculus 17Lys Val Ser Asn Arg Phe Ser1 51821DNAMus musculus 18aaagtttcca accgattttc t 21199PRTMus musculus 19Phe Gln Val Ser His Val Pro Pro Thr1 52027DNAMus musculus 20tttcaagttt cacatgttcc tccgacg 2721455PRTMus musculus 21Gln Val Thr Leu Lys Glu Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Thr Thr Ser 20 25 30Ala Met Gly Val Ser Trp Val Arg Gln Pro Ser Arg Lys Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Ser Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Thr Ala Thr Tyr Tyr 85 90 95Cys Ala Arg Lys Gly Leu Gly Gly Ala Met Asp Tyr Trp Gly Gln Gly 100 105 110Thr Ser Val Thr Val Ser Ser Ala Lys Thr Thr Pro Pro Ser Val Tyr 115 120 125Pro Leu Ala Pro Gly Cys Gly Asp Thr Thr Gly Ser Ser Val Thr Leu 130 135 140Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Ser Val Thr Val Thr Trp145 150 155 160Asn Ser Gly Ser Leu Ser Ser Ser Val His Thr Phe Pro Ala Leu Leu 165 170 175Gln Ser Gly Leu Tyr Thr Met Ser Ser Ser Val Thr Val Pro Ser Ser 180 185 190Thr Trp Pro Ser Gln Thr Val Thr Cys Ser Val Ala His Pro Ala Ser 195 200 205Ser Thr Thr Val Asp Lys Lys Leu Glu Pro Ser Gly Pro Ile Ser Thr 210 215 220Ile Asn Pro Cys Pro Pro Cys Lys Glu Cys His Lys Cys Pro Ala Pro225 230 235 240Asn Leu Glu Gly Gly Pro Ser Val Phe Ile Phe Pro Pro Asn Ile Lys 245 250 255Asp Val Leu Met Ile Ser Leu Thr Pro Lys Val Thr Cys Val Val Val 260 265 270Asp Val Ser Glu Asp Asp Pro Asp Val Gln Ile Ser Trp Phe Val Asn 275 280 285Asn Val Glu Val His Thr Ala Gln Thr Gln Thr His Arg Glu Asp Tyr 290 295 300Asn Ser Thr Ile Arg Val Val Ser Thr Leu Pro Ile Gln His Gln Asp305 310 315 320Trp Met Ser Gly Lys Glu Phe Lys Cys Lys Val Asn Asn Lys Asp Leu 325 330 335Pro Ser Pro Ile Glu Arg Thr Ile Ser Lys Ile Lys Gly Leu Val Arg 340 345 350Ala Pro Gln Val Tyr Ile Leu Pro Pro Pro Ala Glu Gln Leu Ser Arg 355 360 365Lys Asp Val Ser Leu Thr Cys Leu Val Val Gly Phe Asn Pro Gly Asp 370 375 380Ile Ser Val Glu Trp Thr Ser Asn Gly His Thr Glu Glu Asn Tyr Lys385 390 395 400Asp Thr Ala Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe Ile Tyr Ser 405 410 415Lys Leu Asn Met Lys Thr Ser Lys Trp Glu Lys Thr Asp Ser Phe Ser 420 425 430Cys Asn Val Arg His Glu Gly Leu Lys Asn Tyr Tyr Leu Lys Lys Thr 435 440 445Ile Ser Arg Ser Pro Gly Lys 450 455221368DNAMus musculus 22caggttactc tgaaagagtc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctggatt ttcactgacc acttctgcta tgggtgtgag ctgggttcgt 120cagccttcaa gaaagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag caaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gatactgcca catactactg tgctcgaaag 300ggactgggag gtgctatgga ctactggggt caaggaacct cagtcaccgt ctcctcagcc 360aaaacaacac ccccatcagt ctatccactg gcccctgggt gtggagatac aactggttcc 420tccgtgactc tgggatgcct ggtcaagggc tacttccctg agtcagtgac tgtgacttgg 480aactctggat ccctgtccag cagtgtgcac accttcccag ctctcctgca gtctggactc 540tacactatga gcagctcagt gactgtcccc tccagcacct ggccaagtca gaccgtcacc 600tgcagcgttg ctcacccagc cagcagcacc acggtggaca aaaaacttga gcccagcggg 660cccatttcaa caatcaaccc ctgtcctcca tgcaaggagt gtcacaaatg cccagctcct 720aacctcgagg gtggaccatc cgtcttcatc ttccctccaa atatcaagga tgtactcatg 780atctccctga cacccaaggt cacgtgtgtg gtggtggatg tgagcgagga tgacccagac 840gtccagatca gctggtttgt gaacaacgtg gaagtacaca cagctcagac acaaacccat 900agagaggatt acaacagtac tatccgggtg gtcagcaccc tccccatcca gcaccaggac 960tggatgagtg gcaaggagtt caaatgcaag gtcaacaaca aagacctccc atcacccatc 1020gagagaacca tctcaaaaat taaagggcta gtcagagctc cacaagtata catcttgccg 1080ccaccagcag agcagttgtc caggaaagat gtcagtctca cttgcctggt cgtgggcttc 1140aaccctggag acatcagtgt ggagtggacc agcaatgggc atacagagga gaactacaag 1200gacaccgcac cagtcctgga ctctgacggt tcttacttca tatatagcaa gctcaatatg 1260aaaacaagca agtgggagaa aacagattcc ttctcatgca acgtgagaca cgagggtctg 1320aaaaattact acctgaagaa gaccatctcc cggtctccgg gtaaatga 136823219PRTMus musculus 23Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Leu His Ser 20 25 30Asn Gly Asn Thr Tyr Leu His Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser 85 90 95Thr His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 110Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu 115 120 125Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe 130 135 140Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg145 150 155 160Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser 165 170 175Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu 180 185 190Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser 195 200 205Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 21524660DNAMus musculus 24gatgttgtga tgacccaaac tccgctctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagccttcta cacagtaatg gaaacaccta tttacattgg 120tacctgcaga agccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga

tctgggagtt tatttctgct ctcaaagtac acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaacggg ctgatgctgc accaactgta 360tccatcttcc caccatccag tgagcagtta acatctggag gtgcctcagt cgtgtgcttc 420ttgaacaact tctaccccaa agacatcaat gtcaagtgga agattgatgg cagtgaacga 480caaaatggcg tcctgaacag ttggactgat caggacagca aagacagcac ctacagcatg 540agcagtaccc tcacgttgac caaggacgag tatgaacgac ataacagcta tacctgtgag 600gccactcaca agacatcaac ttcacccatt gtcaagagct tcaacaggaa tgagtgttag 66025119PRTMus musculus 25Gln Val Thr Leu Lys Glu Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Thr Thr Ser 20 25 30Ala Met Gly Val Ser Trp Val Arg Gln Pro Ser Arg Lys Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Ser Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Thr Ala Thr Tyr Tyr 85 90 95Cys Ala Arg Lys Gly Leu Gly Gly Ala Met Asp Tyr Trp Gly Gln Gly 100 105 110Thr Ser Val Thr Val Ser Ser 11526357DNAMus musculus 26caggttactc tgaaagagtc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctggatt ttcactgacc acttctgcta tgggtgtgag ctgggttcgt 120cagccttcaa gaaagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag caaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gatactgcca catactactg tgctcgaaag 300ggactgggag gtgctatgga ctactggggt caaggaacct cagtcaccgt ctcctca 35727112PRTMus musculus 27Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Leu His Ser 20 25 30Asn Gly Asn Thr Tyr Leu His Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser 85 90 95Thr His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 11028336DNAMus musculus 28gatgttgtga tgacccaaac tccgctctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagccttcta cacagtaatg gaaacaccta tttacattgg 120tacctgcaga agccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tatttctgct ctcaaagtac acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaa 336297PRTMus musculus 29Thr Ser Ala Met Gly Val Ser1 53021DNAMus musculus 30acttctgcta tgggtgtgag c 21319PRTMus musculus 31His Ile Tyr Trp Asp Asp Asp Lys Arg1 53227DNAMus musculus 32cacatttact gggatgatga caagcgc 27339PRTMus musculus 33Lys Gly Leu Gly Gly Ala Met Asp Tyr1 53427DNAMus musculus 34aagggactgg gaggtgctat ggactac 273516PRTMus musculus 35Arg Ser Ser Gln Ser Leu Leu His Ser Asn Gly Asn Thr Tyr Leu His1 5 10 153648DNAMus musculus 36agatctagtc agagccttct acacagtaat ggaaacacct atttacat 48377PRTMus musculus 37Lys Val Ser Asn Arg Phe Ser1 53821DNAMus musculus 38aaagtttcca accgattttc t 21399PRTMus musculus 39Ser Gln Ser Thr His Val Pro Leu Thr1 54027DNAMus musculus 40tctcaaagta cacatgttcc gctcacg 2741447PRTMus musculus 41Gln Val Thr Leu Lys Asp Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Ser Thr Ser 20 25 30Gly Met Gly Val Ser Trp Ile Arg Gln Pro Ser Gly Lys Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Ser Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Ser Ala Thr Tyr Tyr 85 90 95Cys Ser Thr Met Ile Thr Gly Phe Val Tyr Trp Gly Gln Gly Thr Leu 100 105 110Val Thr Val Ser Ala Ala Lys Thr Thr Ala Pro Ser Val Tyr Pro Leu 115 120 125Ala Pro Val Cys Gly Asp Thr Thr Gly Ser Ser Val Ala Leu Gly Cys 130 135 140Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr Leu Thr Trp Asn Ser145 150 155 160Gly Ser Leu Ser Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser 165 170 175Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr Val Thr Ser Ser Thr Trp 180 185 190Pro Ser Gln Ser Ile Thr Cys Asn Val Ala His Pro Ala Ser Ser Thr 195 200 205Lys Val Asp Lys Lys Ile Glu Pro Arg Gly Pro Thr Ile Lys Pro Cys 210 215 220Pro Pro Cys Lys Cys Pro Ala Pro Asn Leu Leu Gly Gly Pro Ser Val225 230 235 240Phe Ile Phe Pro Pro Lys Ile Lys Asp Val Leu Met Ile Ser Leu Ser 245 250 255Pro Ile Val Thr Cys Val Val Val Asp Val Ser Glu Asp Asp Pro Asp 260 265 270Val Gln Ile Ser Trp Phe Val Asn Asn Val Glu Val His Thr Ala Gln 275 280 285Thr Gln Thr His Arg Glu Asp Tyr Asn Ser Thr Leu Arg Val Val Ser 290 295 300Ala Leu Pro Ile Gln His Gln Asp Trp Met Ser Gly Lys Glu Phe Lys305 310 315 320Cys Lys Val Asn Asn Lys Asp Leu Pro Ala Pro Ile Glu Arg Thr Ile 325 330 335Ser Lys Pro Lys Gly Ser Val Arg Ala Pro Gln Val Tyr Val Leu Pro 340 345 350Pro Pro Glu Glu Glu Met Thr Lys Lys Gln Val Thr Leu Thr Cys Met 355 360 365Val Thr Asp Phe Met Pro Glu Asp Ile Tyr Val Glu Trp Thr Asn Asn 370 375 380Gly Lys Thr Glu Leu Asn Tyr Lys Asn Thr Glu Pro Val Leu Asp Ser385 390 395 400Asp Gly Ser Tyr Phe Met Tyr Ser Lys Leu Arg Val Glu Lys Lys Asn 405 410 415Trp Val Glu Arg Asn Ser Tyr Ser Cys Ser Val Val His Glu Gly Leu 420 425 430His Asn His His Thr Thr Lys Ser Phe Ser Arg Thr Pro Gly Lys 435 440 445421344DNAMus musculus 42caggttactc tgaaagactc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctgggtt ttcactgagc acttctggta tgggtgtgag ctggattcgt 120cagccttcag gaaagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag caaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gattctgcca catactactg ttccactatg 300attacggggt ttgtttactg gggccaaggg actctggtca ctgtctctgc agccaaaaca 360acagccccat cggtctatcc cctggcccct gtgtgtggag atacaactgg ctcctcggtg 420gctctaggat gcctggtcaa gggttatttc cctgagccag tgaccttgac ctggaactct 480ggatccctgt ccagtggtgt gcacaccttc ccagctgtcc tgcagtctga cctctacacc 540ctcagcagct cagtgactgt aacctcgagc acctggccca gccagtccat cacctgcaat 600gtggcccacc cggcaagcag caccaaggtg gacaagaaaa ttgagcccag agggcccaca 660atcaagccct gtcctccatg caaatgccca gcacctaacc tcttgggtgg accatccgtc 720ttcatcttcc ctccaaagat caaggatgta ctcatgatct ccctgagccc catagtcaca 780tgtgtggtgg tggatgtgag cgaggatgac ccagatgtcc agatcagctg gtttgtgaac 840aacgtggaag tacacacagc tcagacacaa acccatagag aggattacaa cagtactctc 900cgggtggtca gtgccctccc catccagcac caggactgga tgagtggcaa ggagttcaaa 960tgcaaggtca acaacaaaga cctcccagcg cccatcgaga gaaccatctc aaaacccaaa 1020gggtcagtaa gagctccaca ggtatatgtc ttgcctccac cagaagaaga gatgactaag 1080aaacaggtca ctctgacctg catggtcaca gacttcatgc ctgaagacat ttacgtggag 1140tggaccaaca acgggaaaac agagctaaac tacaagaaca ctgaaccagt cctggactct 1200gatggttctt acttcatgta cagcaagctg agagtggaaa agaagaactg ggtggaaaga 1260aatagctact cctgttcagt ggtccacgag ggtctgcaca atcaccacac gactaagagc 1320ttctcccgga ctccgggtaa atga 134443219PRTMus musculus 43Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 110Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu 115 120 125Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe 130 135 140Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg145 150 155 160Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser 165 170 175Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu 180 185 190Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser 195 200 205Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 21544660DNAMus musculus 44gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agtagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaacggg ctgatgctgc accaactgta 360tccatcttcc caccatccag tgagcagtta acatctggag gtgcctcagt cgtgtgcttc 420ttgaacaact tctaccccaa agacatcaat gtcaagtgga agattgatgg cagtgaacga 480caaaatggcg tcctgaacag ttggactgat caggacagca aagacagcac ctacagcatg 540agcagtaccc tcacgttgac caaggacgag tatgaacgac ataacagcta tacctgtgag 600gccactcaca agacatcaac ttcacccatt gtcaagagct tcaacaggaa tgagtgttag 66045117PRTMus musculus 45Gln Val Thr Leu Lys Asp Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Ser Thr Ser 20 25 30Gly Met Gly Val Ser Trp Ile Arg Gln Pro Ser Gly Lys Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Ser Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Ser Ala Thr Tyr Tyr 85 90 95Cys Ser Thr Met Ile Thr Gly Phe Val Tyr Trp Gly Gln Gly Thr Leu 100 105 110Val Thr Val Ser Ala 11546351DNAMus musculus 46caggttactc tgaaagactc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctgggtt ttcactgagc acttctggta tgggtgtgag ctggattcgt 120cagccttcag gaaagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag caaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gattctgcca catactactg ttccactatg 300attacggggt ttgtttactg gggccaaggg actctggtca ctgtctctgc a 35147112PRTMus musculus 47Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 11048336DNAMus musculus 48gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agtagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaa 336497PRTMus musculus 49Thr Ser Gly Met Gly Val Ser1 55021DNAMus musculus 50acttctggta tgggtgtgag c 21519PRTMus musculus 51His Ile Tyr Trp Asp Asp Asp Lys Arg1 55227DNAMus musculus 52cacatttact gggatgatga caagcgc 27537PRTMus musculus 53Met Ile Thr Gly Phe Val Tyr1 55421DNAMus musculus 54atgattacgg ggtttgttta c 215516PRTMus musculus 55Arg Ser Ser Gln Ser Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu1 5 10 155648DNAMus musculus 56agatctagtc agagcattgt acatagtaat ggaaacacct atttagaa 48577PRTMus musculus 57Lys Val Ser Asn Arg Phe Ser1 55821DNAMus musculus 58aaagtttcca accgattttc t 21599PRTMus musculus 59Phe Gln Gly Ser His Val Pro Leu Thr1 56027DNAMus musculus 60tttcaaggtt cacatgttcc gctcacg 2761445PRTMus musculus 61Asp Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Arg Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Phe 20 25 30Gly Met His Trp Val Arg Gln Ala Pro Glu Lys Gly Leu Glu Trp Val 35 40 45Ala Tyr Ile Ser Ser Gly Ser Ser Thr Ile Tyr Tyr Ala Asp Thr Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Pro Lys Asn Thr Leu Phe65 70 75 80Leu Gln Met Thr Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95Ala Gly Thr Gly Thr Arg Ala Tyr Trp Gly Gln Gly Thr Leu Val Thr 100 105 110Val Ser Ala Ala Lys Thr Thr Ala Pro Ser Val Tyr Pro Leu Ala Pro 115 120 125Val Cys Gly Asp Thr Thr Gly Ser Ser Val Ala Leu Gly Cys Leu Val 130 135 140Lys Gly Tyr Phe Pro Glu Pro Val Thr Leu Thr Trp Asn Ser Gly Ser145 150 155 160Leu Ser Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Asp Leu 165 170 175Tyr Thr Leu Ser Ser Ser Val Thr Val Thr Ser Ser Thr Trp Pro Ser 180 185 190Gln Ser Ile Thr Cys Asn Val Ala His Pro Ala Ser Ser Thr Lys Val 195 200 205Asp Lys Lys Ile Glu Pro Arg Gly Pro Thr Ile Lys Pro Cys Pro Pro 210 215 220Cys Lys Cys Pro Ala Pro Asn Leu Leu Gly Gly Pro Ser Val Phe Ile225 230 235 240Phe Pro Pro Lys Ile Lys Asp Val Leu Met Ile Ser Leu Ser Pro Ile 245 250 255Val Thr Cys Val Val Val Asp Val Ser Glu Asp Asp Pro Asp Val Gln 260 265 270Ile Ser Trp Phe Val Asn Asn Val Glu Val His Thr Ala Gln Thr Gln 275 280 285Thr His Arg Glu Asp Tyr Asn Ser Thr Leu Arg Val Val Ser Ala Leu 290 295 300Pro Ile Gln His Gln Asp Trp Met Ser Gly Lys Glu Phe Lys Cys Lys305 310 315 320Val Asn Asn Lys Asp Leu Pro Ala Pro Ile Glu Arg Thr Ile Ser Lys 325 330 335Pro Lys Gly Ser Val Arg Ala Pro Gln Val Tyr Val Leu Pro Pro Pro 340 345 350Glu Glu Glu Met Thr Lys Lys Gln Val Thr Leu Thr Cys Met Val Thr 355 360 365Asp Phe Met Pro Glu Asp Ile Tyr Val Glu Trp Thr Asn Asn Gly Lys 370 375 380Thr Glu Leu Asn Tyr Lys Asn Thr

Glu Pro Val Leu Asp Ser Asp Gly385 390 395 400Ser Tyr Phe Met Tyr Ser Lys Leu Arg Val Glu Lys Lys Asn Trp Val 405 410 415Glu Arg Asn Ser Tyr Ser Cys Ser Val Val His Glu Gly Leu His Asn 420 425 430His His Thr Thr Lys Ser Phe Ser Arg Thr Pro Gly Lys 435 440 445621338DNAMus musculus 62gatgtgcagc tggtggagtc tgggggaggc ttagtgcagc ctggagggtc ccggaaactc 60tcctgtgcag cctctggatt cactttcagt agctttggaa tgcactgggt tcgtcaggct 120ccagagaagg ggctggagtg ggtcgcatac attagtagtg gcagtagtac catctactat 180gcagacacag tgaagggccg attcaccatc tccagagaca atcccaagaa caccctgttc 240ctgcaaatga ccagtctaag gtctgaggac acggccatgt attactgtgc cgggactggg 300acgagagctt actggggcca agggactctg gtcactgtct ctgcagccaa aacaacagcc 360ccatcggtct atcccctggc ccctgtgtgt ggagatacaa ctggctcctc ggtggctcta 420ggatgcctgg tcaagggtta tttccctgag ccagtgacct tgacctggaa ctctggatcc 480ctgtccagtg gtgtgcacac cttcccagct gtcctgcagt ctgacctcta caccctcagc 540agctcagtga ctgtaacctc gagcacctgg cccagccagt ccatcacctg caatgtggcc 600cacccggcaa gcagcaccaa ggtggacaag aaaattgagc ccagagggcc cacaatcaag 660ccctgtcctc catgcaaatg cccagcacct aacctcttgg gtggaccatc cgtcttcatc 720ttccctccaa agatcaagga tgtactcatg atctccctga gccccatagt cacatgtgtg 780gtggtggatg tgagcgagga tgacccagat gtccagatca gctggtttgt gaacaacgtg 840gaagtacaca cagctcagac acaaacccat agagaggatt acaacagtac tctccgggtg 900gtcagtgccc tccccatcca gcaccaggac tggatgagtg gcaaggagtt caaatgcaag 960gtcaacaaca aagacctccc agcgcccatc gagagaacca tctcaaaacc caaagggtca 1020gtaagagctc cacaggtata tgtcttgcct ccaccagaag aagagatgac taagaaacag 1080gtcactctga cctgcatggt cacagacttc atgcctgaag acatttacgt ggagtggacc 1140aacaacggga aaacagagct aaactacaag aacactgaac cagtcctgga ctctgatggt 1200tcttacttca tgtacagcaa gctgagagtg gaaaagaaga actgggtgga aagaaatagc 1260tactcctgtt cagtggtcca cgagggtctg cacaatcacc acacgactaa gagcttctcc 1320cggactccgg gtaaatga 133863219PRTMus musculus 63Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Pro Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys 100 105 110Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu 115 120 125Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe 130 135 140Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg145 150 155 160Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser 165 170 175Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu 180 185 190Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser 195 200 205Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 21564660DNAMus musculus 64gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctcctaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttcct 300ccgacgttcg gtggaggcac caagctggaa atcaaacggg ctgatgctgc accaactgta 360tccatcttcc caccatccag tgagcagtta acatctggag gtgcctcagt cgtgtgcttc 420ttgaacaact tctaccccaa agacatcaat gtcaagtgga agattgatgg cagtgaacga 480caaaatggcg tcctgaacag ttggactgat caggacagca aagacagcac ctacagcatg 540agcagtaccc tcacgttgac caaggacgag tatgaacgac ataacagcta tacctgtgag 600gccactcaca agacatcaac ttcacccatt gtcaagagct tcaacaggaa tgagtgttag 66065115PRTMus musculus 65Asp Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly1 5 10 15Ser Arg Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Ser Phe 20 25 30Gly Met His Trp Val Arg Gln Ala Pro Glu Lys Gly Leu Glu Trp Val 35 40 45Ala Tyr Ile Ser Ser Gly Ser Ser Thr Ile Tyr Tyr Ala Asp Thr Val 50 55 60Lys Gly Arg Phe Thr Ile Ser Arg Asp Asn Pro Lys Asn Thr Leu Phe65 70 75 80Leu Gln Met Thr Ser Leu Arg Ser Glu Asp Thr Ala Met Tyr Tyr Cys 85 90 95Ala Gly Thr Gly Thr Arg Ala Tyr Trp Gly Gln Gly Thr Leu Val Thr 100 105 110Val Ser Ala 11566345DNAMus musculus 66gatgtgcagc tggtggagtc tgggggaggc ttagtgcagc ctggagggtc ccggaaactc 60tcctgtgcag cctctggatt cactttcagt agctttggaa tgcactgggt tcgtcaggct 120ccagagaagg ggctggagtg ggtcgcatac attagtagtg gcagtagtac catctactat 180gcagacacag tgaagggccg attcaccatc tccagagaca atcccaagaa caccctgttc 240ctgcaaatga ccagtctaag gtctgaggac acggccatgt attactgtgc cgggactggg 300acgagagctt actggggcca agggactctg gtcactgtct ctgca 34567112PRTMus musculus 67Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Pro Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys 100 105 11068336DNAMus musculus 68gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctcctaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttcct 300ccgacgttcg gtggaggcac caagctggaa atcaaa 336695PRTMus musculus 69Ser Phe Gly Met His1 57015DNAMus musculus 70agctttggaa tgcac 157110PRTMus musculus 71Tyr Ile Ser Ser Gly Ser Ser Thr Ile Tyr1 5 107230DNAMus musculus 72tacattagta gtggcagtag taccatctac 30736PRTMus musculus 73Thr Gly Thr Arg Ala Tyr1 57418DNAMus musculus 74actgggacga gagcttac 187516PRTMus musculus 75Arg Ser Ser Gln Ser Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu1 5 10 157648DNAMus musculus 76agatctagtc agagcattgt acatagtaat ggaaacacct atttagaa 48777PRTMus musculus 77Lys Val Ser Asn Arg Phe Ser1 57821DNAMus musculus 78aaagtttcca accgattttc t 21799PRTMus musculus 79Phe Gln Gly Ser His Val Pro Pro Thr1 58027DNAMus musculus 80tttcaaggtt cacatgttcc tccgacg 2781442PRTMus musculus 81Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Lys Pro Gly Ala1 5 10 15Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr 20 25 30Trp Met His Trp Val Lys Leu Arg Pro Gly Gln Gly Phe Glu Trp Ile 35 40 45Gly Glu Ile Asn Pro Arg Asn Gly Gly Thr Asn Asn Asn Glu Asn Phe 50 55 60Lys Arg Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Ser Thr Ala Tyr65 70 75 80Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys 85 90 95Thr Arg Asp Gly Asn Tyr Asp Pro Phe Ala Tyr Trp Gly Gln Gly Thr 100 105 110Leu Val Thr Val Ser Ala Ala Lys Thr Thr Pro Pro Ser Val Tyr Pro 115 120 125Leu Ala Pro Gly Ser Ala Ala Gln Thr Asn Ser Met Val Thr Leu Gly 130 135 140Cys Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr Val Thr Trp Asn145 150 155 160Ser Gly Ser Leu Ser Ser Gly Val His Thr Phe Pro Ala Val Leu Gln 165 170 175Ser Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr Val Pro Ser Ser Thr 180 185 190Trp Pro Ser Glu Thr Val Thr Cys Asn Val Ala His Pro Ala Ser Ser 195 200 205Thr Lys Val Asp Lys Lys Ile Val Pro Arg Asp Cys Gly Cys Lys Pro 210 215 220Cys Ile Cys Thr Val Pro Glu Val Ser Ser Val Phe Ile Phe Pro Pro225 230 235 240Lys Pro Lys Asp Val Leu Thr Ile Thr Leu Thr Pro Lys Val Thr Cys 245 250 255Val Val Val Asp Ile Ser Lys Asp Asp Pro Glu Val Gln Phe Ser Trp 260 265 270Phe Val Asp Asp Val Glu Val His Thr Ala Gln Thr Gln Pro Arg Glu 275 280 285Glu Gln Phe Asn Ser Thr Phe Arg Ser Val Ser Glu Leu Pro Ile Met 290 295 300His Gln Asp Trp Leu Asn Gly Lys Glu Phe Lys Cys Arg Val Asn Ser305 310 315 320Ala Ala Phe Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly 325 330 335Arg Pro Lys Ala Pro Gln Val Tyr Thr Ile Pro Pro Pro Lys Glu Gln 340 345 350Met Ala Lys Asp Lys Val Ser Leu Thr Cys Met Ile Thr Asp Phe Phe 355 360 365Pro Glu Asp Ile Thr Val Glu Trp Gln Trp Asn Gly Gln Pro Ala Glu 370 375 380Asn Tyr Lys Asn Thr Gln Pro Ile Met Asp Thr Asp Gly Ser Tyr Phe385 390 395 400Val Tyr Ser Lys Leu Asn Val Gln Lys Ser Asn Trp Glu Ala Gly Asn 405 410 415Thr Phe Thr Cys Ser Val Leu His Glu Gly Leu Leu Asn Leu His Thr 420 425 430Glu Lys Ser Leu Ser Leu Ser Pro Gly Lys 435 440821329DNAMus musculus 82caggtccaac tccagcagcc tggggctgaa ctggtgaagc ctggggcttc agtgaagttg 60tcctgcaagg cttctggcta caccttcacc agctactgga tgcactgggt gaagctgagg 120cctggacaag gctttgagtg gattggagag attaatccta gaaatggtgg tactaacaac 180aatgagaact tcaagagaaa ggccacactg actgtagaca aatcctccag cacagcctac 240atgcaactca gcagcctgac atctgaggac tctgcggtct attactgtac aagagatggt 300aactacgacc cctttgctta ctggggccaa gggactctgg tcactgtctc tgcagccaaa 360acgacacccc catctgtcta tccactggcc cctggatctg ctgcccaaac taactccatg 420gtgaccctgg gatgcctggt caagggctat ttccctgagc cagtgacagt gacctggaac 480tctggatccc tgtccagcgg tgtgcacacc ttcccagctg tcctgcagtc tgacctctac 540actctgagca gctcagtgac tgtcccctcc agcacctggc ccagcgagac cgtcacctgc 600aacgttgccc acccggccag cagcaccaag gtggacaaga aaattgtgcc cagggattgt 660ggttgtaagc cttgcatatg tacagtccca gaagtatcat ctgtcttcat cttcccccca 720aagcccaagg atgtgctcac cattactctg actcctaagg tcacgtgtgt tgtggtagac 780atcagcaagg atgatcccga ggtccagttc agctggtttg tagatgatgt ggaggtgcac 840acagctcaga cgcaaccccg ggaggagcag ttcaacagca ctttccgctc agtcagtgaa 900cttcccatca tgcaccagga ctggctcaat ggcaaggagt tcaaatgcag ggtcaacagt 960gcagctttcc ctgcccccat cgagaaaacc atctccaaaa ccaaaggcag accgaaggct 1020ccacaggtgt acaccattcc acctcccaag gagcagatgg ccaaggataa agtcagtctg 1080acctgcatga taacagactt cttccctgaa gacattactg tggagtggca gtggaatggg 1140cagccagcgg agaactacaa gaacactcag cccatcatgg acacagatgg ctcttacttc 1200gtttacagca agctcaatgt gcagaagagc aactgggagg caggaaatac tttcacctgc 1260tctgtgttac atgagggcct gctcaacctc catactgaga agagcctctc cctctctcct 1320ggtaaatga 132983220PRTMus musculus 83Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu His Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser 85 90 95Thr His Val Pro Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile 100 105 110Lys Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser 115 120 125Glu Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn 130 135 140Phe Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu145 150 155 160Arg Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp 165 170 175Ser Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr 180 185 190Glu Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr 195 200 205Ser Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 215 22084663DNAMus musculus 84gatgttgtga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagccttgta cacagtaatg gaaacaccta tttacattgg 120tacctgcaga agccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tatttctgct ctcaaagtac acatgttcct 300ccgtacacgt tcggaggggg gaccaagctg gaaataaaac gggctgatgc tgcaccaact 360gtatccatct tcccaccatc cagtgagcag ttaacatctg gaggtgcctc agtcgtgtgc 420ttcttgaaca acttctaccc caaagacatc aatgtcaagt ggaagattga tggcagtgaa 480cgacaaaatg gcgtcctgaa cagttggact gatcaggaca gcaaagacag cacctacagc 540atgagcagta ccctcacgtt gaccaaggac gagtatgaac gacataacag ctatacctgt 600gaggccactc acaagacatc aacttcaccc attgtcaaga gcttcaacag gaatgagtgt 660tag 66385118PRTMus musculus 85Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Lys Pro Gly Ala1 5 10 15Ser Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr 20 25 30Trp Met His Trp Val Lys Leu Arg Pro Gly Gln Gly Phe Glu Trp Ile 35 40 45Gly Glu Ile Asn Pro Arg Asn Gly Gly Thr Asn Asn Asn Glu Asn Phe 50 55 60Lys Arg Lys Ala Thr Leu Thr Val Asp Lys Ser Ser Ser Thr Ala Tyr65 70 75 80Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys 85 90 95Thr Arg Asp Gly Asn Tyr Asp Pro Phe Ala Tyr Trp Gly Gln Gly Thr 100 105 110Leu Val Thr Val Ser Ala 11586354DNAMus musculus 86caggtccaac tccagcagcc tggggctgaa ctggtgaagc ctggggcttc agtgaagttg 60tcctgcaagg cttctggcta caccttcacc agctactgga tgcactgggt gaagctgagg 120cctggacaag gctttgagtg gattggagag attaatccta gaaatggtgg tactaacaac 180aatgagaact tcaagagaaa ggccacactg actgtagaca aatcctccag cacagcctac 240atgcaactca gcagcctgac atctgaggac tctgcggtct attactgtac aagagatggt 300aactacgacc cctttgctta ctggggccaa gggactctgg tcactgtctc tgca 35487113PRTMus musculus 87Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu His Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser 85 90 95Thr His Val Pro Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile 100 105 110Lys88339DNAMus musculus 88gatgttgtga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagccttgta cacagtaatg gaaacaccta tttacattgg 120tacctgcaga agccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc

cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tatttctgct ctcaaagtac acatgttcct 300ccgtacacgt tcggaggggg gaccaagctg gaaataaaa 339895PRTMus musculus 89Ser Tyr Trp Met His1 59015DNAMus musculus 90agctactgga tgcac 159110PRTMus musculus 91Glu Ile Asn Pro Arg Asn Gly Gly Thr Asn1 5 109230DNAMus musculus 92gagattaatc ctagaaatgg tggtactaac 30939PRTMus musculus 93Asp Gly Asn Tyr Asp Pro Phe Ala Tyr1 59427DNAMus musculus 94gatggtaact acgacccctt tgcttac 279516PRTMus musculus 95Arg Ser Ser Gln Ser Leu Val His Ser Asn Gly Asn Thr Tyr Leu His1 5 10 159648DNAMus musculus 96agatctagtc agagccttgt acacagtaat ggaaacacct atttacat 48977PRTMus musculus 97Lys Val Ser Asn Arg Phe Ser1 59821DNAMus musculus 98aaagtttcca accgattttc t 219910PRTMus musculus 99Ser Gln Ser Thr His Val Pro Pro Tyr Thr1 5 1010030DNAMus musculus 100tctcaaagta cacatgttcc tccgtacacg 30101453PRTMus musculus 101Gln Val Thr Leu Lys Glu Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Ser Thr Ser 20 25 30Gly Met Gly Val Ser Trp Ile Arg Gln Pro Ser Gly Glu Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Arg Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Thr Ala Thr Tyr Tyr 85 90 95Cys Gly Arg Tyr Arg Tyr Gly Phe Ala Tyr Trp Gly Gln Gly Thr Leu 100 105 110Val Thr Val Ser Ala Ala Lys Thr Thr Pro Pro Ser Val Tyr Pro Leu 115 120 125Ala Pro Gly Cys Gly Asp Thr Thr Gly Ser Ser Val Thr Leu Gly Cys 130 135 140Leu Val Lys Gly Tyr Phe Pro Glu Ser Val Thr Val Thr Trp Asn Ser145 150 155 160Gly Ser Leu Ser Ser Ser Val His Thr Phe Pro Ala Leu Leu Gln Ser 165 170 175Gly Leu Tyr Thr Met Ser Ser Ser Val Thr Val Pro Ser Ser Thr Trp 180 185 190Pro Ser Gln Thr Val Thr Cys Ser Val Ala His Pro Ala Ser Ser Thr 195 200 205Thr Val Asp Lys Lys Leu Glu Pro Ser Gly Pro Ile Ser Thr Ile Asn 210 215 220Pro Cys Pro Pro Cys Lys Glu Cys His Lys Cys Pro Ala Pro Asn Leu225 230 235 240Glu Gly Gly Pro Ser Val Phe Ile Phe Pro Pro Asn Ile Lys Asp Val 245 250 255Leu Met Ile Ser Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Val 260 265 270Ser Glu Asp Asp Pro Asp Val Gln Ile Ser Trp Phe Val Asn Asn Val 275 280 285Glu Val His Thr Ala Gln Thr Gln Thr His Arg Glu Asp Tyr Asn Ser 290 295 300Thr Ile Arg Val Val Ser Thr Leu Pro Ile Gln His Gln Asp Trp Met305 310 315 320Ser Gly Lys Glu Phe Lys Cys Lys Val Asn Asn Lys Asp Leu Pro Ser 325 330 335Pro Ile Glu Arg Thr Ile Ser Lys Ile Lys Gly Leu Val Arg Ala Pro 340 345 350Gln Val Tyr Ile Leu Pro Pro Pro Ala Glu Gln Leu Ser Arg Lys Asp 355 360 365Val Ser Leu Thr Cys Leu Val Val Gly Phe Asn Pro Gly Asp Ile Ser 370 375 380Val Glu Trp Thr Ser Asn Gly His Thr Glu Glu Asn Tyr Lys Asp Thr385 390 395 400Ala Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe Ile Tyr Ser Lys Leu 405 410 415Asn Met Lys Thr Ser Lys Trp Glu Lys Thr Asp Ser Phe Ser Cys Asn 420 425 430Val Arg His Glu Gly Leu Lys Asn Tyr Tyr Leu Lys Lys Thr Ile Ser 435 440 445Arg Ser Pro Gly Lys 4501021362DNAMus musculus 102caggttactc tgaaagagtc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctgggtt ttcactgagc acttctggta tgggtgtgag ctggattcgt 120cagccttcag gagagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag aaaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gatactgcca catactactg tggtcgatat 300aggtacggct ttgcttactg gggccaaggg actctggtca ctgtctctgc agccaaaaca 360acacccccat cagtctatcc actggcccct gggtgtggag atacaactgg ttcctccgtg 420actctgggat gcctggtcaa gggctacttc cctgagtcag tgactgtgac ttggaactct 480ggatccctgt ccagcagtgt gcacaccttc ccagctctcc tgcagtctgg actctacact 540atgagcagct cagtgactgt cccctccagc acctggccaa gtcagaccgt cacctgcagc 600gttgctcacc cagccagcag caccacggtg gacaaaaaac ttgagcccag cgggcccatt 660tcaacaatca acccctgtcc tccatgcaag gagtgtcaca aatgcccagc tcctaacctc 720gagggtggac catccgtctt catcttccct ccaaatatca aggatgtact catgatctcc 780ctgacaccca aggtcacgtg tgtggtggtg gatgtgagcg aggatgaccc agacgtccag 840atcagctggt ttgtgaacaa cgtggaagta cacacagctc agacacaaac ccatagagag 900gattacaaca gtactatccg ggtggtcagc accctcccca tccagcacca ggactggatg 960agtggcaagg agttcaaatg caaggtcaac aacaaagacc tcccatcacc catcgagaga 1020accatctcaa aaattaaagg gctagtcaga gctccacaag tatacatctt gccgccacca 1080gcagagcagt tgtccaggaa agatgtcagt ctcacttgcc tggtcgtggg cttcaaccct 1140ggagacatca gtgtggagtg gaccagcaat gggcatacag aggagaacta caaggacacc 1200gcaccagtcc tggactctga cggttcttac ttcatatata gcaagctcaa tatgaaaaca 1260agcaagtggg agaaaacaga ttccttctca tgcaacgtga gacacgaggg tctgaaaaat 1320tactacctga agaagaccat ctcccggtct ccgggtaaat ga 1362103219PRTMus musculus 103Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 110Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu 115 120 125Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe 130 135 140Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg145 150 155 160Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser 165 170 175Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu 180 185 190Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser 195 200 205Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys 210 215104660DNAMus musculus 104gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaacggg ctgatgctgc accaactgta 360tccatcttcc caccatccag tgagcagtta acatctggag gtgcctcagt cgtgtgcttc 420ttgaacaact tctaccccaa agacatcaat gtcaagtgga agattgatgg cagtgaacga 480caaaatggcg tcctgaacag ttggactgat caggacagca aagacagcac ctacagcatg 540agcagtaccc tcacgttgac caaggacgag tatgaacgac ataacagcta tacctgtgag 600gccactcaca agacatcaac ttcacccatt gtcaagagct tcaacaggaa tgagtgttag 660105116PRTMus musculus 105Gln Val Thr Leu Lys Glu Ser Gly Pro Gly Ile Leu Gln Pro Ser Gln1 5 10 15Thr Leu Ser Leu Thr Cys Ser Phe Ser Gly Phe Ser Leu Ser Thr Ser 20 25 30Gly Met Gly Val Ser Trp Ile Arg Gln Pro Ser Gly Glu Gly Leu Glu 35 40 45Trp Leu Ala His Ile Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Ser 50 55 60Leu Lys Ser Arg Leu Thr Ile Ser Lys Asp Thr Ser Arg Asn Gln Val65 70 75 80Phe Leu Lys Ile Thr Ser Val Asp Thr Ala Asp Thr Ala Thr Tyr Tyr 85 90 95Cys Gly Arg Tyr Arg Tyr Gly Phe Ala Tyr Trp Gly Gln Gly Thr Leu 100 105 110Val Thr Val Ser 115106348DNAMus musculus 106caggttactc tgaaagagtc tggccctggg atattgcagc cctcccagac cctcagtctg 60acttgttctt tctctgggtt ttcactgagc acttctggta tgggtgtgag ctggattcgt 120cagccttcag gagagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180tataacccat ccctgaagag ccggctcaca atctccaagg atacctccag aaaccaggta 240ttcctcaaga tcaccagtgt ggacactgca gatactgcca catactactg tggtcgatat 300aggtacggct ttgcttactg gggccaaggg actctggtca ctgtctct 348107112PRTMus musculus 107Asp Val Leu Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly1 5 10 15Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser 20 25 30Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser 35 40 45Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro 50 55 60Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile65 70 75 80Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Phe Gln Gly 85 90 95Ser His Val Pro Leu Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys 100 105 110108336DNAMus musculus 108gatgttttga tgacccaaac tccactctcc ctgcctgtca gtcttggaga tcaagcctcc 60atctcttgca gatctagtca gagcattgta catagtaatg gaaacaccta tttagaatgg 120tacctgcaga aaccaggcca gtctccaaag ctcctgatct acaaagtttc caaccgattt 180tctggggtcc cagacaggtt cagtggcagt ggatcaggga cagatttcac actcaagatc 240agcagagtgg aggctgagga tctgggagtt tattactgct ttcaaggttc acatgttccg 300ctcacgttcg gtgctgggac caagctggag ctgaaa 3361097PRTMus musculus 109Thr Ser Gly Met Gly Val Ser1 511021DNAMus musculus 110acttctggta tgggtgtgag c 211119PRTMus musculus 111His Ile Tyr Trp Asp Asp Asp Lys Arg1 511227DNAMus musculus 112cacatttact gggatgatga caagcgc 271137PRTMus musculus 113Tyr Arg Tyr Gly Phe Ala Tyr1 511421DNAMus musculus 114tataggtacg gctttgctta c 2111516PRTMus musculus 115Arg Ser Ser Gln Ser Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu1 5 10 1511648DNAMus musculus 116agatctagtc agagcattgt acatagtaat ggaaacacct atttagaa 481177PRTMus musculus 117Lys Val Ser Asn Arg Phe Ser1 511821DNAMus musculus 118aaagtttcca accgattttc t 211199PRTMus musculus 119Phe Gln Gly Ser His Val Pro Leu Thr1 512027DNAMus musculus 120tttcaaggtt cacatgttcc gctcacg 27

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed