NZ618914B2 - Anti-alpha synuclein binding molecules - Google Patents
Anti-alpha synuclein binding molecules Download PDFInfo
- Publication number
- NZ618914B2 NZ618914B2 NZ618914A NZ61891412A NZ618914B2 NZ 618914 B2 NZ618914 B2 NZ 618914B2 NZ 618914 A NZ618914 A NZ 618914A NZ 61891412 A NZ61891412 A NZ 61891412A NZ 618914 B2 NZ618914 B2 NZ 618914B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- antibody
- seq
- antigen
- synuclein
- amino acid
- Prior art date
Links
- 230000027455 binding Effects 0.000 title claims abstract description 336
- 102000019355 Synucleins Human genes 0.000 title description 43
- 108050006783 Synucleins Proteins 0.000 title description 43
- 102000004965 antibodies Human genes 0.000 claims abstract description 589
- 108090001123 antibodies Proteins 0.000 claims abstract description 589
- 239000000427 antigen Substances 0.000 claims abstract description 154
- 108091007172 antigens Proteins 0.000 claims abstract description 150
- 102000038129 antigens Human genes 0.000 claims abstract description 150
- 125000003275 alpha amino acid group Chemical group 0.000 claims abstract description 124
- 150000001413 amino acids Chemical class 0.000 claims abstract description 104
- 210000004027 cells Anatomy 0.000 claims description 137
- 229920001184 polypeptide Polymers 0.000 claims description 134
- 229920000023 polynucleotide Polymers 0.000 claims description 83
- 239000002157 polynucleotide Substances 0.000 claims description 82
- 102000004169 proteins and genes Human genes 0.000 claims description 73
- 108090000623 proteins and genes Proteins 0.000 claims description 73
- 201000010099 disease Diseases 0.000 claims description 61
- 239000000203 mixture Substances 0.000 claims description 56
- 150000007523 nucleic acids Chemical class 0.000 claims description 42
- 108020004707 nucleic acids Proteins 0.000 claims description 40
- 206010061536 Parkinson's disease Diseases 0.000 claims description 33
- 239000003814 drug Substances 0.000 claims description 26
- 230000004807 localization Effects 0.000 claims description 22
- 206010067889 Dementia with Lewy body Diseases 0.000 claims description 20
- 201000002832 Lewy body dementia Diseases 0.000 claims description 19
- 102000003802 alpha-Synuclein Human genes 0.000 claims description 19
- 108090000185 alpha-Synuclein Proteins 0.000 claims description 19
- 238000004519 manufacturing process Methods 0.000 claims description 17
- 102000004190 Enzymes Human genes 0.000 claims description 16
- 108090000790 Enzymes Proteins 0.000 claims description 16
- 239000000969 carrier Substances 0.000 claims description 13
- 230000035693 Fab Effects 0.000 claims description 12
- 241000700605 Viruses Species 0.000 claims description 11
- 239000003795 chemical substances by application Substances 0.000 claims description 10
- 238000003384 imaging method Methods 0.000 claims description 8
- 108010070144 Single-Chain Antibodies Proteins 0.000 claims description 6
- 150000002632 lipids Chemical class 0.000 claims description 6
- 230000004044 response Effects 0.000 claims description 6
- 102000005632 Single-Chain Antibodies Human genes 0.000 claims description 5
- 229920001223 polyethylene glycol Polymers 0.000 claims description 5
- 241000124008 Mammalia Species 0.000 claims description 4
- 230000003287 optical Effects 0.000 claims description 2
- 239000008177 pharmaceutical agent Substances 0.000 claims description 2
- 238000002600 positron emission tomography Methods 0.000 claims description 2
- 238000002603 single-photon emission computed tomography Methods 0.000 claims description 2
- 238000003325 tomography Methods 0.000 claims description 2
- 240000006240 Linum usitatissimum Species 0.000 claims 2
- 238000002595 magnetic resonance imaging Methods 0.000 claims 1
- 239000003607 modifier Substances 0.000 claims 1
- 235000001014 amino acid Nutrition 0.000 description 118
- 230000014509 gene expression Effects 0.000 description 69
- 235000018102 proteins Nutrition 0.000 description 68
- 238000000034 method Methods 0.000 description 59
- 102000018358 Immunoglobulins Human genes 0.000 description 54
- 108060003951 Immunoglobulins Proteins 0.000 description 54
- 238000006467 substitution reaction Methods 0.000 description 41
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 39
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 37
- 150000002500 ions Chemical class 0.000 description 32
- 108010045030 monoclonal antibodies Proteins 0.000 description 28
- 102000005614 monoclonal antibodies Human genes 0.000 description 28
- 229920001405 Coding region Polymers 0.000 description 24
- 102000004196 processed proteins & peptides Human genes 0.000 description 24
- 108090000765 processed proteins & peptides Proteins 0.000 description 24
- 230000004048 modification Effects 0.000 description 23
- 238000006011 modification reaction Methods 0.000 description 23
- 239000000047 product Substances 0.000 description 23
- 241000699666 Mus <mouse, genus> Species 0.000 description 22
- 238000004166 bioassay Methods 0.000 description 21
- 230000001225 therapeutic Effects 0.000 description 21
- 229920000160 (ribonucleotides)n+m Polymers 0.000 description 19
- 210000004408 Hybridomas Anatomy 0.000 description 17
- 239000000523 sample Substances 0.000 description 17
- 229920001850 Nucleic acid sequence Polymers 0.000 description 16
- 230000035897 transcription Effects 0.000 description 16
- 210000004556 Brain Anatomy 0.000 description 15
- 108090001095 Immunoglobulin G Proteins 0.000 description 15
- 102000004851 Immunoglobulin G Human genes 0.000 description 15
- 108020001507 fusion proteins Proteins 0.000 description 15
- 102000037240 fusion proteins Human genes 0.000 description 15
- 230000035772 mutation Effects 0.000 description 15
- 108020004999 Messenger RNA Proteins 0.000 description 14
- 230000000694 effects Effects 0.000 description 14
- 229920002106 messenger RNA Polymers 0.000 description 14
- 238000003127 radioimmunoassay Methods 0.000 description 14
- 108060001277 CDR2 Proteins 0.000 description 13
- 102100008744 CDR2 Human genes 0.000 description 13
- 101700027814 CDR3 Proteins 0.000 description 13
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 13
- 230000015572 biosynthetic process Effects 0.000 description 13
- 229960000060 monoclonal antibodies Drugs 0.000 description 13
- 210000001519 tissues Anatomy 0.000 description 13
- 210000003719 B-Lymphocytes Anatomy 0.000 description 12
- 239000003550 marker Substances 0.000 description 12
- 239000000463 material Substances 0.000 description 12
- 238000000746 purification Methods 0.000 description 12
- 239000000243 solution Substances 0.000 description 12
- 241000588724 Escherichia coli Species 0.000 description 11
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 11
- 108091006028 chimera Proteins 0.000 description 11
- 230000000295 complement Effects 0.000 description 11
- 238000001514 detection method Methods 0.000 description 11
- 230000004927 fusion Effects 0.000 description 11
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 11
- 229920002676 Complementary DNA Polymers 0.000 description 10
- 102000006496 Immunoglobulin Heavy Chains Human genes 0.000 description 10
- 108010019476 Immunoglobulin Heavy Chains Proteins 0.000 description 10
- 238000004113 cell culture Methods 0.000 description 10
- VYFYYTLLBUKUHU-UHFFFAOYSA-N dopamine Chemical compound NCCC1=CC=C(O)C(O)=C1 VYFYYTLLBUKUHU-UHFFFAOYSA-N 0.000 description 10
- 229940079593 drugs Drugs 0.000 description 10
- 230000002068 genetic Effects 0.000 description 10
- 210000004602 germ cell Anatomy 0.000 description 10
- 238000000338 in vitro Methods 0.000 description 10
- 102000005962 receptors Human genes 0.000 description 10
- 108020003175 receptors Proteins 0.000 description 10
- 229940088598 Enzyme Drugs 0.000 description 9
- 101710009640 IGKV1D-8 Proteins 0.000 description 9
- 101710024481 IGKV2-29 Proteins 0.000 description 9
- 101710009635 IGKV2D-26 Proteins 0.000 description 9
- 102100005978 IGKV3-20 Human genes 0.000 description 9
- 101710024987 IGKV3-20 Proteins 0.000 description 9
- 101710009651 IGKV3D-15 Proteins 0.000 description 9
- 239000002299 complementary DNA Substances 0.000 description 9
- 238000005516 engineering process Methods 0.000 description 9
- 230000002708 enhancing Effects 0.000 description 9
- -1 polyA+ RNA Chemical class 0.000 description 9
- 241000894007 species Species 0.000 description 9
- 239000000126 substance Substances 0.000 description 9
- NFGXHKASABOEEW-UHFFFAOYSA-N (+)-methoprene Chemical compound COC(C)(C)CCCC(C)CC=CC(C)=CC(=O)OC(C)C NFGXHKASABOEEW-UHFFFAOYSA-N 0.000 description 8
- 210000004369 Blood Anatomy 0.000 description 8
- 102100008989 IGHV1-2 Human genes 0.000 description 8
- 101710003461 IGHV1-2 Proteins 0.000 description 8
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 8
- 235000014680 Saccharomyces cerevisiae Nutrition 0.000 description 8
- 238000007792 addition Methods 0.000 description 8
- 239000008280 blood Substances 0.000 description 8
- 238000004587 chromatography analysis Methods 0.000 description 8
- 238000010367 cloning Methods 0.000 description 8
- 150000001875 compounds Chemical class 0.000 description 8
- 238000005755 formation reaction Methods 0.000 description 8
- 230000001965 increased Effects 0.000 description 8
- 210000004962 mammalian cells Anatomy 0.000 description 8
- 239000002609 media Substances 0.000 description 8
- 230000036499 Half live Effects 0.000 description 7
- 210000002381 Plasma Anatomy 0.000 description 7
- 108020004511 Recombinant DNA Proteins 0.000 description 7
- 125000000539 amino acid group Chemical group 0.000 description 7
- 238000004458 analytical method Methods 0.000 description 7
- 238000003776 cleavage reaction Methods 0.000 description 7
- 239000011248 coating agent Substances 0.000 description 7
- 238000000576 coating method Methods 0.000 description 7
- 230000000875 corresponding Effects 0.000 description 7
- 230000003899 glycosylation Effects 0.000 description 7
- 238000006206 glycosylation reaction Methods 0.000 description 7
- 239000012528 membrane Substances 0.000 description 7
- 210000001806 memory B lymphocyte Anatomy 0.000 description 7
- 229910052751 metal Inorganic materials 0.000 description 7
- 239000002184 metal Substances 0.000 description 7
- 125000003729 nucleotide group Chemical group 0.000 description 7
- 230000001575 pathological Effects 0.000 description 7
- 238000003259 recombinant expression Methods 0.000 description 7
- 239000011780 sodium chloride Substances 0.000 description 7
- 239000007790 solid phase Substances 0.000 description 7
- 238000003786 synthesis reaction Methods 0.000 description 7
- 229940098773 Bovine Serum Albumin Drugs 0.000 description 6
- 108091003117 Bovine Serum Albumin Proteins 0.000 description 6
- 210000004558 Lewy Bodies Anatomy 0.000 description 6
- 210000002966 Serum Anatomy 0.000 description 6
- 229940035295 Ting Drugs 0.000 description 6
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 6
- 201000011510 cancer Diseases 0.000 description 6
- 229920002678 cellulose Polymers 0.000 description 6
- 238000002649 immunization Methods 0.000 description 6
- 231100000350 mutagenesis Toxicity 0.000 description 6
- 210000002569 neurons Anatomy 0.000 description 6
- 239000002773 nucleotide Substances 0.000 description 6
- 239000008194 pharmaceutical composition Substances 0.000 description 6
- 230000002797 proteolythic Effects 0.000 description 6
- 238000010186 staining Methods 0.000 description 6
- 206010001897 Alzheimer's disease Diseases 0.000 description 5
- 241000894006 Bacteria Species 0.000 description 5
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 5
- 108020004684 Internal Ribosome Entry Sites Proteins 0.000 description 5
- 231100000765 Toxin Toxicity 0.000 description 5
- 238000004220 aggregation Methods 0.000 description 5
- 230000002776 aggregation Effects 0.000 description 5
- 230000004075 alteration Effects 0.000 description 5
- 239000001913 cellulose Substances 0.000 description 5
- 235000010980 cellulose Nutrition 0.000 description 5
- 239000003153 chemical reaction reagent Substances 0.000 description 5
- 239000003636 conditioned culture media Substances 0.000 description 5
- 229960003638 dopamine Drugs 0.000 description 5
- 239000003623 enhancer Substances 0.000 description 5
- 210000003527 eukaryotic cell Anatomy 0.000 description 5
- 238000002474 experimental method Methods 0.000 description 5
- 230000036541 health Effects 0.000 description 5
- 230000028993 immune response Effects 0.000 description 5
- 230000003053 immunization Effects 0.000 description 5
- 238000003018 immunoassay Methods 0.000 description 5
- 238000003780 insertion Methods 0.000 description 5
- 230000003993 interaction Effects 0.000 description 5
- 238000002955 isolation Methods 0.000 description 5
- 230000000670 limiting Effects 0.000 description 5
- 238000002703 mutagenesis Methods 0.000 description 5
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 5
- 230000001105 regulatory Effects 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 230000002588 toxic Effects 0.000 description 5
- 231100000331 toxic Toxicity 0.000 description 5
- 239000003053 toxin Substances 0.000 description 5
- 108020003112 toxins Proteins 0.000 description 5
- 206010003694 Atrophy Diseases 0.000 description 4
- 210000001124 Body Fluids Anatomy 0.000 description 4
- 241000699802 Cricetulus griseus Species 0.000 description 4
- 108010087819 Fc receptors Proteins 0.000 description 4
- 102000009109 Fc receptors Human genes 0.000 description 4
- RAXXELZNTBOGNW-UHFFFAOYSA-N Imidazole Chemical compound C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 4
- 210000003734 Kidney Anatomy 0.000 description 4
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-methionine Chemical compound CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 4
- 239000004472 Lysine Substances 0.000 description 4
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 4
- 210000002241 Neurites Anatomy 0.000 description 4
- 229920000272 Oligonucleotide Polymers 0.000 description 4
- 206010035226 Plasma cell myeloma Diseases 0.000 description 4
- GSEJCLTVZPLZKY-UHFFFAOYSA-N Tris Chemical compound OCCN(CCO)CCO GSEJCLTVZPLZKY-UHFFFAOYSA-N 0.000 description 4
- 239000007983 Tris buffer Substances 0.000 description 4
- 241000251539 Vertebrata <Metazoa> Species 0.000 description 4
- 238000006640 acetylation reaction Methods 0.000 description 4
- 230000003321 amplification Effects 0.000 description 4
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 4
- 229960002685 biotin Drugs 0.000 description 4
- 235000020958 biotin Nutrition 0.000 description 4
- 239000011616 biotin Substances 0.000 description 4
- 239000010839 body fluid Substances 0.000 description 4
- 150000001720 carbohydrates Chemical group 0.000 description 4
- 230000002860 competitive Effects 0.000 description 4
- 238000007796 conventional method Methods 0.000 description 4
- 238000010168 coupling process Methods 0.000 description 4
- 238000011161 development Methods 0.000 description 4
- 230000018109 developmental process Effects 0.000 description 4
- 239000000839 emulsion Substances 0.000 description 4
- 230000012010 growth Effects 0.000 description 4
- 230000001900 immune effect Effects 0.000 description 4
- 230000001939 inductive effect Effects 0.000 description 4
- 230000002401 inhibitory effect Effects 0.000 description 4
- 239000003446 ligand Substances 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 230000001404 mediated Effects 0.000 description 4
- 150000002739 metals Chemical class 0.000 description 4
- 239000000178 monomer Substances 0.000 description 4
- 201000000050 myeloid neoplasm Diseases 0.000 description 4
- 230000000926 neurological Effects 0.000 description 4
- 230000001264 neutralization Effects 0.000 description 4
- 238000003199 nucleic acid amplification method Methods 0.000 description 4
- 238000002823 phage display Methods 0.000 description 4
- 238000001556 precipitation Methods 0.000 description 4
- 230000002285 radioactive Effects 0.000 description 4
- 230000028327 secretion Effects 0.000 description 4
- 239000006228 supernatant Substances 0.000 description 4
- 230000002194 synthesizing Effects 0.000 description 4
- 238000001262 western blot Methods 0.000 description 4
- 229920000936 Agarose Polymers 0.000 description 3
- 108020004705 Codon Proteins 0.000 description 3
- QIVBCDIJIAJPQS-SECBINFHSA-N D-tryptophane Chemical compound C1=CC=C2C(C[C@@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-SECBINFHSA-N 0.000 description 3
- 241000196324 Embryophyta Species 0.000 description 3
- 102000005720 Glutathione Transferase family Human genes 0.000 description 3
- 108010070675 Glutathione Transferase family Proteins 0.000 description 3
- 108010004889 Heat-Shock Proteins Proteins 0.000 description 3
- 102000002812 Heat-Shock Proteins Human genes 0.000 description 3
- 241000238631 Hexapoda Species 0.000 description 3
- 108090001124 Immunoglobulin E Proteins 0.000 description 3
- 102000013463 Immunoglobulin Light Chains Human genes 0.000 description 3
- 108010065825 Immunoglobulin Light Chains Proteins 0.000 description 3
- 108010067060 Immunoglobulin Variable Region Proteins 0.000 description 3
- 102000017727 Immunoglobulin Variable Region Human genes 0.000 description 3
- 108010004484 Immunotoxins Proteins 0.000 description 3
- XUJNEKJLAYXESH-REOHCLBHSA-N L-cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 3
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 3
- 210000004698 Lymphocytes Anatomy 0.000 description 3
- 241001465754 Metazoa Species 0.000 description 3
- 241000699660 Mus musculus Species 0.000 description 3
- 206010053643 Neurodegenerative disease Diseases 0.000 description 3
- 210000001672 Ovary Anatomy 0.000 description 3
- 208000003715 Parkinsonian Disorders Diseases 0.000 description 3
- 206010034010 Parkinsonism Diseases 0.000 description 3
- 229920001949 Transfer RNA Polymers 0.000 description 3
- 108020004566 Transfer RNA Proteins 0.000 description 3
- 238000009825 accumulation Methods 0.000 description 3
- 230000002378 acidificating Effects 0.000 description 3
- 238000007112 amidation reaction Methods 0.000 description 3
- 230000000890 antigenic Effects 0.000 description 3
- 230000000903 blocking Effects 0.000 description 3
- 235000014633 carbohydrates Nutrition 0.000 description 3
- 230000001413 cellular Effects 0.000 description 3
- 238000006243 chemical reaction Methods 0.000 description 3
- 238000007398 colorimetric assay Methods 0.000 description 3
- 230000001808 coupling Effects 0.000 description 3
- 238000005859 coupling reaction Methods 0.000 description 3
- 238000010192 crystallographic characterization Methods 0.000 description 3
- 125000004122 cyclic group Chemical group 0.000 description 3
- 235000018417 cysteine Nutrition 0.000 description 3
- 231100000599 cytotoxic agent Toxicity 0.000 description 3
- 238000003745 diagnosis Methods 0.000 description 3
- 238000002405 diagnostic procedure Methods 0.000 description 3
- 150000002019 disulfides Chemical class 0.000 description 3
- 239000001963 growth media Substances 0.000 description 3
- 229910001385 heavy metal Inorganic materials 0.000 description 3
- 239000002596 immunotoxin Substances 0.000 description 3
- 230000003834 intracellular Effects 0.000 description 3
- 238000001990 intravenous administration Methods 0.000 description 3
- 230000003902 lesions Effects 0.000 description 3
- 125000005647 linker group Chemical group 0.000 description 3
- 238000004811 liquid chromatography Methods 0.000 description 3
- 230000011987 methylation Effects 0.000 description 3
- 238000007069 methylation reaction Methods 0.000 description 3
- 238000010369 molecular cloning Methods 0.000 description 3
- 238000010172 mouse model Methods 0.000 description 3
- 230000001537 neural Effects 0.000 description 3
- 230000002887 neurotoxic Effects 0.000 description 3
- 238000006366 phosphorylation reaction Methods 0.000 description 3
- 230000000865 phosphorylative Effects 0.000 description 3
- 239000004417 polycarbonate Substances 0.000 description 3
- 108091008117 polyclonal antibodies Proteins 0.000 description 3
- 230000001323 posttranslational Effects 0.000 description 3
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 3
- 230000002265 prevention Effects 0.000 description 3
- 230000002829 reduced Effects 0.000 description 3
- 102220069125 rs77384282 Human genes 0.000 description 3
- 230000003248 secreting Effects 0.000 description 3
- 231100000486 side effect Toxicity 0.000 description 3
- 238000002741 site-directed mutagenesis Methods 0.000 description 3
- 239000004055 small Interfering RNA Substances 0.000 description 3
- 230000004083 survival Effects 0.000 description 3
- 239000000725 suspension Substances 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- 230000002103 transcriptional Effects 0.000 description 3
- 230000001131 transforming Effects 0.000 description 3
- 239000003981 vehicle Substances 0.000 description 3
- 238000005406 washing Methods 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- SJRHAKCJDLBPHV-UHFFFAOYSA-N 2-(3,4-dihydroxyphenyl)-N-oxoacetamide Chemical compound OC1=CC=C(CC(=O)N=O)C=C1O SJRHAKCJDLBPHV-UHFFFAOYSA-N 0.000 description 2
- 101700027111 3SA0 Proteins 0.000 description 2
- 229940022698 Acetylcholinesterase Drugs 0.000 description 2
- 108010022752 Acetylcholinesterase Proteins 0.000 description 2
- 102000012440 Acetylcholinesterase Human genes 0.000 description 2
- 108090000206 Autoantibodies Proteins 0.000 description 2
- 102000003852 Autoantibodies Human genes 0.000 description 2
- 235000014469 Bacillus subtilis Nutrition 0.000 description 2
- 210000001218 Blood-Brain Barrier Anatomy 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 210000000349 Chromosomes Anatomy 0.000 description 2
- 241000699800 Cricetinae Species 0.000 description 2
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 description 2
- 101700061444 DDX25 Proteins 0.000 description 2
- 101710007887 DHFR Proteins 0.000 description 2
- 206010012289 Dementia Diseases 0.000 description 2
- 231100000491 EC50 Toxicity 0.000 description 2
- 238000002965 ELISA Methods 0.000 description 2
- 241000283074 Equus asinus Species 0.000 description 2
- 101710013836 HSPD1 Proteins 0.000 description 2
- 108010012104 Heterophile Antibodies Proteins 0.000 description 2
- 108010093488 His-His-His-His-His-His Proteins 0.000 description 2
- 229940088597 Hormone Drugs 0.000 description 2
- 229960000310 ISOLEUCINE Drugs 0.000 description 2
- 210000000987 Immune System Anatomy 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- 102000004090 Immunoglobulin Isotypes Human genes 0.000 description 2
- 108090000539 Immunoglobulin Isotypes Proteins 0.000 description 2
- 102000012745 Immunoglobulin Subunits Human genes 0.000 description 2
- 108010079585 Immunoglobulin Subunits Proteins 0.000 description 2
- 231100000608 Immunotoxin Toxicity 0.000 description 2
- 210000003000 Inclusion Bodies Anatomy 0.000 description 2
- 229940047124 Interferons Drugs 0.000 description 2
- 102000014150 Interferons Human genes 0.000 description 2
- 108010050904 Interferons Proteins 0.000 description 2
- 229940047122 Interleukins Drugs 0.000 description 2
- 102000015696 Interleukins Human genes 0.000 description 2
- 108010063738 Interleukins Proteins 0.000 description 2
- 108010029660 Intrinsically Disordered Proteins Proteins 0.000 description 2
- SZVJSHCCFOBDDC-UHFFFAOYSA-N Iron(II,III) oxide Chemical compound O=[Fe]O[Fe]O[Fe]=O SZVJSHCCFOBDDC-UHFFFAOYSA-N 0.000 description 2
- 241000726306 Irus Species 0.000 description 2
- 102000004195 Isomerases Human genes 0.000 description 2
- 108090000769 Isomerases Proteins 0.000 description 2
- AGPKZVBTJJNPAG-WHFBIAKZSA-N L-isoleucine Chemical compound CC[C@H](C)[C@H](N)C(O)=O AGPKZVBTJJNPAG-WHFBIAKZSA-N 0.000 description 2
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 2
- COLNVLDHVKWLRT-QMMMGPOBSA-N L-phenylalanine Chemical compound OC(=O)[C@@H](N)CC1=CC=CC=C1 COLNVLDHVKWLRT-QMMMGPOBSA-N 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 2
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 2
- KZSNJWFQEVHDMF-BYPYZUCNSA-N L-valine Chemical compound CC(C)[C@H](N)C(O)=O KZSNJWFQEVHDMF-BYPYZUCNSA-N 0.000 description 2
- HWYHZTIRURJOHG-UHFFFAOYSA-N Luminol Chemical compound O=C1NNC(=O)C2=C1C(N)=CC=C2 HWYHZTIRURJOHG-UHFFFAOYSA-N 0.000 description 2
- QPCDCPDFJACHGM-UHFFFAOYSA-N N,N-bis{2-[bis(carboxymethyl)amino]ethyl}glycine Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(=O)O)CCN(CC(O)=O)CC(O)=O QPCDCPDFJACHGM-UHFFFAOYSA-N 0.000 description 2
- 208000009025 Nervous System Disease Diseases 0.000 description 2
- 206010029305 Neurological disorder Diseases 0.000 description 2
- 241000283973 Oryctolagus cuniculus Species 0.000 description 2
- 101700040790 PH Proteins 0.000 description 2
- 229960005190 Phenylalanine Drugs 0.000 description 2
- 208000009144 Pure Autonomic Failure Diseases 0.000 description 2
- 102200036626 SNCA A53T Human genes 0.000 description 2
- 241000235070 Saccharomyces Species 0.000 description 2
- 229920001891 Small hairpin RNA Polymers 0.000 description 2
- 108010090804 Streptavidin Proteins 0.000 description 2
- 108060008443 TPPP Proteins 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 102000003978 Tissue plasminogen activator Human genes 0.000 description 2
- 108090000373 Tissue plasminogen activator Proteins 0.000 description 2
- 241000723873 Tobacco mosaic virus Species 0.000 description 2
- QORWJWZARLRLPR-UHFFFAOYSA-H Tricalcium phosphate Chemical compound [Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O QORWJWZARLRLPR-UHFFFAOYSA-H 0.000 description 2
- 101710042194 Trpgamma Proteins 0.000 description 2
- 210000002700 Urine Anatomy 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 230000004913 activation Effects 0.000 description 2
- 238000001042 affinity chromatography Methods 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 230000001580 bacterial Effects 0.000 description 2
- 230000003115 biocidal Effects 0.000 description 2
- 239000001506 calcium phosphate Substances 0.000 description 2
- 229910000389 calcium phosphate Inorganic materials 0.000 description 2
- 235000011010 calcium phosphates Nutrition 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 239000003593 chromogenic compound Substances 0.000 description 2
- 230000001684 chronic Effects 0.000 description 2
- 230000021615 conjugation Effects 0.000 description 2
- 101700067609 ctx Proteins 0.000 description 2
- 230000009089 cytolysis Effects 0.000 description 2
- 239000002619 cytotoxin Substances 0.000 description 2
- 230000002939 deleterious Effects 0.000 description 2
- 230000001419 dependent Effects 0.000 description 2
- 238000001212 derivatisation Methods 0.000 description 2
- 238000003795 desorption Methods 0.000 description 2
- 230000001809 detectable Effects 0.000 description 2
- 239000008121 dextrose Substances 0.000 description 2
- 239000000032 diagnostic agent Substances 0.000 description 2
- 238000010790 dilution Methods 0.000 description 2
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 2
- KCXVZYZYPLLWCC-UHFFFAOYSA-N edta Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 239000012530 fluid Substances 0.000 description 2
- GNBHRKFJIUUOQI-UHFFFAOYSA-N fluorescein Chemical compound O1C(=O)C2=CC=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 GNBHRKFJIUUOQI-UHFFFAOYSA-N 0.000 description 2
- MHMNJMPURVTYEJ-UHFFFAOYSA-N fluorescein-5-isothiocyanate Chemical compound O1C(=O)C2=CC(N=C=S)=CC=C2C21C1=CC=C(O)C=C1OC1=CC(O)=CC=C21 MHMNJMPURVTYEJ-UHFFFAOYSA-N 0.000 description 2
- 238000006170 formylation reaction Methods 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 239000005556 hormone Substances 0.000 description 2
- 230000000640 hydroxylating Effects 0.000 description 2
- 238000005805 hydroxylation reaction Methods 0.000 description 2
- 230000002163 immunogen Effects 0.000 description 2
- 230000016784 immunoglobulin production Effects 0.000 description 2
- 230000002055 immunohistochemical Effects 0.000 description 2
- 238000003364 immunohistochemistry Methods 0.000 description 2
- 238000001114 immunoprecipitation Methods 0.000 description 2
- 230000002637 immunotoxin Effects 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 230000002757 inflammatory Effects 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 238000005304 joining Methods 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 230000004576 lipid-binding Effects 0.000 description 2
- 125000003588 lysine group Chemical group [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 2
- 230000002934 lysing Effects 0.000 description 2
- 230000005291 magnetic Effects 0.000 description 2
- 238000004949 mass spectrometry Methods 0.000 description 2
- 230000035800 maturation Effects 0.000 description 2
- OKKJLVBELUTLKV-UHFFFAOYSA-N methanol Chemical compound OC OKKJLVBELUTLKV-UHFFFAOYSA-N 0.000 description 2
- 229960000485 methotrexate Drugs 0.000 description 2
- 238000002493 microarray Methods 0.000 description 2
- 244000005700 microbiome Species 0.000 description 2
- 230000000051 modifying Effects 0.000 description 2
- 230000000626 neurodegenerative Effects 0.000 description 2
- 239000004090 neuroprotective agent Substances 0.000 description 2
- 231100000189 neurotoxic Toxicity 0.000 description 2
- PXHVJJICTQNCMI-UHFFFAOYSA-N nickel Chemical compound [Ni] PXHVJJICTQNCMI-UHFFFAOYSA-N 0.000 description 2
- 239000012188 paraffin wax Substances 0.000 description 2
- 230000036961 partial Effects 0.000 description 2
- 230000001402 polyadenylating Effects 0.000 description 2
- 229920000642 polymer Polymers 0.000 description 2
- 238000002360 preparation method Methods 0.000 description 2
- 210000001236 prokaryotic cell Anatomy 0.000 description 2
- DNIAPMSPPWPWGF-UHFFFAOYSA-N propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 2
- 230000002633 protecting Effects 0.000 description 2
- 230000001681 protective Effects 0.000 description 2
- 238000000159 protein binding assay Methods 0.000 description 2
- 238000005215 recombination Methods 0.000 description 2
- 238000003757 reverse transcription PCR Methods 0.000 description 2
- 238000002864 sequence alignment Methods 0.000 description 2
- 239000007787 solid Substances 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 238000002198 surface plasmon resonance spectroscopy Methods 0.000 description 2
- 201000003356 synucleinopathy Diseases 0.000 description 2
- 125000003396 thiol group Chemical group [H]S* 0.000 description 2
- 229960000187 tissue plasminogen activator Drugs 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 238000002054 transplantation Methods 0.000 description 2
- ORHBXUUXSCNDEV-UHFFFAOYSA-N umbelliferone Chemical compound C1=CC(=O)OC2=CC(O)=CC=C21 ORHBXUUXSCNDEV-UHFFFAOYSA-N 0.000 description 2
- 241000701161 unidentified adenovirus Species 0.000 description 2
- 241000701447 unidentified baculovirus Species 0.000 description 2
- 238000002255 vaccination Methods 0.000 description 2
- 229960005486 vaccines Drugs 0.000 description 2
- 239000004474 valine Substances 0.000 description 2
- 230000000007 visual effect Effects 0.000 description 2
- 239000002023 wood Substances 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N β-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- YMXHPSHLTSZXKH-RVBZMBCESA-N (2,5-dioxopyrrolidin-1-yl) 5-[(3aS,4S,6aR)-2-oxo-1,3,3a,4,6,6a-hexahydrothieno[3,4-d]imidazol-4-yl]pentanoate Chemical compound C([C@H]1[C@H]2NC(=O)N[C@H]2CS1)CCCC(=O)ON1C(=O)CCC1=O YMXHPSHLTSZXKH-RVBZMBCESA-N 0.000 description 1
- NKCXQMYPWXSLIZ-PSRDDEIFSA-N (2S)-2-[[(2S)-1-[(2S)-5-amino-2-[[2-[[(2S)-6-amino-2-[[2-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S,3R)-2-[[(2S)-2-[[(2S,3R)-2-amino-3-hydroxybutanoyl]amino]-3-(1H-indol-3-yl)propanoyl]amino]-3-hydroxybutanoyl]amino]propanoyl]amino]-4-oxobutanoyl]amino]-3-m Chemical compound O=C([C@H](CCC(N)=O)NC(=O)CNC(=O)[C@H](CCCCN)NC(=O)CNC(=O)[C@@H](NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](C)NC(=O)[C@@H](NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@@H](N)[C@@H](C)O)[C@@H](C)O)C(C)C)N1CCC[C@H]1C(=O)N[C@@H](CO)C(O)=O NKCXQMYPWXSLIZ-PSRDDEIFSA-N 0.000 description 1
- HUDPLKWXRLNSPC-UHFFFAOYSA-N 4-Aminophthalhydrazide Chemical compound O=C1NNC(=O)C=2C1=CC(N)=CC=2 HUDPLKWXRLNSPC-UHFFFAOYSA-N 0.000 description 1
- XPDXVDYUQZHFPV-UHFFFAOYSA-N 5-Dimethylaminonaphthyl-5-sulfonyl chloride Chemical compound C1=CC=C2C(N(C)C)=CC=CC2=C1S(Cl)(=O)=O XPDXVDYUQZHFPV-UHFFFAOYSA-N 0.000 description 1
- 229940117976 5-hydroxylysine Drugs 0.000 description 1
- ODHCTXKNWHHXJC-VKHMYHEASA-M 5-oxo-L-prolinate Chemical compound [O-]C(=O)[C@@H]1CCC(=O)N1 ODHCTXKNWHHXJC-VKHMYHEASA-M 0.000 description 1
- 101710006746 7.5K Proteins 0.000 description 1
- 229940116904 ANTIINFLAMMATORY THERAPEUTIC RADIOPHARMACEUTICALS Drugs 0.000 description 1
- 102100007409 APRT Human genes 0.000 description 1
- 229960003272 ASPARAGINASE Drugs 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 108010024223 Adenine Phosphoribosyltransferase Proteins 0.000 description 1
- 108010000239 Aequorin Proteins 0.000 description 1
- HTIQEAQVCYTUBX-UHFFFAOYSA-N Amlodipine Chemical compound CCOC(=O)C1=C(COCCN)NC(C)=C(C(=O)OC)C1C1=CC=CC=C1Cl HTIQEAQVCYTUBX-UHFFFAOYSA-N 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N Ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229920000856 Amylose Polymers 0.000 description 1
- 244000303258 Annona diversifolia Species 0.000 description 1
- 235000002198 Annona diversifolia Nutrition 0.000 description 1
- 108010055216 Anti-Idiotypic Antibodies Proteins 0.000 description 1
- 229940064005 Antibiotic throat preparations Drugs 0.000 description 1
- 229940083879 Antibiotics FOR TREATMENT OF HEMORRHOIDS AND ANAL FISSURES FOR TOPICAL USE Drugs 0.000 description 1
- 229940042052 Antibiotics for systemic use Drugs 0.000 description 1
- 210000000628 Antibody-Producing Cells Anatomy 0.000 description 1
- 229940042786 Antitubercular Antibiotics Drugs 0.000 description 1
- 102000007592 Apolipoproteins Human genes 0.000 description 1
- 108010071619 Apolipoproteins Proteins 0.000 description 1
- 239000004475 Arginine Substances 0.000 description 1
- 206010003445 Ascites Diseases 0.000 description 1
- 108010024976 Asparaginase Proteins 0.000 description 1
- 102000015790 Asparaginase Human genes 0.000 description 1
- 229960001230 Asparagine Drugs 0.000 description 1
- 229960005261 Aspartic Acid Drugs 0.000 description 1
- 241001367049 Autographa Species 0.000 description 1
- 206010003816 Autoimmune disease Diseases 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 108090001008 Avidin Proteins 0.000 description 1
- 239000010751 BS 2869 Class A2 Substances 0.000 description 1
- 240000008371 Bacillus subtilis Species 0.000 description 1
- 229940075615 Bacillus subtilis Drugs 0.000 description 1
- 230000036912 Bioavailability Effects 0.000 description 1
- 230000037177 Biodistribution Effects 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 208000000409 Breast Neoplasms Diseases 0.000 description 1
- 241000208199 Buxus sempervirens Species 0.000 description 1
- 108010075254 C-Peptide Proteins 0.000 description 1
- 102000003930 C-Type Lectins Human genes 0.000 description 1
- 108090000342 C-Type Lectins Proteins 0.000 description 1
- 210000004899 C-terminal region Anatomy 0.000 description 1
- 229940105657 CATALASE Drugs 0.000 description 1
- 101700020617 CD22 Proteins 0.000 description 1
- 102100000189 CD22 Human genes 0.000 description 1
- 102100019461 CD28 Human genes 0.000 description 1
- 101700033362 CD28 Proteins 0.000 description 1
- 101700078950 CD44 Proteins 0.000 description 1
- 102100003735 CD44 Human genes 0.000 description 1
- 102100005310 CTLA4 Human genes 0.000 description 1
- 101700054183 CTLA4 Proteins 0.000 description 1
- 241000282836 Camelus dromedarius Species 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- UHBYWPGGCSDKFX-UHFFFAOYSA-N Carboxyglutamic acid Chemical compound OC(=O)C(N)CC(C(O)=O)C(O)=O UHBYWPGGCSDKFX-UHFFFAOYSA-N 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 102000000844 Cell Surface Receptors Human genes 0.000 description 1
- 108010001857 Cell Surface Receptors Proteins 0.000 description 1
- 231100000023 Cell-mediated cytotoxicity Toxicity 0.000 description 1
- 206010057250 Cell-mediated cytotoxicity Diseases 0.000 description 1
- 210000003169 Central Nervous System Anatomy 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 210000001175 Cerebrospinal Fluid Anatomy 0.000 description 1
- 241000251730 Chondrichthyes Species 0.000 description 1
- 230000037250 Clearance Effects 0.000 description 1
- 241000557626 Corvus corax Species 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- 102100005838 DHFR Human genes 0.000 description 1
- 229940039227 DIAGNOSTIC AGENTS Drugs 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 101700011961 DPOM Proteins 0.000 description 1
- 108010016626 Dipeptides Proteins 0.000 description 1
- 206010061818 Disease progression Diseases 0.000 description 1
- 230000036947 Dissociation constant Effects 0.000 description 1
- 229940028937 Divalproex Sodium Drugs 0.000 description 1
- 108020004461 Double-Stranded RNA Proteins 0.000 description 1
- 235000019227 E-number Nutrition 0.000 description 1
- 239000004243 E-number Substances 0.000 description 1
- 102000013460 EC 1.1.1.37 Human genes 0.000 description 1
- 108010026217 EC 1.1.1.37 Proteins 0.000 description 1
- 108010015776 EC 1.1.3.4 Proteins 0.000 description 1
- 108010053835 EC 1.11.1.6 Proteins 0.000 description 1
- 102000016938 EC 1.11.1.6 Human genes 0.000 description 1
- 108010091358 EC 2.4.2.8 Proteins 0.000 description 1
- 102000018251 EC 2.4.2.8 Human genes 0.000 description 1
- 108010067770 Endopeptidase K Proteins 0.000 description 1
- 210000002889 Endothelial Cells Anatomy 0.000 description 1
- 241000588921 Enterobacteriaceae Species 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- LVGKNOAMLMIIKO-QXMHVHEDSA-N Ethyl oleate Chemical compound CCCCCCCC\C=C/CCCCCCCC(=O)OCC LVGKNOAMLMIIKO-QXMHVHEDSA-N 0.000 description 1
- 108010074860 Factor Xa Proteins 0.000 description 1
- 102100011343 GLB1 Human genes 0.000 description 1
- 101700014779 GLB1 Proteins 0.000 description 1
- 229940116332 GLUCOSE OXIDASE Drugs 0.000 description 1
- 102000006395 Globulins Human genes 0.000 description 1
- 108010044091 Globulins Proteins 0.000 description 1
- 108010073178 Glucan 1,4-alpha-Glucosidase Proteins 0.000 description 1
- 239000004366 Glucose oxidase Substances 0.000 description 1
- 108010018962 Glucosephosphate Dehydrogenase Proteins 0.000 description 1
- 102000002794 Glucosephosphate Dehydrogenase Human genes 0.000 description 1
- 229960002989 Glutamic Acid Drugs 0.000 description 1
- RWSXRVCMGQZWBV-WDSKDSINSA-N Glutathione Chemical compound OC(=O)[C@@H](N)CCC(=O)N[C@@H](CS)C(=O)NCC(O)=O RWSXRVCMGQZWBV-WDSKDSINSA-N 0.000 description 1
- 229960003180 Glutathione Drugs 0.000 description 1
- 108010024636 Glutathione Proteins 0.000 description 1
- 102000006602 Glyceraldehyde-3-Phosphate Dehydrogenases Human genes 0.000 description 1
- 108020004445 Glyceraldehyde-3-Phosphate Dehydrogenases Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- ZJYYHGLJYGJLLN-UHFFFAOYSA-N Guanidinium thiocyanate Chemical compound SC#N.NC(N)=N ZJYYHGLJYGJLLN-UHFFFAOYSA-N 0.000 description 1
- 229940093922 Gynecological Antibiotics Drugs 0.000 description 1
- 102100019126 HBB Human genes 0.000 description 1
- 241000606768 Haemophilus influenzae Species 0.000 description 1
- 229940047650 Haemophilus influenzae Drugs 0.000 description 1
- 210000002216 Heart Anatomy 0.000 description 1
- 108091005902 Hemoglobin subunit beta Proteins 0.000 description 1
- 208000009889 Herpes Simplex Diseases 0.000 description 1
- 210000001320 Hippocampus Anatomy 0.000 description 1
- 208000006572 Human Influenza Diseases 0.000 description 1
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 1
- 241000701044 Human gammaherpesvirus 4 Species 0.000 description 1
- 102000015434 Humanized Monoclonal Antibodies Human genes 0.000 description 1
- 108010064750 Humanized Monoclonal Antibodies Proteins 0.000 description 1
- 210000004754 Hybrid Cells Anatomy 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 102000009438 IgE Receptors Human genes 0.000 description 1
- 108010073816 IgE Receptors Proteins 0.000 description 1
- 206010022000 Influenza Diseases 0.000 description 1
- UGQMRVRMYYASKQ-KMPDEGCQSA-N Inosine Natural products O[C@H]1[C@H](O)[C@@H](CO)O[C@@H]1N1C(N=CNC2=O)=C2N=C1 UGQMRVRMYYASKQ-KMPDEGCQSA-N 0.000 description 1
- UGQMRVRMYYASKQ-KQYNXXCUSA-N Inosine Chemical compound O[C@@H]1[C@H](O)[C@@H](CO)O[C@H]1N1C2=NC=NC(O)=C2N=C1 UGQMRVRMYYASKQ-KQYNXXCUSA-N 0.000 description 1
- 229920002459 Intron Polymers 0.000 description 1
- 108020004391 Introns Proteins 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- 108010025815 Kanamycin Kinase Proteins 0.000 description 1
- 241000235058 Komagataella pastoris Species 0.000 description 1
- 102000016551 L-Selectin Human genes 0.000 description 1
- 108010092694 L-Selectin Proteins 0.000 description 1
- QNAYBMKLOCPYGJ-REOHCLBHSA-N L-alanine Chemical compound C[C@H](N)C(O)=O QNAYBMKLOCPYGJ-REOHCLBHSA-N 0.000 description 1
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 1
- CKLJMWTZIZZHCS-REOHCLBHSA-N L-aspartic acid Chemical compound OC(=O)[C@@H](N)CC(O)=O CKLJMWTZIZZHCS-REOHCLBHSA-N 0.000 description 1
- WHUUTDBJXJRKMK-VKHMYHEASA-N L-glutamic acid Chemical compound OC(=O)[C@@H](N)CCC(O)=O WHUUTDBJXJRKMK-VKHMYHEASA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- UKAUYVFTDYCKQA-VKHMYHEASA-N L-homoserine zwitterion Chemical compound OC(=O)[C@@H](N)CCO UKAUYVFTDYCKQA-VKHMYHEASA-N 0.000 description 1
- ROHFNLRQFUQHCH-YFKPBYRVSA-N L-leucine Chemical compound CC(C)C[C@H](N)C(O)=O ROHFNLRQFUQHCH-YFKPBYRVSA-N 0.000 description 1
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 1
- 101800001171 Leader peptide Proteins 0.000 description 1
- 208000008404 Lewy Body Variant of Alzheimer Disease Diseases 0.000 description 1
- 239000005089 Luciferase Substances 0.000 description 1
- 108060001084 Luciferase family Proteins 0.000 description 1
- 210000002751 Lymph Anatomy 0.000 description 1
- 101710029649 MDV043 Proteins 0.000 description 1
- 102100008175 MGAM Human genes 0.000 description 1
- 101710032250 MICAL1 Proteins 0.000 description 1
- 108010059724 Micrococcal Nuclease Proteins 0.000 description 1
- 101710017500 MitHPPK/DHPS Proteins 0.000 description 1
- 108010006519 Molecular Chaperones Proteins 0.000 description 1
- 102000005431 Molecular Chaperones Human genes 0.000 description 1
- 240000002769 Morchella esculenta Species 0.000 description 1
- 235000002779 Morchella esculenta Nutrition 0.000 description 1
- 241000713333 Mouse mammary tumor virus Species 0.000 description 1
- 210000002027 Muscle, Skeletal Anatomy 0.000 description 1
- 229960000951 Mycophenolic Acid Drugs 0.000 description 1
- JDHILDINMRGULE-LURJTMIESA-N N(pros)-methyl-L-histidine Chemical compound CN1C=NC=C1C[C@H](N)C(O)=O JDHILDINMRGULE-LURJTMIESA-N 0.000 description 1
- ZTLGJPIZUOVDMT-UHFFFAOYSA-N N,N-dichlorotriazin-4-amine Chemical compound ClN(Cl)C1=CC=NN=N1 ZTLGJPIZUOVDMT-UHFFFAOYSA-N 0.000 description 1
- 210000004897 N-terminal region Anatomy 0.000 description 1
- 108091007229 NSP3 Papain-like protease domain Proteins 0.000 description 1
- 108091007228 NSP3 acidic domain Proteins 0.000 description 1
- 229940097496 Nasal Spray Drugs 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 206010061309 Neoplasm progression Diseases 0.000 description 1
- 210000001577 Neostriatum Anatomy 0.000 description 1
- 241001182492 Nes Species 0.000 description 1
- 208000001094 Neuroaxonal Dystrophy Diseases 0.000 description 1
- 206010029350 Neurotoxicity Diseases 0.000 description 1
- 239000004677 Nylon Substances 0.000 description 1
- 229940074726 OPHTHALMOLOGIC ANTIINFLAMMATORY AGENTS Drugs 0.000 description 1
- 229940074730 OPHTHAMOLOGIC DIAGNOSTIC AGENTS Drugs 0.000 description 1
- 108020005187 Oligonucleotide Probes Proteins 0.000 description 1
- YBYRMVIVWMBXKQ-UHFFFAOYSA-N PMSF Chemical compound FS(=O)(=O)CC1=CC=CC=C1 YBYRMVIVWMBXKQ-UHFFFAOYSA-N 0.000 description 1
- 101700061424 POLB Proteins 0.000 description 1
- 208000002593 Pantothenate Kinase-Associated Neurodegeneration Diseases 0.000 description 1
- 229940055729 Papain Drugs 0.000 description 1
- 108090000526 Papain Proteins 0.000 description 1
- 108090000284 Pepsin A Proteins 0.000 description 1
- 210000001428 Peripheral Nervous System Anatomy 0.000 description 1
- 108010004729 Phycoerythrin Proteins 0.000 description 1
- 230000037249 Plasma Binding Effects 0.000 description 1
- 230000035728 Plasma binding activity Effects 0.000 description 1
- 241000276498 Pollachius virens Species 0.000 description 1
- 239000004698 Polyethylene (PE) Substances 0.000 description 1
- 241001505332 Polyomavirus sp. Species 0.000 description 1
- 239000004743 Polypropylene Substances 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 210000000063 Presynaptic Terminals Anatomy 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 108010078762 Protein Precursors Proteins 0.000 description 1
- 102000014961 Protein Precursors Human genes 0.000 description 1
- 108010001267 Protein Subunits Proteins 0.000 description 1
- 102000002067 Protein Subunits Human genes 0.000 description 1
- 231100000654 Protein toxin Toxicity 0.000 description 1
- 229940043131 Pyroglutamate Drugs 0.000 description 1
- 101700054624 RF1 Proteins 0.000 description 1
- 108010033725 Recombinant Proteins Proteins 0.000 description 1
- 102000007312 Recombinant Proteins Human genes 0.000 description 1
- 210000003660 Reticulum Anatomy 0.000 description 1
- 210000001525 Retina Anatomy 0.000 description 1
- PYWVYCXTNDRMGF-UHFFFAOYSA-N Rhodamine B Chemical compound [Cl-].C=12C=CC(=[N+](CC)CC)C=C2OC2=CC(N(CC)CC)=CC=C2C=1C1=CC=CC=C1C(O)=O PYWVYCXTNDRMGF-UHFFFAOYSA-N 0.000 description 1
- AUNGANRZJHBGPY-SCRDCRAPSA-N Riboflavin Chemical compound OC[C@@H](O)[C@@H](O)[C@@H](O)CN1C=2C=C(C)C(C)=CC=2N=C2C1=NC(=O)NC2=O AUNGANRZJHBGPY-SCRDCRAPSA-N 0.000 description 1
- 108010083644 Ribonucleases Proteins 0.000 description 1
- 102000006382 Ribonucleases Human genes 0.000 description 1
- 241000724205 Rice stripe tenuivirus Species 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 102100013834 SLC3A2 Human genes 0.000 description 1
- 101710007458 SLC3A2 Proteins 0.000 description 1
- 235000003534 Saccharomyces carlsbergensis Nutrition 0.000 description 1
- 229940081969 Saccharomyces cerevisiae Drugs 0.000 description 1
- 241000607142 Salmonella Species 0.000 description 1
- 206010039447 Salmonellosis Diseases 0.000 description 1
- 101710043164 Segment-4 Proteins 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 1
- 210000003625 Skull Anatomy 0.000 description 1
- 108020004459 Small Interfering RNA Proteins 0.000 description 1
- 229920001985 Small interfering RNA Polymers 0.000 description 1
- 241000256248 Spodoptera Species 0.000 description 1
- 241000194017 Streptococcus Species 0.000 description 1
- 239000012505 Superdex™ Substances 0.000 description 1
- 108091008153 T cell receptors Proteins 0.000 description 1
- 102000016266 T-Cell Antigen Receptors Human genes 0.000 description 1
- 210000001744 T-Lymphocytes Anatomy 0.000 description 1
- 108090000190 Thrombin Proteins 0.000 description 1
- 108020004440 Thymidine Kinase Proteins 0.000 description 1
- 102000006601 Thymidine Kinase Human genes 0.000 description 1
- 229940024982 Topical Antifungal Antibiotics Drugs 0.000 description 1
- 206010044221 Toxic encephalopathy Diseases 0.000 description 1
- 102000003298 Tumor Necrosis Factor Receptors Human genes 0.000 description 1
- 108060008683 Tumor Necrosis Factor Receptors Proteins 0.000 description 1
- ZHSGGJXRNHWHRS-VIDYELAYSA-N Tunicamycin Chemical compound O([C@H]1[C@@H]([C@H]([C@@H](O)[C@@H](CC(O)[C@@H]2[C@H]([C@@H](O)[C@@H](O2)N2C(NC(=O)C=C2)=O)O)O1)O)NC(=O)/C=C/CC(C)C)[C@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1NC(C)=O ZHSGGJXRNHWHRS-VIDYELAYSA-N 0.000 description 1
- 108010046334 Urease Proteins 0.000 description 1
- 241001459538 Ute Species 0.000 description 1
- 101700038759 VP1 Proteins 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- ASCUXPQGEXGEMJ-GPLGTHOPSA-N [(2R,3S,4S,5R,6S)-3,4,5-triacetyloxy-6-[[(2R,3R,4S,5R,6R)-3,4,5-triacetyloxy-6-(4-methylanilino)oxan-2-yl]methoxy]oxan-2-yl]methyl acetate Chemical compound CC(=O)O[C@@H]1[C@@H](OC(C)=O)[C@@H](OC(C)=O)[C@@H](COC(=O)C)O[C@@H]1OC[C@@H]1[C@@H](OC(C)=O)[C@H](OC(C)=O)[C@@H](OC(C)=O)[C@H](NC=2C=CC(C)=CC=2)O1 ASCUXPQGEXGEMJ-GPLGTHOPSA-N 0.000 description 1
- ZBKFYXZXZJPWNQ-UHFFFAOYSA-N [N-]=C=S Chemical compound [N-]=C=S ZBKFYXZXZJPWNQ-UHFFFAOYSA-N 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- DZBUGLKDJFMEHC-UHFFFAOYSA-O acridine;hydron Chemical class C1=CC=CC2=CC3=CC=CC=C3[NH+]=C21 DZBUGLKDJFMEHC-UHFFFAOYSA-O 0.000 description 1
- 239000004480 active ingredient Substances 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 235000004279 alanine Nutrition 0.000 description 1
- 230000001476 alcoholic Effects 0.000 description 1
- 150000003862 amino acid derivatives Chemical class 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 238000005571 anion exchange chromatography Methods 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 239000003242 anti bacterial agent Substances 0.000 description 1
- 239000002260 anti-inflammatory agent Substances 0.000 description 1
- 230000000845 anti-microbial Effects 0.000 description 1
- 230000000111 anti-oxidant Effects 0.000 description 1
- 239000004599 antimicrobial Substances 0.000 description 1
- 239000003963 antioxidant agent Substances 0.000 description 1
- 235000006708 antioxidants Nutrition 0.000 description 1
- 239000008365 aqueous carrier Substances 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 235000009582 asparagine Nutrition 0.000 description 1
- 125000000613 asparagine group Chemical group N[C@@H](CC(N)=O)C(=O)* 0.000 description 1
- 235000003704 aspartic acid Nutrition 0.000 description 1
- 201000007197 atypical autism Diseases 0.000 description 1
- 230000001363 autoimmune Effects 0.000 description 1
- 201000009596 autoimmune hypersensitivity disease Diseases 0.000 description 1
- 230000003608 autoimmunological Effects 0.000 description 1
- 230000002567 autonomic Effects 0.000 description 1
- 238000000376 autoradiography Methods 0.000 description 1
- 244000052616 bacterial pathogens Species 0.000 description 1
- 239000011324 bead Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000003542 behavioural Effects 0.000 description 1
- 108010005774 beta-Galactosidase Proteins 0.000 description 1
- 239000011230 binding agent Substances 0.000 description 1
- 230000035514 bioavailability Effects 0.000 description 1
- 239000003139 biocide Substances 0.000 description 1
- 239000000560 biocompatible material Substances 0.000 description 1
- 230000033228 biological regulation Effects 0.000 description 1
- 238000001574 biopsy Methods 0.000 description 1
- 230000037348 biosynthesis Effects 0.000 description 1
- 238000001815 biotherapy Methods 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- BPKIGYQJPYCAOW-FFJTTWKXSA-I calcium;potassium;disodium;(2S)-2-hydroxypropanoate;dichloride;dihydroxide;hydrate Chemical compound O.[OH-].[OH-].[Na+].[Na+].[Cl-].[Cl-].[K+].[Ca+2].C[C@H](O)C([O-])=O BPKIGYQJPYCAOW-FFJTTWKXSA-I 0.000 description 1
- 201000009030 carcinoma Diseases 0.000 description 1
- 230000017455 cell-cell adhesion Effects 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 201000008779 central nervous system disease Diseases 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 239000000919 ceramic Substances 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 238000007385 chemical modification Methods 0.000 description 1
- 239000000544 cholinesterase inhibitor Substances 0.000 description 1
- 230000035512 clearance Effects 0.000 description 1
- 238000007374 clinical diagnostic method Methods 0.000 description 1
- 238000000975 co-precipitation Methods 0.000 description 1
- 230000001149 cognitive Effects 0.000 description 1
- 238000004737 colorimetric analysis Methods 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 230000004154 complement system Effects 0.000 description 1
- 230000001268 conjugating Effects 0.000 description 1
- 230000023298 conjugation with cellular fusion Effects 0.000 description 1
- 239000000470 constituent Substances 0.000 description 1
- 238000010276 construction Methods 0.000 description 1
- 239000000599 controlled substance Substances 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 229910052802 copper Inorganic materials 0.000 description 1
- 239000010949 copper Substances 0.000 description 1
- RYGMFSIKBFXOCR-UHFFFAOYSA-N copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 1
- 239000007822 coupling agent Substances 0.000 description 1
- 238000004132 cross linking Methods 0.000 description 1
- 210000004748 cultured cells Anatomy 0.000 description 1
- 230000001472 cytotoxic Effects 0.000 description 1
- 239000002254 cytotoxic agent Substances 0.000 description 1
- 230000003247 decreasing Effects 0.000 description 1
- 230000017858 demethylation Effects 0.000 description 1
- 238000010520 demethylation reaction Methods 0.000 description 1
- 238000000326 densiometry Methods 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 108020001096 dihydrofolate reductase family Proteins 0.000 description 1
- 102000004419 dihydrofolate reductase family Human genes 0.000 description 1
- 230000003467 diminishing Effects 0.000 description 1
- 201000009910 diseases by infectious agent Diseases 0.000 description 1
- 238000010494 dissociation reaction Methods 0.000 description 1
- 230000005593 dissociations Effects 0.000 description 1
- 238000009826 distribution Methods 0.000 description 1
- 239000003136 dopamine receptor stimulating agent Substances 0.000 description 1
- 230000003291 dopaminomimetic Effects 0.000 description 1
- 238000005553 drilling Methods 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 239000003792 electrolyte Substances 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 230000002124 endocrine Effects 0.000 description 1
- 230000002255 enzymatic Effects 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- 108010002591 epsilon receptor Proteins 0.000 description 1
- YSMODUONRAFBET-UHNVWZDZSA-N erythro-5-hydroxy-L-lysine Chemical compound NC[C@H](O)CC[C@H](N)C(O)=O YSMODUONRAFBET-UHNVWZDZSA-N 0.000 description 1
- 239000003797 essential amino acid Substances 0.000 description 1
- 235000020776 essential amino acid Nutrition 0.000 description 1
- 150000002148 esters Chemical class 0.000 description 1
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 1
- 229940093471 ethyl oleate Drugs 0.000 description 1
- 230000001747 exhibiting Effects 0.000 description 1
- 238000000605 extraction Methods 0.000 description 1
- 238000000684 flow cytometry Methods 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 238000003197 gene knockdown Methods 0.000 description 1
- 238000010353 genetic engineering Methods 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- 235000019420 glucose oxidase Nutrition 0.000 description 1
- 230000000848 glutamatergic Effects 0.000 description 1
- 235000013922 glutamic acid Nutrition 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 150000003278 haem Chemical group 0.000 description 1
- 101700005460 hemA Proteins 0.000 description 1
- 230000002949 hemolytic Effects 0.000 description 1
- 238000004128 high performance liquid chromatography Methods 0.000 description 1
- 230000001744 histochemical Effects 0.000 description 1
- 239000003276 histone deacetylase inhibitor Substances 0.000 description 1
- 239000012510 hollow fiber Substances 0.000 description 1
- 230000002209 hydrophobic Effects 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 238000002318 immunoblotting Methods 0.000 description 1
- 239000000367 immunologic factor Substances 0.000 description 1
- 239000003547 immunosorbent Substances 0.000 description 1
- 230000003308 immunostimulating Effects 0.000 description 1
- 230000001976 improved Effects 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 238000010348 incorporation Methods 0.000 description 1
- 239000011261 inert gas Substances 0.000 description 1
- 230000028709 inflammatory response Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 229960003786 inosine Drugs 0.000 description 1
- 229940079866 intestinal antibiotics Drugs 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000009114 investigational therapy Methods 0.000 description 1
- 238000006192 iodination reaction Methods 0.000 description 1
- 238000005342 ion exchange Methods 0.000 description 1
- 238000004255 ion exchange chromatography Methods 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- BPHPUYQFMNQIOC-NXRLNHOXSA-N isopropyl β-D-thiogalactopyranoside Chemical compound CC(C)S[C@@H]1O[C@H](CO)[C@H](O)[C@H](O)[C@H]1O BPHPUYQFMNQIOC-NXRLNHOXSA-N 0.000 description 1
- 239000007951 isotonicity adjuster Substances 0.000 description 1
- 230000002147 killing Effects 0.000 description 1
- 229910052747 lanthanoid Inorganic materials 0.000 description 1
- 150000002602 lanthanoids Chemical class 0.000 description 1
- 230000001665 lethal Effects 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 239000007791 liquid phase Substances 0.000 description 1
- 238000004020 luminiscence type Methods 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000003340 mental Effects 0.000 description 1
- 230000002503 metabolic Effects 0.000 description 1
- 230000002906 microbiologic Effects 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 230000003278 mimic Effects 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 230000001459 mortal Effects 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 239000007922 nasal spray Substances 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 201000006443 neuroaxonal dystrophy Diseases 0.000 description 1
- 201000007601 neurodegeneration with brain iron accumulation Diseases 0.000 description 1
- 210000004255 neuroglia Anatomy 0.000 description 1
- 230000000324 neuroprotective Effects 0.000 description 1
- 231100000228 neurotoxicity Toxicity 0.000 description 1
- 229910052759 nickel Inorganic materials 0.000 description 1
- QJGQUHMNIGDVPM-UHFFFAOYSA-N nitrogen group Chemical group [N] QJGQUHMNIGDVPM-UHFFFAOYSA-N 0.000 description 1
- 239000012457 nonaqueous media Substances 0.000 description 1
- 230000036963 noncompetitive Effects 0.000 description 1
- 239000000346 nonvolatile oil Substances 0.000 description 1
- 238000010606 normalization Methods 0.000 description 1
- 235000015097 nutrients Nutrition 0.000 description 1
- 229920001778 nylon Polymers 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 239000002751 oligonucleotide probe Substances 0.000 description 1
- 150000002482 oligosaccharides Polymers 0.000 description 1
- 239000004006 olive oil Substances 0.000 description 1
- 235000008390 olive oil Nutrition 0.000 description 1
- 229940005935 ophthalmologic Antibiotics Drugs 0.000 description 1
- 230000014207 opsonization Effects 0.000 description 1
- 230000003571 opsonizing Effects 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 150000002895 organic esters Chemical class 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 230000003647 oxidation Effects 0.000 description 1
- 238000007254 oxidation reaction Methods 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 239000001301 oxygen Substances 0.000 description 1
- MYMOFIZGZYHOMD-UHFFFAOYSA-N oxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 1
- 235000019834 papain Nutrition 0.000 description 1
- 230000005298 paramagnetic Effects 0.000 description 1
- 238000007911 parenteral administration Methods 0.000 description 1
- 239000002245 particle Substances 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 239000008188 pellet Substances 0.000 description 1
- 230000000149 penetrating Effects 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 1
- 229940111202 pepsin Drugs 0.000 description 1
- 239000000137 peptide hydrolase inhibitor Substances 0.000 description 1
- 239000011886 peripheral blood Substances 0.000 description 1
- 239000002953 phosphate buffered saline Substances 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- 230000003169 placental Effects 0.000 description 1
- 229920002401 polyacrylamide Polymers 0.000 description 1
- 229920000515 polycarbonate Polymers 0.000 description 1
- 229920000573 polyethylene Polymers 0.000 description 1
- 229920001155 polypropylene Polymers 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 229920000915 polyvinyl chloride Polymers 0.000 description 1
- 239000004800 polyvinyl chloride Substances 0.000 description 1
- 230000004481 post-translational protein modification Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 239000000651 prodrug Substances 0.000 description 1
- 229940002612 prodrugs Drugs 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 230000000750 progressive Effects 0.000 description 1
- 230000000069 prophylaxis Effects 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- 238000000575 proteomic Methods 0.000 description 1
- 210000001938 protoplasts Anatomy 0.000 description 1
- 230000001003 psychopharmacologic Effects 0.000 description 1
- 239000002287 radioligand Substances 0.000 description 1
- 239000002464 receptor antagonist Substances 0.000 description 1
- 230000010837 receptor-mediated endocytosis Effects 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000001718 repressive Effects 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 238000007363 ring formation reaction Methods 0.000 description 1
- 238000004805 robotic Methods 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 125000003616 serine group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 description 1
- 238000007086 side reaction Methods 0.000 description 1
- 238000001542 size-exclusion chromatography Methods 0.000 description 1
- 238000004513 sizing Methods 0.000 description 1
- AWUCVROLDVIAJX-GSVOUGTGSA-N sn-glycerol 3-phosphate Chemical compound OC[C@@H](O)COP(O)(O)=O AWUCVROLDVIAJX-GSVOUGTGSA-N 0.000 description 1
- 235000017557 sodium bicarbonate Nutrition 0.000 description 1
- 229910000030 sodium bicarbonate Inorganic materials 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- PHPHRWZFQRLJIA-UZUGEDCSSA-M sodium;(2S,3R,4S,5S,6R)-6-(hydroxymethyl)oxane-2,3,4,5-tetrol;chloride Chemical compound [Na+].[Cl-].OC[C@H]1O[C@H](O)[C@H](O)[C@@H](O)[C@@H]1O PHPHRWZFQRLJIA-UZUGEDCSSA-M 0.000 description 1
- 230000000392 somatic Effects 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 238000009987 spinning Methods 0.000 description 1
- 210000000130 stem cell Anatomy 0.000 description 1
- 239000008174 sterile solution Substances 0.000 description 1
- 230000004960 subcellular localization Effects 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000005670 sulfation reaction Methods 0.000 description 1
- 125000004434 sulfur atoms Chemical group 0.000 description 1
- 230000000576 supplementary Effects 0.000 description 1
- 239000000829 suppository Substances 0.000 description 1
- 238000004114 suspension culture Methods 0.000 description 1
- 230000005062 synaptic transmission Effects 0.000 description 1
- 230000005700 syncytium formation by plasma membrane fusion Effects 0.000 description 1
- 238000010189 synthetic method Methods 0.000 description 1
- 238000004885 tandem mass spectrometry Methods 0.000 description 1
- 229960004072 thrombin Drugs 0.000 description 1
- 231100000730 tolerability Toxicity 0.000 description 1
- 230000000699 topical Effects 0.000 description 1
- 230000001052 transient Effects 0.000 description 1
- 230000014621 translational initiation Effects 0.000 description 1
- 230000001296 transplacental Effects 0.000 description 1
- 230000001960 triggered Effects 0.000 description 1
- 210000004881 tumor cells Anatomy 0.000 description 1
- 238000000539 two dimensional gel electrophoresis Methods 0.000 description 1
- 238000010798 ubiquitination Methods 0.000 description 1
- 241000701366 unidentified nuclear polyhedrosis viruses Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- MSRILKIQRXUYCT-UHFFFAOYSA-M valproate semisodium Chemical compound [Na+].CCCC(C(O)=O)CCC.CCCC(C([O-])=O)CCC MSRILKIQRXUYCT-UHFFFAOYSA-M 0.000 description 1
- 229960000604 valproic acid Drugs 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 230000003612 virological Effects 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 108020001612 μ-opioid receptors Proteins 0.000 description 1
- 102000037275 μ-opioid receptors Human genes 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/68—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6849—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/14—Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
- A61P25/16—Anti-Parkinson drugs
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P25/00—Drugs for disorders of the nervous system
- A61P25/28—Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/34—Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/565—Complementarity determining region [CDR]
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2333/00—Assays involving biological materials from specific organisms or of a specific nature
- G01N2333/435—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
- G01N2333/46—Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
- G01N2333/47—Assays involving proteins of known structure or function as defined in the subgroups
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/28—Neurological disorders
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/28—Neurological disorders
- G01N2800/2814—Dementia; Cognitive disorders
- G01N2800/2828—Prion diseases
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N2800/00—Detection or diagnosis of diseases
- G01N2800/28—Neurological disorders
- G01N2800/2835—Movement disorders, e.g. Parkinson, Huntington, Tourette
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/68—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
- G01N33/6893—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
- G01N33/6896—Neurological disorders, e.g. Alzheimer's disease
Abstract
Disclosed is an isolated antibody or antigen-binding fragment thereof that specifically binds to human ?-synuclein (SEQ ID NO: 1), said antibody or antigen-binding fragment comprising: -a VH CDR1 region comprising the amino acid sequence of NYAMH or a sequence that differs from NYAMH by one amino acid; -a VH CDR2 region comprising the amino acid sequence of WINAGNGKRKYSQKFQD or a sequence that differs from WINAGNGKRKYSQKFQD by one amino acid; -a VH CDR3 region comprising the amino acid sequence of EEDHAGSGSYLSMDV or a sequence that differs from EEDHAGSGSYLSMDV by one amino acid; -a VL CDR1 region comprising the amino acid sequence of KSSQNVLYSSNNKNYLA or a sequence that differs from KSSQNVLYSSNNKNYLA by one amino acid; -a VL CDR2 region comprising the amino acid sequence of WASTRES or a sequence that differs from WASTRES by one amino acid; and -a VL CDR3 region comprising the amino acid sequence of QQYYSSPLT or a sequence that differs from QQYYSSPLT by one amino acid. cid; -a VH CDR2 region comprising the amino acid sequence of WINAGNGKRKYSQKFQD or a sequence that differs from WINAGNGKRKYSQKFQD by one amino acid; -a VH CDR3 region comprising the amino acid sequence of EEDHAGSGSYLSMDV or a sequence that differs from EEDHAGSGSYLSMDV by one amino acid; -a VL CDR1 region comprising the amino acid sequence of KSSQNVLYSSNNKNYLA or a sequence that differs from KSSQNVLYSSNNKNYLA by one amino acid; -a VL CDR2 region comprising the amino acid sequence of WASTRES or a sequence that differs from WASTRES by one amino acid; and -a VL CDR3 region comprising the amino acid sequence of QQYYSSPLT or a sequence that differs from QQYYSSPLT by one amino acid.
Description
ANTI-ALPHA SYNUCLEIN BINDING LES
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
The content of the electronically submitted sequence listing in ASCII text file
(Name: celisting_ascii.txt; Size: 23KB; and Date of Creation: June 23, 2011) filed
with the application is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
The present invention generally s to novel uclein-specific binding
molecules, particularly human antibodies as well as fragments, derivatives and variants
thereof that ize u-synuclein and aggregated forms of u-synuclein, respectively. In
addition, the present invention relates to ceutical and diagnostic compositions
sing such binding molecules, antibodies and mimics thereof valuable both as a
diagnostic tool to identify toxic species of (x-synuclein in plasma and CSF and also in
passive vaccination strategies for treating ers related to aggregates of (x-synuclein
such as Parkinson’s disease (PD), ia with Lewy bodies (DLB) and Lewy body
variant of Alzheimer’s disease (AD) and other synucleinopathic es.
Protein misfolding and aggregation are pathological aspects of numerous
neurodegenerative diseases. Aggregates of (x-synuclein are major components of the
Lewy bodies and Lewy neurites associated with Parkinson's disease (PD). A natively
unfolded protein, (x-synuclein can adopt different aggregated morphologies, including
oligomers, protofibrils and fibrils. The small oligomeric aggregates have been shown to
be particularly toxic.
Naturally occurring autoantibodies against (x-synuclein have been ed in
healthy persons and altered levels in patients were associated with particular
neurodegenerative ers; see for review Neff et al., Autoimmun. Rev. 7 (2008), 501-
507. Thus, naturally occurring antibodies in patients suffering from son’s disease,
either spontaneously or upon vaccination, in particular in healthy patients can serve a
protective role with respect to (x-synuclein aggregation; see, e. g., Woulfe et al.,
Neurology 58 (2002), 1435-1436 and Papachroni et al., J. Neurochem. 101 (2007), 749-
756. Hitherto, the therapeutic significance of autoantibodies had been difficult to assess.
This is mostly due to the lack of ht-forward experimental approaches for their
isolation and subsequent characterization in vitro.
Recently, oligomeric species of u-synuclein have been reported ellularly in
plasma and CSF (El-Agnaf et al., FASEB J. 20 (2006), 5) and immunization
studies in mouse models of PD show that extracellular mouse monoclonal antibodies
against u-synuclein can reduce accumulation of intracellular u-synuclein ates
(Masliah et al., Neuron, 46 (2005), 857—868) supporting the idea that dies that
neutralize the neurotoxic aggregates without ering with beneficial functions of
monomeric u-synuclein can be useful therapeutics. However, the therapeutic utility of
murine based dies in human is ed by the human anti-mouse antibody
(HAMA) response in view of their non-human origin.
Emadi et al. in J. Mol. Biol. 368 (2007), 1132—1144, describe the isolation of
single chain antibody fragments (scFvs) from a phage displayed antibody library based on
human sequences against u-synuclein, which bind only to an oligomeric form of 0L-
synuclein and inhibit both aggregation and toxicity of u-synuclein in vitro. However,
although the generation of scFvs from phage display is rather simple, this technique has
severe drawbacks since the antibodies so produced bear the risk of undesired
crossreactivity against self-antigens and lack the characteristics of evolutionary optimized
natural human antibodies produced by the human immune system. Furthermore, such
antibodies may not be specific enough because of cross-reactivity with other proteins
and/or with the target protein in context with normal logical environment and
function. In case of Parkinson’s disease, for example, antibodies that also cross-react with
physiological derivatives of u-synuclein bear the ial to cause side effects related to
the normal functions of the physiologic target structures. In this respect, an undesired
autoimmune disease would downrightly be d — a hardly calculable risk also in the
conceptual design of active immunization ments employing protein structures that,
in variant form, also occur physiologically.
More recently, Seitz et al. (81. Kongress der Deutschen Gesellschaft fur
Neurologie mit ldungsakademie Hamburg lO.-l3.09.2008), reported on the
isolation of anti-(x-synuclein polyclonal autoantibody from different immunoglobulin
solutions and s of single blood donors h affinity chromatography. However,
besides the fact that this approach provides mere limited amounts of the desired antibody,
W0 2012/177972 2012/043701
_ 3 _
polyclonal antibodies are of only limited use for therapeutic application, for example
because of their geneity and the risk of being contaminated with other (x-synuclein
associated les which have undesired side effects. Likewise, the diagnostic value of
polyclonal antibodies is reduced since the variability of the composition of the antibodies
will influence the overall specificity and reactivity. This is all the more true for antibodies
against proteins subject of aggregation and deposition due to misfolding.
Thus, there is a need to overcome the above-described limitations and to provide a
therapeutic and diagnostic human antibody against uclein.
SUMMARY OF THE INVENTION
In one embodiment, the present invention provides an isolated binding molecule
which specifically binds to an epitope within amino acids 4 to 15 of human (x-synuclein
(SEQ ID NO:1). In certain aspects the binding molecule itively inhibits the
binding of reference monoclonal antibody NI-202.12F4 to (x-synuclein. A binding
molecule of the invention can be an antibody, or an antigen-binding fragment thereof In
particular embodiments the binding molecule is not human monoclonal antibody NI-
202.12F4, or an antigen-binding nt, t, or tive thereof.
Another embodiment es an isolated binding molecule which specifically
binds to an epitope within amino acids 113 to 123 or within amino acids 117 to 123 of 0L-
synuclein (SEQ ID NO:1). In certain aspects the binding molecule specifically binds to
the same human (x-synuclein epitope as the reference monoclonal antibody NI-
202.22D11, or competitively inhibits the binding reference monoclonal antibody NI-
202.22D11 to human uclein. A binding molecule of the invention can be an
antibody, or an antigen-binding fragment thereof. A particular binding molecule of the
invention is the human monoclonal antibody NI-202.22D11, or an antigen-binding
fragment, variant, or derivative f.
The invention further provides an isolated antibody or n binding fragment
thereof that specifically binds to human uclein, comprising an globulin
heavy chain le region (VH) and an immunoglobulin light chain variable region
(VL), where the VH comprises a polypeptide sequence at least 90%, or 100% identical to
SEQ ID NO:15 or SEQ ID NO:20. Also provided is an isolated antibody or antigen
binding fragment thereof that specifically binds to human (x-synuclein, comprising a VH
2012/043701
and a VL, where the VL comprises a polypeptide sequence at least 90%, or 100%
identical to SEQ ID NO:22 or SEQ ID NO:26. Similarly, the invention provides an
isolated antibody or antigen binding fragment thereof that cally binds to human or-
synuclein, comprising a VH and a VL where the VH and VL comprise, respectively,
polypeptide sequences at least 90%, or 100% identical to the reference ptides SEQ
ID NO:lS and SEQ ID NO:22, SEQ ID NO:lS and SEQ ID NO:26, SEQ ID N020 and
SEQ ID NO:22, or SEQ ID N020 and SEQ ID NO:26.
Further ed are isolated polypeptides, including an isolated ptide
comprising a VH, where the CDRl region of the VH is identical, or identical except for
less than 3 conservative amino acid substitutions, to reference heavy chain CDRl
sequence SEQ ID NO:l6, an isolated polypeptide comprising a VH, where the CDR2
region of the VH is identical, or identical except for less than 5 conservative amino acid
substitutions, to reference heavy chain CDR2 sequence SEQ ID NO:l7, an isolated
ptide comprising a VH, where the CDR3 region of the VH is identical, or identical
except for less than 5 conservative amino acid tutions, to reference heavy chain
CDR3 sequence SEQ ID NO:18, an isolated polypeptide comprising a VL, where the
CDRl region of the VL is identical, or identical except for less than 5 conservative amino
acid substitutions, to reference light chain CDRl sequence SEQ ID NO:23, an isolated
polypeptide comprising a VL, where the CDR2 region of the VL is identical, or identical
except for less than 3 conservative amino acid substitutions, to reference heavy chain
CDR2 sequence SEQ ID N024, and an isolated polypeptide comprising a VL, where the
CDR3 region of the VL is identical, or identical except for less than 3 conservative amino
acid substitutions, to reference heavy chain CDR3 sequence SEQ ID NO:25. In each of
the above stated polypeptides, an antibody or antigen-binding fragment thereof
comprising the polypeptide specifically binds to human clein.
In certain embodiments the invention the isolated dy or fragment f
preferentially binds to a non-linear mational epitope of human (x-synuclein. In
other embodiments the isolated antibody or fragment f preferentially binds human
(x-synuclein in the oligomeric or aggregated form. In filrther embodiments the isolated
antibody or fragment thereof does not specifically bind to human B-synuclein or human y-
synuclein, and/or does not specifically bind to murine u-synuclein.
Also provided is a composition comprising an antibody or fragment thereof as
described above, and a carrier. The composition may be a therapeutic or a diagnostic
composition.
Further provide are one or more isolated polynucleotides encoding a polypeptide
or binding molecule as described herein, and vectors and host cells for expressing such
binding molecules.
It is a particular object of the present ion to provide methods for treating or
preventing a einopathic disease such as, but not limited to Parkinson's disease
(PD), dementia with Lewy bodies (DLB), and multiple systems atrophy (MSA). The
methods comprise administering an effective concentration of anti-human a-synuclein
binding molecule, e.g., an antibody or antigen-binding nt, variant, or derivative
to the subject where the antibody targets a-synuclein.
It is also an object of the invention to provide a method of diagnosing a
synucleinopathic disease in a subject, sing assessing the level, zation,
conformation or a combination thereof of a-synuclein in a subject to be diagnosed with
an antibody or fragment thereof of the invention and comparing the level, localization,
conformation or ation thereof of a-synuclein in the subject to one or more
reference standards derived from one or more control samples, where a difference or
similarity between the level, localization, conformation or combination thereof of asynuclein
in the t and the nce standard indicates whether the subject has a
synucleinopathic disease.
Diagnostic methods of the invention can be through in vitro assay of patient
samples, or by in vivo imaging techniques.
[0018a] Definitions of specific embodiments of the ion as claimed herein follow.
[0018b] According to a first embodiment of the invention, there is provided an isolated
antibody or antigen-binding fragment thereof that specifically binds to human αsynuclein
(SEQ ID NO:1), said dy or antigen-binding fragment comprising:
a VH CDR1 region comprising the amino acid ce of SEQ ID NO:16 or a
ce that differs from SEQ ID NO:16 by one amino acid;
a VH CDR2 region comprising the amino acid ce of SEQ ID NO:17 or a
ce that differs from SEQ ID NO:17 by one amino acid;
a VH CDR3 region sing the amino acid sequence of SEQ ID NO:18 or a
sequence that differs from SEQ ID NO:18 by one amino acid;
- 5a -
a VL CDR1 region comprising the amino acid sequence of SEQ ID NO:23 or a
sequence that differs from SEQ ID NO:23 by one amino acid;
a VL CDR2 region comprising the amino acid sequence of SEQ ID NO:24 or a
ce that differs from SEQ ID NO:24 by one amino acid; and
a VL CDR3 region comprising the amino acid sequence of SEQ ID NO:25 or a
sequence that differs from SEQ ID NO:25 by one amino acid.
[0018c] According to a second ment of the invention, there is provided an
isolated dy or antigen-binding fragment thereof that specifically binds to human
α-synuclein (SEQ ID NO:1) comprising:
a VH CDR1 region comprising the amino acid sequence of SEQ ID NO:16;
a VH CDR2 region comprising the amino acid sequence of SEQ ID NO:17;
a VH CDR3 region comprising the amino acid sequence of SEQ ID NO:18;
a VL CDR1 region comprising the amino acid sequence of SEQ ID NO:23;
a VL CDR2 region comprising the amino acid sequence of SEQ ID NO:24; and
a VL CDR3 region comprising the amino acid sequence of SEQ ID NO:25.
[0018d] According to a third embodiment of the invention, there is provided a
composition comprising the antibody or antigen-binding fragment of the first or second
embodiments and a carrier.
[0018e] According to a fourth embodiment of the invention, there is provided an isolated
polynucleotide comprising a nucleic acid encoding the antibody or antigen-binding
fragment thereof of the first or second ments.
[0018f] According to a fifth embodiment of the invention, there is provided a
composition sing the cleotide of the fourth ment and a carrier.
[0018g] According to a sixth embodiment of the invention, there is provided a vector
comprising the polynucleotide of the fourth embodiment.
[0018h] According to a seventh embodiment of the invention, there is provided an
isolated host cell comprising the polynucleotide of the fourth embodiment or the vector
of the sixth embodiment.
] According to an eighth embodiment of the invention, there is provided a method
of producing an anti-human clein antibody or antigen-binding fragment f,
said method comprising culturing the host cell of the seventh embodiment and
recovering the antibody or antigen-binding nt thereof.
- 5b -
[0018j] According to a ninth embodiment of the invention, there is provided an anti-
human α-synuclein antibody or antigen-binding fragment thereof produced by the
method of the eighth embodiment.
[0018k] According to a tenth embodiment of the invention, there is provided use of the
antibody or antigen-binding nt of the first or ninth embodiments, the composition
of the third or fifth embodiments, the polynucleotide of the fourth embodiment, the
vector of the sixth embodiment or the host cell of the seventh embodiment in the
manufacture of a medicament for ng or preventing a synucleinopathic disease in a
subject.
[0018l] According to an eleventh embodiment of the invention, there is provided use of
the antibody or antigen-binding fragment thereof of the first or ninth embodiments in
the cture of a medicament for diagnosing whether a subject has a
synucleinopathic disease, wherein said diagnosing ses:
(a) assessing a level, localization, conformation or a combination thereof of
clein in the subject to be diagnosed with the medicament; and
(b) comparing the level, localization, mation or combination thereof of
said α-synuclein in the subject to one or more reference rds derived from one or
more control samples,
wherein a difference or similarity between the level, localization, conformation
or combination thereof of said α-synuclein in the subject and the one or more reference
standards indicates r the subject has a synucleinopathic e.
Further embodiments of the present invention will be apparent from the
description and Examples that .
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 Amino acid and nucleotide sequences of the variable region, i.e. heavy
chain and kappa light chains of human antibody .21D11. Framework (FR) and
complementarity determining regions (CDRs) are indicated with the CDRs being
underlined. Due to the cloning strategy the amino acid sequence at the N-terminus of
the heavy chain and light chain may ially contain primer-induced alterations in
FR1,
[Text continues on page 6]
W0 2012/177972 PCT/U82012/043701
_ 6 _
which however do not substantially affect the biological activity of the antibody. In order
to provide a consensus human antibody, the nucleotide and amino acid sequences of the
original clone were aligned with and tuned in accordance with the pertinent human germ
line variable region sequences in the database; see e.g., Vbase (vbase.mrc-cpe.cam.ac.uk)
hosted by the MRC Centre for Protein Engineering (Cambridge, UK). Those amino acids,
which are ered to potentially deviate from the consensus germ line sequence and
thus could be due to the PCR , are indicated in bold.
Figure 2 Recombinant human NI-202.21Dll selectively binds human (x-synuclein
over [3-, y-synuclein and murine u-synuclein in a direct ELISA. Recombinant human u-,B-
,y- synuclein and recombinant human and murine His-tagged (it-synuclein were coated
onto ELISA plates at equal concentration (2 ug/ml). Plates were then probed with
recombinant human NI-202.21Dl 1, human NI-202.12F4 and with a nuclein
antibody. (A) Recombinant NI-202.21D11 selectively binds u-synuclein whereas pan —
synuclein antibody binds to all three synuclein proteins ing equal coating of
inant proteins. (B) Recombinant NI-202.21Dll is selective for human vs murine
(x-synuclein. On the other hand NI-202.12F4 binds to both human and murine (x-synuclein
in a direct ELISA.
Figure 3 Recombinant .21Dll entially binds to high density coated
(it-synuclein. Recombinant human u-synuclein was coated onto ELISA plates at indicated
concentrations and probed with various concentrations of .21Dll by direct ELISA
(El 20 ug/ml;A Sug/mlg91 ug/ml;l 0.25 ug/ml; V 0.1 ug/ml). The half maximal
effective concentration (ECSO) indicating the potency of the antibody was determined for
each coating concentration.
Figure 4 Immunohistochemical binding analysis of NI-202.21Dll showed
prominent staining of (x-synuclein pathology including Lewy body and Lewy neurite like
inclusions in paraffin sections from (A) transgenic mice overexpressing human 0L-
synuclein A53T and (C) from human brain tissue of a Dementia with Lewy Bodies
patient. (B) No staining was observed in wild-type mouse tissue and (bottom right) in a
secondary dy only control. HC=Hippocampus, CTX=Cortex, instem.
Figure 5 Epitope mapping revealed a C-terminal g epitope within human 0L-
synuclein (aa 117-123) for NI-202.21Dll. (A) Recombinant .21Dll bound to the
C-terminal domain of human (x-synuclein in a direct ELISA. (x-synuclein truncations were
coated onto ELISA plates at equal trations (2ug/ml). NI-202.2lDll bound only to
truncated (x-synuclein aa 61-140 and 95-140 but not to truncations aa l-60, l-95. (B)
Pepscan analysis showed binding of NI-202.21Dll to overlapping peptides B08 (aa 109-
123), B09 (aa 113-127) and B10 (aa 117-131) of human (x-synuclein ting that the
l sequence required for NI-202.21Dll binding is PVDPDNE (aa 117-123) within
human (x-synuclein. (C) Recombinant NI-202.21Dll showed reduced binding to human
(x-synuclein DlZlG/N122S in a direct ELISA. Recombinant wt and mutated (x-synuclein
proteins were coated at equal concentration (2ug/ml) onto ELISA plates and tested for
recombinant NI-202.2lDl 1 g.
Figure 6 NI-202.12F4 ively binds to very N-terminus of (x-synuclein. (A)
Pepscan analysis shows binding of NI-202. 12F4 to peptide AOl showing that the minimal
recognition sequence is within residue l-lS of (x-synuclein. (B) Synthetic (x-synuclein
peptides from residue 1-30, 4-30 and 5-30 were tested for NI-202.12F4 binding in an in-
solution ELISA. NI-202.12F4 bound aa l-30 and 4-30 but not 5-30. This showed that
N1202.12F4 epitope sequence starts at residue 4 of (x-synuclein. (C) Residue K10 within
NI-202.12F4 epitope is a key amino acid for selectivity of NI-202.12F4 for (x-synuclein
over B-synuclein. Recombinant wt and mutant (x- and B-synuclein proteins were tested by
direct ELISA for .12F4 binding. NI-202.12F4 bound to wt (x-synuclein and mutant
B-synuclein MlOK but not to wt B-synuclein and mutant (x-synuclein KlOM. This shows
that residue K10 is responsible for NI-202.12F4 u-synuclein selectivity.
DETAILED DESCRIPTION OF THE ION
I. ions
einopathic es or synucleinopathies are a diverse group of
neurodegenerative disorders that share a common pathologic lesion composed of
aggregates of insoluble u-synuclein protein in selectively vulnerable populations of
neurons and glia. These disorders include Parkinson's disease (PD), son’s Disease
Dementia (PDD), dementia with Lewy bodies (DLB), juvenile-onset generalized
neuroaxonal dystrophy (Hallervorden-Spatz disease), pure autonomic failure (PAF),
le system atrophy (MSA) and neurodegeneration with brain iron accumulation
type-l (NBIA-I). Clinically, they are characterized by a chronic and progressive decline
in motor, ive, behavioral, and autonomic fianctions, depending on the distribution of
the lesions.
Parkinson's disease is an age-dependent neurodegenerative disease with unknown
etiology. It is believed that ic Parkinson's disease results from a combination of
genetic vulnerability and nmental insults. It is r ed that Parkinson's
disease (PD) while triggered by disparate mechanisms follows a shared hysiologic
pathway. One shared node is the involvement of u-synuclein. Linkage of this protein with
Parkinson's disease pathogenesis has been ished by the identification of both point
mutations and triplication of the gene in familial cases, the localization of u-synuclein to
Lewy bodies, one of the hallmark pathological features of Parkinson's disease, and the
correlation of u-synuclein expression and disease pathology in neurotoxic models of
Parkinson's disease. r ce indicates that particular forms of u-synuclein (e.g.,
misfolded and clein bonded dopamine) are involved in sporadic disease.
Synucleins are small, soluble proteins expressed primarily in neural tissue and in
certain tumors. The family includes three known proteins: u-synuclein, B-synuclein, and
y-synuclein. All synucleins have in common a highly conserved u-helical lipid-binding
motif with similarity to the class-A2 lipid-binding domains of the exchangeable
apolipoproteins. Synuclein family members are not found outside vertebrates, although
they have some conserved structural similarity with plant 'late-embryo-abundant' ns.
The 0L- and B-synuclein proteins are found primarily in brain tissue, where they are seen
mainly in presynaptic terminals. The y-synuclein protein is found primarily in the
peripheral nervous system and retina, but its expression in breast tumors is a marker for
tumor progression. Normal cellular fianctions have not been determined for any of the
ein proteins, although some data suggest a role in the regulation of membrane
stability and/or turnover. Mutations in u-synuclein are associated with rare familial cases
of early-onset son's disease, and the protein accumulates ally in Parkinson's
disease, Alzheimer's disease, and several other egenerative illnesses. For review
see, e. g., George, Genome Biol. 3 (2002), reviews3002.l—reviews3002.6 published online
December 20, 2001, in which Table l catalogs the unique members of the synuclein
family that are currently listed in k, the disclosure content of which is
incorporated herein by reference.
(x-synuclein was originally identified in human brains as the sor protein of
the amyloid component of (NAC) of Alzheimer’s disease (AD) plaques; see, e. g.,
Ueda et al, Proc. Natl. Acad. Sci. USA. 90 (1993), 1282-1286. u-synuclein, also termed
the precursor of the non-AB component of AD amyloid (NACP), is a protein of 140
amino acids. (x-synuclein exists in its native form as a random coil; however, changes in
pH, molecular crowding, heavy metal content, and dopamine levels all affect protein
conformation. Changes in conformation to eric, proto-fibrillar, fibrillar, and
aggregate moieties are t to regulate protein toxicity. Increasing evidence indicates
that dopamine-adducted (x-synuclein has a faster time course to fibril formation compared
to non-adducted n. Furthermore, dopamine in the background of (x-synuclein
overexpression is toxic.
In this specification, the terms "(x-synuclein", "alpha-synuclein", uclein" and
"aSyn" are used interchangeable to specifically refer to the native monomer form of 0L-
ein. The term "(x-synuclein" is also used to generally identify other conformers of
u-synuclein, for example, (x-synuclein bonded to dopamine-quinone (DAQ) and
oligomers or aggregates of u-synuclein. The term "(x-synuclein" is also used to refer
collectively to all types and forms of u-synuclein. The protein sequence for human 0L-
synuclein is
MDVFMKGLSKAKEGVVAAAEKTKQGVAEAAGKTKEGVLYVGSKTKEGVVHG
VATVAEKTKEQVTNVGGAVVTGVTAVAQKTVEGAGSIAAATGFVKKDQLGKN
EEGAPQEGILEDMPVDPDNEAYEMPSEEGYQDYEPEA (SEQ ID NO: 1). The amino
acid ce of (x-synuclein can be retrieved from the literature and pertinent databases;
see, e.g., Ueda et al., ibid.; GenBank swissprot: locus SYUA_HUMAN, accession
number P37840. The non-AB component of AD d (NAC) is derived from 0L-
synuclein. NAC, a highly hydrophobic domain within (x-synuclein, is a peptide consisting
of at least 28 amino acids residues (residues 60-87) and optionally 35 amino acid residues
ues 61-95). NAC displays a tendency to form a beta-sheet ure (Iwai, et al.,
Biochemistry, 34 (1995) 10139-10145). The amino acid sequences ofNAC are described
in Jensen et al., Biochem. J. 310 (1995), 91-94; k accession number S56746 and
Ueda et al., PNAS USA 90 (1993), 1282-11286.
Disaggregated (x-synuclein or fragments thereof, including NAC, means
monomeric peptide units. Disaggregated (x-synuclein or fragments thereof are generally
PCT/U82012/043701
_ 10 _
soluble, and are capable of self-aggregating to form soluble oligomers. Oligomers of 0L-
ein and fragments thereof are usually soluble and exist predominantly as ces.
Monomeric clein can be prepared in vitro by dissolving lyophilized e in neat
DMSO with sonication. The resulting solution is centrifuged to remove any insoluble
particulates. Aggregated u-synuclein or fragments thereof, including NAC, means
oligomers of u-synuclein or fragments thereof which have associate into insoluble B-sheet
lies. Aggregated u-synuclein or fragments thereof, including NAC, means also
means fibrillar polymers. Fibrils are usually insoluble. Some dies bind either
soluble u-synuclein or fragments thereof or aggregated u-synuclein or fragments thereof.
Some antibodies bind to oligomers of u-synuclein more strongly than to monomeric
forms or fibrillar forms. Some antibodies bind both soluble and ated u-synuclein or
fragments thereof, and optionally oligomeric forms as well.
The human anti-u-synuclein antibodies disclosed herein cally bind 0L-
ein and epitopes thereof and to various conformations of u-synuclein and epitopes
thereof. For e, disclosed herein are antibodies that specifically bind u-synuclein, 0L-
synuclein in its native monomer form, fiJll-length and truncated u-synuclein and 0t-
synuclein aggregates. As used herein, reference to an antibody that "specifically binds",
"selectively binds", or "preferentially binds" u-synuclein refers to an antibody that does
not bind other unrelated proteins. In one example, an u-synuclein antibody disclosed
herein can bind u-synuclein or an epitope thereof and show no binding above about 1.5
times background for other proteins. An dy that "specifically binds" or "selectively
binds" u-synuclein conformer refers to an antibody that does not bind all mations
of u-synuclein, z'.e., does not bind at least one other u-synuclein conformer. For example,
disclosed herein are antibodies that can distinguish among monomeric and ated
forms of u-synuclein, human and mouse u-synuclein; full-length u-synuclein and
truncated forms as well as human u-synuclein versus [3- and y-synuclein. Since the human
anti-(x-synuclein antibodies of the present invention have been isolated from a pool of
elderly subjects with no signs of Parkinsonism and exhibiting an u-synuclein-specific
immune response the x-synuclein antibodies of the present invention are also referred
to as "human ntibodies" in order to emphasize that those antibodies were indeed
expressed by the subjects and have not been isolated from, for example a human
-1]-
immunoglobulin expressing phage library, which hitherto represented one common
method for trying to provide human-like antibodies.
It is to be noted that the term "a" or "an" entity refers to one or more of that entity;
for example, "an antibody," is understood to represent one or more antibodies. As such,
the terms "a" (or "an" "one or more," and "at least one" can be used interchangeably
herein.
As used herein, the term "polypeptide" is intended to encompass a singular
"polypeptide" as well as plural "polypeptides," and refers to a molecule composed of
monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds).
The term eptide" refers to any chain or chains of two or more amino acids, and
does not refer to a specific length of the product. Thus, peptides, dipeptides, tripeptides,
eptides, "protein, amino acid chain," or any other term used to refer to a chain or
chains of two or more amino acids, are included within the definition of "polypeptide,"
and the term "polypeptide" can be used instead of, or hangeably with any of these
terms.
The term "polypeptide" is also intended to refer to the products of post-expression
modifications of the polypeptide, including without limitation glycosylation, ation,
phosphorylation, ion, derivatization by known protecting/blocking groups,
proteolytic cleavage, or modification by non-naturally occurring amino acids. A
polypeptide can be derived from a natural biological source or produced by recombinant
technology, but is not arily translated from a ated nucleic acid sequence. It
an be generated in any manner, including by chemical synthesis.
A ptide of the invention can be of a size of about 3 or more, 5 or more, 10
or more, 20 or more, 25 or more, 50 or more, 75 or more, 100 or more, 200 or more, 500
or more, 1,000 or more, or 2,000 or more amino acids. Polypeptides can have a defined
three-dimensional structure, although they do not arily have such structure.
ptides with a defined three-dimensional structure are referred to as folded, and
polypeptides which do not possess a defined three-dimensional structure, but rather can
adopt a large number of different conformations, and are referred to as unfolded. As used
herein, the term glycoprotein refers to a protein coupled to at least one carbohydrate
moiety that is attached to the protein via an oxygen-containing or a nitrogen-containing
side chain of an amino acid residue, e.g., a serine residue or an asparagine residue.
PCT/U82012/043701
_ 12 _
By an "isolated" polypeptide or a fragment, t, or derivative f is
intended a polypeptide that is not in its natural milieu. No particular level of purification
is required. For example, an isolated polypeptide can be removed from its native or
natural environment. Recombinantly produced polypeptides and proteins expressed in
host cells are considered isolated for purposed of the invention, as are native or
inant polypeptides which have been separated, fractionated, or partially or
substantially purified by any suitable technique.
Also ed as polypeptides of the present invention are fragments, derivatives,
analogs, or ts of the foregoing polypeptides, and any combination thereof. The
terms "fragment," "variant," "derivative" and "analog" when referring to antibodies or
antibody polypeptides of the present invention include any polypeptides which retain at
least some of the antigen-binding properties of the corresponding native binding
molecule, antibody, or polypeptide. Fragments of polypeptides of the present ion
include proteolytic fragments, as well as deletion fragments, in addition to specific
antibody fragments discussed elsewhere herein. Variants of antibodies and antibody
polypeptides of the t invention include nts as described above, and also
polypeptides with altered amino acid ces due to amino acid substitutions,
deletions, or insertions. Variants can occur naturally or be non-naturally occurring. Non-
naturally ing variants can be produced using own mutagenesis techniques.
Variant polypeptides can comprise conservative or non-conservative amino acid
substitutions, deletions or additions. Derivatives of u-synuclein specific binding
molecules, e. g., dies and antibody polypeptides of the present invention, are
polypeptides which have been altered so as to exhibit onal features not found on the
native polypeptide. Examples include fusion proteins. Variant polypeptides are also
referred to herein as "polypeptide analogs". As used herein a "derivative" of a binding
le or fragment thereof, an antibody, or an dy polypeptide refers to a subject
ptide having one or more residues chemically derivatized by reaction of a
functional side group. Also included as "derivatives" are those peptides which contain one
or more naturally occurring amino acid derivatives of the twenty standard amino acids.
For example, oxyproline can be substituted for proline; 5-hydroxylysine can be
substituted for lysine; 3-methylhistidine can be substituted for histidine; homoserine can
be substituted for serine; and omithine can be substituted for lysine.
The term "polynucleotide" is intended to encompass a singular nucleic acid as
well as plural nucleic acids, and refers to an isolated nucleic acid molecule or construct,
e. g., messenger RNA (mRNA) or plasmid DNA . A polynucleotide can comprise
a conventional phosphodiester bond or a non-conventional bond (e.g., an amide bond,
such as found in peptide nucleic acids (PNA)). The term "nucleic acid" refers to any one
or more nucleic acid segments, e.g., DNA or RNA fragments, present in a polynucleotide.
By ted" nucleic acid or cleotide is intended a nucleic acid molecule, DNA or
RNA, which has been removed from its native environment. For example, a recombinant
polynucleotide encoding an dy contained in a vector is considered isolated for the
purposes of the present invention. Further examples of an isolated polynucleotide include
recombinant polynucleotides maintained in heterologous host cells or purified (partially
or substantially) polynucleotides in solution. Isolated RNA molecules include in vivo or
in vitro RNA transcripts of polynucleotides of the present invention. Isolated
polynucleotides or nucleic acids according to the present invention r include such
les produced synthetically. In on, a polynucleotide or a nucleic acid can be
or can include a regulatory element such as a er, ribosome binding site, or a
transcription terminator.
As used herein, a "coding region" is a portion of nucleic acid which ts of
codons ated into amino acids. Although a "stop codon" (TAG, TGA, or TAA) is not
translated into an amino acid, it can be considered to be part of a coding , but any
flanking ces, for example ers, ribosome binding sites, transcriptional
terminators, introns, and the like, are not part of a coding region. Two or more coding
s of the present invention can be present in a single polynucleotide construct, e.g.,
on a single vector, or in separate polynucleotide constructs, e.g., on separate (different)
vectors. Furthermore, any vector can contain a single coding region, or comprise two or
more coding regions, e.g., a single vector can separately encode an immunoglobulin
heavy chain variable region and an immunoglobulin light chain variable region. In
addition, a vector, polynucleotide, or nucleic acid of the invention can encode
heterologous coding regions, either fused or unfiJsed to a nucleic acid encoding a binding
molecule, an antibody, or nt, variant, or derivative thereof. Heterologous coding
regions include without tion specialized elements or motifs, such as a secretory
signal peptide or a heterologous fianctional domain.
In certain embodiments, the polynucleotide or nucleic acid is DNA. In the case of
DNA, a polynucleotide comprising a nucleic acid which encodes a polypeptide can
include a promoter and/or other transcription or translation control elements operably
associated with one or more coding regions. An operable association is when a coding
region for a gene product, e.g., a polypeptide, is associated with one or more regulatory
sequences in such a way as to place expression of the gene product under the ce or
control of the regulatory sequence(s). Two DNA fragments (such as a polypeptide coding
region and a promoter ated therewith) are "operably associated" or "operably
linked" if induction of promoter function results in the transcription of mRNA encoding
the d gene product and if the nature of the linkage between the two DNA fragments
does not interfere with the ability of the expression regulatory sequences to direct the
expression of the gene product or interfere with the ability of the DNA template to be
transcribed. Thus, a promoter region would be ly associated with a nucleic acid
encoding a polypeptide if the er was capable of effecting transcription of that
nucleic acid. The promoter can be a pecif1c promoter that s substantial
transcription of the DNA only in predetermined cells. Other transcription control
elements, besides a promoter, for example enhancers, operators, repressors, and
transcription ation signals, can be operably associated with the polynucleotide to
direct pecific transcription. Suitable promoters and other transcription control
regions are disclosed herein.
A variety of transcription control regions are known to those skilled in the art.
These include, without limitation, transcription control regions which function in
vertebrate cells, such as, but not d to, promoter and er segments from
galoviruses (the immediate early promoter, in ction with -A), simian
Virus 40 (the early promoter), and retroviruses (such as Rous sarcoma Virus). Other
transcription control regions include those d from vertebrate genes such as actin,
heat shock protein, bovine growth hormone and rabbit B-globin, as well as other
sequences capable of controlling gene sion in eukaryotic cells. Additional suitable
transcription control regions include tissue-specific promoters and enhancers as well as
lymphokine-inducible promoters (e.g., promoters inducible by interferons or
interleukins).
W0 2012/177972 PCT/U82012/043701
_ 15 _
Similarly, a variety of translation control elements are known to those of ordinary
skill in the art. These include, but are not limited to ribosome binding sites, translation
initiation and termination codons, and elements derived from picomaviruses (particularly
an internal ribosome entry site, or IRES, also referred to as a CITE sequence).
In other embodiments, a polynucleotide of the present invention is RNA, for
example, in the form of messenger RNA (mRNA).
cleotide and nucleic acid coding regions of the present invention can be
associated with additional coding regions which encode secretory or signal peptides,
which direct the secretion of a polypeptide encoded by a polynucleotide of the present
ion. According to the signal hypothesis, proteins secreted by mammalian cells have
a signal peptide or secretory leader sequence which is d from the mature protein
once export of the growing protein chain across the rough asmic reticulum has
been initiated. Those of ordinary skill in the art are aware that polypeptides secreted by
vertebrate cells lly have a signal peptide fused to the N—terminus of the
ptide, which is cleaved from the complete or "full length" polypeptide to produce a
secreted or e" form of the polypeptide. In certain embodiments, the native signal
peptide, e.g., an immunoglobulin heavy chain or light chain signal e is used, or a
functional derivative of that sequence that s the ability to direct the secretion of the
polypeptide that is operably associated with it. atively, a heterologous mammalian
signal peptide, or a functional derivative thereof, can be used. For example, the wild-type
leader sequence can be substituted with the leader sequence of human tissue plasminogen
activator (TPA) or mouse B-glucuronidase.
Unless stated otherwise, the terms "disorder" and "disease" are used
interchangeably herein.
A "binding molecule" as used in the context of the present invention relates
primarily to antibodies, and nts f, but can also refer to other non-antibody
molecules that bind to u-synuclein including but not limited to hormones, receptors,
ligands, major ompatibility complex (MHC) molecules, chaperones such as heat
shock proteins (HSPs) as well as cell-cell adhesion molecules such as members of the
in, intergrin, C-type lectin, immunoglobulin (Ig) superfamilies, and synthetic
binding molecules. Thus, for the sake of clarity only and without restricting the scope of
the present invention most of the ing embodiments are discussed with respect to
PCT/U82012/043701
_ 16 _
antibodies and antibody-like molecules which represent exemplary binding molecules for
the development of therapeutic and diagnostic agents.
The terms "antibody" and "immunoglobulin" are used interchangeably herein. An
antibody or immunoglobulin is an u-synuclein-binding molecule which comprises at least
the variable domain of a heavy chain, and ly comprises at least the variable
domains of a heavy chain and a light chain. Basic globulin structures in
rate systems are relatively well understood; see, e.g., Harlow et al., Antibodies: A
Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988).
As will be discussed in more detail below, the term "immunoglobulin" ses
various broad classes of polypeptides that can be distinguished biochemically. Those
skilled in the art will iate that heavy chains are classified as gamma, mu, alpha,
delta, or epsilon, (y, u, 0t, 8, 8) with some subclasses among them (e. g., yl-y4). It is the
nature of this chain that determines the "class" of the antibody as IgG, IgM, IgA IgG, or
IgE, respectively. The immunoglobulin subclasses (isotypes) e.g., IgGl, IgG2, IgG3,
IgG4, IgAl, etc. are well characterized and are known to confer onal specialization.
Modified versions of each of these classes and es are readily discemable to the
skilled artisan in view of the t disclosure and, ingly, are within the scope of
the instant invention. All immunoglobulin classes are clearly within the scope of the
present invention, the following discussion will generally be directed to the IgG class of
immunoglobulin molecules. With regard to IgG, a rd immunoglobulin molecule
comprises two identical light chain polypeptides of molecular weight approximately
23,000 Daltons, and two identical heavy chain polypeptides of molecular weight 53,000-
70,000. The four chains are typically joined by disulfide bonds in a "Y" configuration
wherein the light chains bracket the heavy chains starting at the mouth of the "Y" and
continuing h the variable region.
Light chains are classified as either kappa or lambda (K, k). Each heavy chain
class can be bound with either a kappa or lambda light chain. In l, the light and
heavy chains are covalently bonded to each other, and the "tail" portions of the two heavy
chains are bonded to each other by covalent disulfide linkages or non-covalent linkages
when the immunoglobulins are generated either by hybridomas, B cells or genetically
engineered host cells. In the heavy chain, the amino acid sequences run from an N-
W0 2012/177972 PCT/U82012/043701
_ 17 _
terminus at the forked ends of the Y ration to the C-terminus at the bottom of each
chain.
Both the light and heavy chains are d into regions of structural and
functional homology. The terms "constant" and "variable" are used filnctionally. In this
regard, it will be appreciated that the variable domains of both the light (VL) and heavy
(VH) chain portions determine antigen recognition and specificity. Conversely, the
constant domains of the light chain (CL) and the heavy chain (CH1, CH2 or CH3) confer
important biological properties such as secretion, transplacental mobility, Fc receptor
binding, complement binding, and the like. By convention the numbering of the constant
region domains increases as they become more distal from the antigen-binding site or
terminus of the antibody. The N-terminal portion is a variable region and at the C-
terminal n is a constant ; the CH3 and CL domains actually comprise the
carboxy-terminus of the heavy and light chain, tively.
As indicated above, the variable region allows the dy to selectively
recognize and specifically bind epitopes on antigens. That is, the VL domain and VH
domain, or subset of the mentarity determining regions , of an antibody
e to form the variable region that defines a three dimensional antigen-binding site.
This quaternary antibody structure forms the antigen-binding site t at the end of
each arm of the Y. More specifically, the antigen-binding site is defined by three CDRs
on each of the VH and VL chains. Any antibody or immunoglobulin fragment which
contains ent structure to specifically bind to u-synuclein is denoted herein
hangeably as a "binding fragment" or an "immunospecific fragment."
In naturally occurring antibodies, an antibody comprises six hypervariable
regions, sometimes called "complementarity determining regions" or "CDRs" present in
each antigen-binding domain, which are short, non-contiguous sequences of amino acids
that are specifically positioned to form the antigen-binding domain as the antibody
assumes its three dimensional configuration in an aqueous environment. The "CDRs" are
flanked by four relatively conserved "framework" regions or "FRs" which show less
inter-molecular variability. The framework regions largely adopt a [3-sheet conformation
and the CDRs form loops which connect, and in some cases form part of, the B-sheet
ure. Thus, framework regions act to form a scaffold that provides for positioning the
CDRs in correct orientation by inter-chain, non-covalent interactions. The antigen-
_ 18 _
binding domain formed by the oned CDRs defines a e complementary to the
epitope on the immunoreactive antigen. This complementary surface promotes the non-
covalent binding of the antibody to its cognate epitope. The amino acids comprising the
CDRs and the framework regions, respectively, can be readily identified for any given
heavy or light chain variable region by one of ordinary skill in the art, since they have
been precisely defined; see, "Sequences of Proteins of Immunological Interest," Kabat,
E., et al., US. Department of Health and Human Services, (1983); and Chothia and Lesk,
J. Mol. Biol., 196 (1987), 901-917, which are incorporated herein by reference in their
entireties.
In the case where there are two or more definitions of a term which is used and/or
accepted within the art, the definition of the term as used herein is intended to include all
such meanings unless explicitly stated to the contrary. A specific example is the use of the
term "complementarity determining region" ("CDR") to describe the ntiguous
antigen ing sites found within the variable region of both heavy and light chain
polypeptides. This particular region has been described by Kabat et al., US. Dept. of
Health and Human Services, "Sequences of Proteins of logical Interest" (1983)
and by Chothia and Lesk, J. Mol. Biol., 196 (1987), 901-917, which are incorporated
herein by reference, where the definitions include overlapping or s of amino acid
residues when compared against each other. Nevertheless, application of either ion
to refer to a CDR of an antibody or ts thereof is intended to be within the scope of
the term as defined and used herein. The appropriate amino acid residues which
encompass the CDRs as defined by each of the above cited references are set forth below
in Table 1 as a comparison. The exact e numbers which encompass a particular
CDR will vary depending on the sequence and size of the CDR. Those skilled in the art
can routinely ine which residues comprise a particular hypervariable region or
CDR of the human IgG subtype of antibody given the le region amino acid
sequence of the antibody.
Table 1: CDR Definitions1
VH CDRl
VH CDR2
VH CDR3
VL CDRl 24-34 26-32
PCT/U82012/043701
_ 19 _
VL CDR2 50-56 50-52
VL CDR3 89-97 91-96
1Numbering of all CDR definitions in Table l is according to the numbering conventions set
forth by Kabat et al. (see below).
Kabat et al. also defined a numbering system for variable domain sequences that
is applicable to any antibody. One of ordinary skill in the art can unambiguously assign
this system of "Kabat numbering" to any variable domain sequence, without reliance on
any mental data beyond the sequence itself. As used herein, "Kabat numbering"
refers to the numbering system set forth by Kabat et al., US. Dept. of Health and Human
Services, nce of Proteins of Immunological Interest" (1983). Unless otherwise
specified, references to the numbering of specific amino acid residue positions in an
antibody or antigen-binding fragment, variant, or tive thereof of the present
invention are according to the Kabat numbering system.
dies or antigen-binding nts, immunospecific fragments, variants, or
derivatives thereof of the invention include, but are not limited to, polyclonal,
monoclonal, multispecific, human, humanized, primatized, murinized or chimeric
antibodies, single chain dies, epitope-binding fragments, e.g., Fab, Fab' and F(ab')2,
Fd, Fvs, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (dev),
fragments comprising either a VL or VH domain, fragments produced by a Fab
expression library, and anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id
antibodies to antibodies disclosed herein). ScFv molecules are known in the art and are
described, e. g., in US patent 5,892,019. Immunoglobulin or dy molecules of the
invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgGl,
IgG2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
In one embodiment, the antibody of the present invention is not IgM or a
derivative thereof with a pentavalent structure. Particular, in c applications of the
present invention, especially eutic use, Ing are less useful than IgG and other
bivalent antibodies or ponding binding les since Ing due to their
pentavalent structure and lack of affinity maturation often show unspecific cross-
reactivities and very low affinity.
In one embodiment, the antibody of the present invention is not a onal
dy, z'.e. it substantially consists of one particular dy species rather than being
a mixture obtained from a plasma immunoglobulin sample.
PCT/U82012/043701
_ 20 _
Antibody fragments, including single-chain antibodies, can comprise the le
region(s) alone or in combination with the ty or a portion of the following: hinge
, CH1, CH2, and CH3 domains. Also included in the invention are u-synuclein-
binding fragments also comprising any combination of variable region(s) with a hinge
region, CH1, CH2, and CH3 domains. Antibodies or specif1c fragments thereof
of the present invention can be from any animal origin ing birds and mammals. In
certain embodiments, the dies are human, murine, donkey, rabbit, goat, guinea pig,
camel, llama, horse, or n antibodies. In another embodiment, the variable region
can be condricthoid in origin (6.g. from sharks).
In one aspect, the antibody of the present invention is a human monoclonal
antibody isolated from a human. Optionally, the framework region of the human antibody
is aligned and adopted in accordance with the pertinent human germ line variable region
sequences in the database; see, e.g., Vbase (vbase.mrc-cpe.cam.ac.uk) hosted by the MRC
Centre for Protein Engineering (Cambridge, UK). For example, amino acids considered to
potentially deviate from the true germ line sequence could be due to the PCR primer
sequences incorporated during the cloning process. Compared to artificially generated
like antibodies such as single chain antibody fragments ) from a phage
displayed antibody library or xenogeneic mice the human monoclonal dy of the
present invention is characterized by (i) being obtained using the human immune
response rather than that of animal surrogates, z'.e. the antibody has been generated in
response to natural u-synuclein in its relevant conformation in the human body, (ii)
having protected the individual or is at least icant for the presence of u-synuclein,
and (iii) since the antibody is of human origin the risks of cross-reactivity against self-
antigens is minimized. Thus, in accordance with the present invention the terms "human
monoclonal antibody", "human monoclonal autoantibody", "human antibody" and the like
are used to denote an u-synuclein binding molecule which is of human origin, z'.e. which
has been isolated from a human cell such as a B cell or hybridoma thereof or the cDNA of
which has been directly cloned from mRNA of a human cell, for example a human
memory B cell. A human antibody is still "human" even if amino acid tutions are
made in the antibody, 6.g. to e binding characteristics.
Antibodies d from human immunoglobulin libraries or from animals
transgenic for one or more human immunoglobulins and that do not express endogenous
PCT/U82012/043701
_ 21 _
immunoglobulins, as described infra and, for example in, US patent no 5,939,598 by
Kucherlapati et al., are denoted human-like antibodies in order distinguish them from
truly human antibodies of the present invention.
As used herein, the term ized antibody" or "murinized immunoglobulin"
refers to an dy comprising one or more CDRs from a human antibody of the present
invention; and a human framework region that contains amino acid substitutions and/or
deletions and/or insertions that are based on a mouse antibody sequence. The human
immunoglobulin providing the CDRs is called the "paren " or "acceptor" and the mouse
antibody ing the framework changes is called the "donor". nt regions need
not be present, but if they are, they are usually substantially identical to mouse antibody
constant regions, z'.e. at least about 85- 90%, or about 95% or more identical. Hence, in
some embodiments, a filll length murinized human heavy or light chain immunoglobulin
contains a mouse constant region, human CDRs, and a substantially human framework
that has a number of "murinizing" amino acid substitutions. Typically, a ized
antibody" is an antibody comprising a zed variable light chain and/or a zed
variable heavy chain. For example, a murinized antibody would not encompass a typical
chimeric antibody, e.g., because the entire variable region of a chimeric antibody is non-
mouse. A modified antibody that has been "murinized" by the s of "murinization"
binds to the same antigen as the parent antibody that provides the CDRs and is usually
less immunogenic in mice, as compared to the parent antibody.
As used herein, the term "heavy chain n" includes amino acid sequences
derived from an immunoglobulin heavy chain. A polypeptide sing a heavy chain
portion comprises at least one of: a CHl domain, a hinge (e.g., upper, middle, and/or
lower hinge region) domain, a CH2 domain, a CH3 , or a variant or nt
thereof. For example, a binding polypeptide for use in the invention can comprise a
polypeptide chain comprising a CHl domain; a polypeptide chain comprising a CHl
domain, at least a portion of a hinge domain, and a CH2 domain; a polypeptide chain
comprising a CHl domain and a CH3 domain; a polypeptide chain comprising a CHl
, at least a portion of a hinge domain, and a CH3 domain, or a polypeptide chain
comprising a CHl domain, at least a portion of a hinge domain, a CH2 domain, and a
CH3 domain. In another embodiment, a polypeptide of the invention comprises a
polypeptide chain comprising a CH3 domain. Further, a g polypeptide for use in
PCT/U82012/043701
_ 22 _
the invention can lack at least a portion of a CH2 domain (e.g., all or part of a CH2
). As set forth above, it will be understood by one of ordinary skill in the art that
these domains (e.g., the heavy chain portions) can be modified such that they vary in
amino acid sequence from the naturally occurring immunoglobulin molecule.
In certain antibodies, or antigen-binding fragments, variants, or derivatives thereof
disclosed herein, the heavy chain portions of one polypeptide chain of a er are
identical to those on a second polypeptide chain of the multimer. Alternatively, heavy
chain portion-containing monomers of the invention are not identical. For e, each
monomer can comprise a different target binding site, forming, for example, a bispecific
antibody or diabody.
In another embodiment, the antibodies, or antigen-binding nts, variants, or
derivatives thereof disclosed herein are composed of a single polypeptide chain such as
scFvs and are to be expressed intracellularly (intrabodies) for potential in viva therapeutic
and stic applications.
The heavy chain portions of a binding polypeptide for use in the diagnostic and
ent methods disclosed herein can be derived from different immunoglobulin
molecules. For e, a heavy chain portion of a ptide can comprise a CHl
domain derived from an IgGl molecule and a hinge region derived from an IgG3
molecule. In another example, a heavy chain portion can se a hinge region derived,
in part, from an IgGl molecule and, in part, from an IgG3 molecule. In another example,
a heavy chain portion can comprise a chimeric hinge derived, in part, from an IgGl
molecule and, in part, from an IgG4 molecule.
As used herein, the term "light chain portion" includes amino acid sequences
derived from an immunoglobulin light chain. In one embodiment, the light chain portion
comprises at least one of a VL or CL domain.
The m size of a e or ptide epitope for an antibody is thought
to be about four to five amino acids. Peptide or polypeptide epitopes can contain, for
e, at least seven, at least nine, or between at least about 15 to about 30 amino
acids. Since a CDR can recognize an antigenic peptide or polypeptide in its tertiary form,
the amino acids comprising an epitope need not be contiguous, and in some cases, may
not even be on the same peptide chain. In the present invention, a peptide or polypeptide
epitope recognized by antibodies of the present invention contains a sequence of at least
PCT/U82012/043701
_ 23 _
4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at
least 25, or between about 15 to about 30 contiguous or non-contiguous amino acids of 0L-
synuclein. As used herein, when an antibody is said to bind "within" a given range of
amino acids, e.g., bind to an epitope "within amino acids 4 to 15 of u-synuclein," it is
meant that the epitope encompasses the full range of stated amino acids or is smaller. In
other words, for an e "within amino acids 4 to 15 of u-synuclein," the epitope can
include the entire 12-amino acid peptide chain of 4 to 15, but can also be smaller, e.g.,
amino acids 4 to 12, amino acids 4 to 10 or amino acids 4 to 8. The person of ordinary
skill in the art will also recognize that amino acids outside of the stated range may
contribute to better binding affinity or increased recognition of a conformational epitope,
but are not required for binding.
By "specifically binding", or "specifically recognizing", used interchangeably
herein, it is generally meant that a binding molecule, e.g., an antibody binds to an epitope
via its n-binding domain, and that the binding entails some complementarity
between the antigen-binding domain and the epitope. According to this definition, an
antibody is said to "specifically bind" to an epitope when it binds to that epitope, via its
antigen-binding domain more readily than it would bind to a , unrelated e.
The term "specificity" is used herein to qualify the relative affinity by which a certain
antibody binds to a certain e. For example, antibody "A" can be deemed to have a
higher specificity for a given epitope than antibody "B," or antibody "A" can be said to
bind to e "C" with a higher specificity than it has for related epitope "D".
Where present, the term ological g characteristics," or other binding
characteristics of an antibody with an n, in all of its grammatical forms, refers to the
specificity, affinity, cross-reactivity, and other binding characteristics of an antibody.
By "preferentially binding", it is meant that the binding molecule, e.g., antibody
specifically binds to an epitope more readily than it would bind to a related, r,
homologous, or analogous epitope. Thus, an dy which "preferentially binds" to a
given epitope would more likely bind to that epitope than to a related epitope, even
though such an antibody can cross-react with the related epitope.
By way of non-limiting example, a binding molecule, e.g., an antibody binds a
first epitope preferentially if it binds said first epitope with a dissociation nt (KD)
that is less than the antibody’s KD for the second epitope. In another non-limiting
e, an antibody binds a first antigen entially if it binds the first epitope with
an affinity that is at least one order of magnitude less than the antibody’s KD for the
second e. In another miting example, an antibody binds a first epitope
preferentially if it binds the first epitope with an affinity that is at least two orders of
magnitude less than the antibody’s KD for the second epitope.
In another miting example, a binding molecule, e.g., an antibody binds a
first epitope preferentially if it binds the first epitope with an off rate (k(off)) that is less
than the antibody’s k(off) for the second epitope. In another non-limiting example, an
antibody binds a first epitope preferentially if it binds the first epitope with an y that
is at least one order of magnitude less than the antibody’s k(off) for the second epitope. In
another non-limiting example, an antibody binds a first epitope preferentially if it binds
the first epitope with an affinity that is at least two orders of magnitude less than the
antibody’s k(off) for the second epitope.
In some embodiments binding molecule, e.g, an antibody or antigen-binding
nt, variant, or derivative disclosed herein can bind a u-synuclein or a fragment or
variant f with an off rate (k(off)) of less than or equal to 5 X 10'2 sec'l, 10'2 sec'l, 5
X 10'3 sec"1 or 10'3 sec'l. In some ments, an antibody of the invention can bind a-
synuclein or a fragment or t thereof with an off rate (k(off)) less than or equal to 5
X 10'4 sec'l, 10'4 sec'l, 5 X 10'5 sec'l, or 10'5 sec"1 5 X 10'6 sec'l, 10'6 sec'l, 5 X 10'7 sec"1
or 10'7 sec'l.
In certain embodiments, a g molecule, e.g., an antibody or antigen-binding
fragment, variant, or derivative disclosed herein can bind u-synuclein or a fragment or
variant thereof with an on rate (k(on)) of greater than or equal to 103 M"1 sec'l, 5 X 103 M"
1 sec'l, 104 M"1 sec"1 or 5 X 104 M"1 sec'l. In some embodiments, an antibody of the
invention can bind u-synuclein or a fragment or t thereof with an on rate (k(on))
greater than or equal to 105 M"1 sec'l, 5 X 105 M"1 sec'l, 106 M"1 sec'l, or 5 X 106 M"1 sec"1
or 107 M"1 sec'l.
A binding molecule, 6.g. an antibody is said to itively inhibit binding of a
reference antibody to a given epitope if it preferentially binds to that epitope to the extent
that it blocks, to some degree, binding of the reference antibody to the epitope.
Competitive inhibition can be determined by any method known in the art, for example,
competition ELISA assays. As an example, an antibody can competitively inhibit binding
W0 2012/177972 2012/043701
_ 25 _
of the reference dy to a given epitope by at least 90%, at least 80%, at least 70%, at
least 60%, or at least 50%.
As used herein, the term "affinity" refers to a measure of the strength of the
binding of an individual epitope with the CDR of a binding molecule, e.g., an
immunoglobulin molecule; see, e.g., Harlow et al., Antibodies: A Laboratory Manual,
Cold Spring Harbor Laboratory Press, 2nd ed. (1988) at pages 27-28. As used herein, the
term "avidity" refers to the overall stability of the complex between a population of
immunoglobulins and an antigen, that is, the onal combining th of an
immunoglobulin mixture with the antigen; see, e.g., Harlow at pages 29-34. Avidity is
related to both the affinity of dual immunoglobulin molecules in the tion with
specific epitopes, and also the valencies of the immunoglobulins and the n. For
example, the ction between a bivalent monoclonal antibody and an n with a
highly repeating epitope structure, such as a polymer, would be one of high avidity. The
affinity or avidity of an antibody for an n can be determined experimentally using
any suitable method; see, for example, Berzofsky et al., "Antibody-Antigen Interactions"
In Fundamental Immunology, Paul, W. E., Ed., Raven Press New York, N Y (1984),
Kuby, Janis Immunology, W. H. Freeman and Company New York, N Y (1992), and
s described herein. General techniques for measuring the affinity of an antibody
for an antigen include ELISA, RIA, and surface plasmon resonance. The measured
affinity of a particular antibody-antigen interaction can vary if ed under different
conditions, e.g., salt concentration, pH. Thus, measurements of affinity and other n-
binding parameters, e.g., KD, IC50, can be made with standardized solutions of antibody
and antigen, and a standardized buffer.
Binding molecules, e.g., antibodies or antigen-binding fragments, variants or
derivatives thereof of the invention can also be described or specified in terms of their
cross-reactivity. As used herein, the term "cross-reactivity" refers to the ability of an
antibody, specific for one antigen, to react with a second antigen; a measure of
relatedness between two different antigenic substances. Thus, an antibody is cross
reactive if it binds to an epitope other than the one that induced its formation. The cross
reactive e generally contains many of the same complementary structural features
as the inducing epitope, and in some cases, can actually fit better than the original.
_ 26 _
For example, certain antibodies have some degree of cross-reactivity, in that they
bind related, but non-identical epitopes, e.g., epitopes with at least 95%, at least 90%, at
least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least
55%, and at least 50% identity (as calculated using methods known in the art and
described herein) to a nce epitope. In some ments, an antibody can be said to
have little or no cross-reactivity if it does not bind epitopes with less than 95%, less than
90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than
60%, less than 55%, and less than 50% identity (as calculated using methods known in
the art and described herein) to a reference epitope. An antibody can be deemed "highly
specific" for a certain epitope, if it does not bind any other analog, ortholog, or homolog
of that epitope.
Binding molecules, e.g., dies or antigen-binding fragments, variants or
derivatives thereof of the invention can also be described or specified in terms of their
binding affinity to u-synuclein. Binding affinities include those with a dissociation
constant or Kd less than 5 x 10‘2 M, 10‘2 M, 5 x 10‘3 M, 10‘3 M, 5 x 10'4M, 10‘4 M, 5 x 10‘5
M, 10‘5 M, 5 x 10'6M, 10‘6 M, 5 X10'7M,10'7M,5 x 10‘8 M, 10'8M, 5 x 10'9M, 10‘9 M, 5
x 10'10M, 10'10M, 5 x 10‘11 M, 10‘11 M, 5 x10'12M,10'12M,5 x 10‘13 M, 10'13M, 5 x10—
14M, 10'14M, 5 x 10‘15 M, or 10‘15 M.
As previously indicated, the subunit structures and three dimensional
configuration of the nt regions of the various immunoglobulin classes are well
known. As used herein, the term "VH domain" includes the amino terminal variable
domain of an immunoglobulin heavy chain and the term "CHl domain" includes the first
(most amino terminal) constant region domain of an immunoglobulin heavy chain. The
CHl domain is nt to the VH domain and is amino terminal to the hinge region of an
immunoglobulin heavy chain molecule.
As used herein the term "CH2 domain" includes the portion of a heavy chain
molecule that extends, e. g., from about residue 244 to residue 360 of an antibody using
conventional numbering s (residues 244 to 360, Kabat ing system; and
es 231-340, EU numbering system; see Kabat EA et al. op. cit). The CH2 domain is
unique in that it is not closely paired with another . Rather, two N—linked branched
ydrate chains are interposed n the two CH2 domains of an intact native IgG
W0 2012/177972
_ 27 _
molecule. It is also well documented that the CH3 domain extends from the CH2 domain
to the C-terminal of the IgG molecule and ses approximately 108 residues.
As used herein, the term "hinge region" includes the n of a heavy chain
le that joins the CH1 domain to the CH2 domain. This hinge region comprises
approximately 25 residues and is flexible, thus allowing the two N-terminal antigen-
binding regions to move independently. Hinge regions can be subdivided into three
distinct domains: upper, , and lower hinge domains; see Roux et al., J. Immunol.
161 (1998), 4083.
As used herein the term "disulfide bond" includes the covalent bond formed
between two sulfur atoms. The amino acid cysteine comprises a thiol group that can form
a de bond or bridge with a second thiol group. In most naturally occurring IgG
molecules, the CH1 and CL regions are linked by a disulfide bond and the two heavy
chains are linked by two disulfide bonds at positions ponding to 239 and 242 using
the Kabat numbering system (position 226 or 229, EU numbering system).
As used herein, the terms "linked," "fused" or n" are used interchangeably.
These terms refer to the joining together of two more elements or components, by
whatever means ing chemical conjugation or recombinant means. An "in-frame
fusion" refers to the joining of two or more polynucleotide open reading frames (ORFs)
to form a uous longer ORF, in a manner that maintains the correct translational
reading frame of the original ORFs. Thus, a recombinant fusion n is a single protein
containing two or more segments that correspond to polypeptides encoded by the original
ORFs (which segments are not normally so joined in nature). gh the reading frame
is thus made continuous throughout the fused segments, the segments can be physically or
spatially separated by, for e, in-frame linker sequence. For example,
polynucleotides encoding the CDRs of an immunoglobulin variable region can be filSGd,
in-frame, but be separated by a polynucleotide encoding at least one immunoglobulin
framework region or additional CDR regions, as long as the "filSGd" CDRs are co-
translated as part of a continuous polypeptide.
The term "expression" as used herein refers to a process by which a gene produces
a biochemical, for example, an RNA or polypeptide. The process includes any
manifestation of the fianctional presence of the gene within the cell including, without
limitation, gene knockdown as well as both transient sion and stable expression. It
PCT/U82012/043701
_ 28 _
includes t limitation transcription of the gene into messenger RNA (mRNA),
transfer RNA (tRNA), small hairpin RNA (shRNA), small interfering RNA ) or
any other RNA product, and the translation of such mRNA into polypeptide(s). If the
final desired product is a biochemical, expression includes the creation of that
biochemical and any precursors. Expression of a gene produces a "gene product." As used
herein, a gene product can be either a nucleic acid, e. g., a messenger RNA produced by
transcription of a gene, or a polypeptide which is translated from a transcript. Gene
products described herein fiarther include nucleic acids with post riptional
modifications, e.g., enylation, or polypeptides with post translational
modifications, e.g., methylation, glycosylation, the addition of lipids, association with
other protein subunits, proteolytic cleavage, and the like.
As used herein, the term "sample" refers to any biological material obtained from
a subject or patient. In one aspect, a sample can comprise blood, cerebrospinal fluid
("CSF"), or urine. In other aspects, a sample can comprise whole blood, plasma, B cells
enriched from blood samples, and cultured cells (e.g., B cells from a t). A sample
can also include a biopsy or tissue sample including neural . In still other aspects, a
sample can comprise whole cells and/or a lysate of the cells. Blood samples can be
collected by methods known in the art. In one aspect, the pellet can be resuspended by
vortexing at 4°C in 200 ul buffer (20 mM Tris, pH. 7.5, 0.5% Nonidet, 1 mM EDTA, 1
mM PMSF, 0.1M NaCl, IX Sigma Protease Inhibitor, and IX Sigma atase
Inhibitors l and 2). The suspension can be kept on ice for 20 minutes with intermittent
vortexing. After spinning at 15,000 x g for 5 minutes at about 4°C, aliquots of supernatant
can be stored at about -70°C.
As used herein, the terms " or "treatment" refer to both therapeutic treatment
and prophylactic or preventative measures, wherein the object is to prevent or slow down
(lessen) an undesired physiological change or disorder, such as the development of
sonism. ial or desired clinical results include, but are not limited to,
alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not
worsening) state of disease, delay or g of disease ssion, ration or
palliation of the disease state, and ion (whether partial or total), whether able
or undetectable. "Treatment" can also mean prolonging survival as compared to expected
survival if not receiving treatment. Those in need of treatment include those already with
PCT/U82012/043701
_ 29 _
the condition or disorder as well as those prone to have the condition or disorder or those
in which the manifestation of the condition or disorder is to be prevented.
By ct" or "individual" or "animal" or nt" or l,” is meant any
subject, particularly a mammalian subject, e.g., a human patient, for whom diagnosis,
prognosis, prevention, or therapy is desired.
11. Antibodies
The present ion generally relates to human anti-(x-synuclein antibodies and
n-binding fragments thereof, which can demonstrate the immunological g
characteristics and/or biological properties as outlined for the dies illustrated in the
Examples. In accordance with the present invention human monoclonal dies
specific for u-synuclein were cloned from a pool of aged subjects.
In the course of the experiments performed in accordance with the present
invention initial attempts failed to clone u-synuclein specific antibodies but almost always
resulted in false-positive clones. Further investigation of these clones revealed that they
produced antibodies recognizing proteins of E. 0012'. In order to circumvent this problem,
antibodies in conditioned media of human memory B cell cultures were screened in
parallel for binding to coated fiJll-length alpha synuclein monomer and e of
g to E. 0011'. proteins and bovine serum albumin (BSA). In particular, B cell
conditioned medium was preabsorbed with E. coli proteins prior to subjecting the
medium to an ELISA assay for screening of u-synuclein binding human antibodies.
Initial attempts at isolating specific antibodies were focused at pools of human
subjects with high plasma binding activity to u-synuclein, tive of elevated levels of
circulating u-synuclein antibodies plasma. These attempts failed to produce u-synuclein
specific human memory B cells and the antibodies described in the current invention were
isolated from pools of subjects with low plasma reactivity to u-synuclein.
Due to this measure, several antibodies were isolated. Selected dies were
further analyzed for class and light chain subclass determination. Selected relevant
antibody messages from memory B cell cultures are then transcribed by RT-PCR, cloned
and combined into expression vectors for recombinant production; see PCT Publication
No. Al. Exemplary anti-human clein antibodies NI-202.12F4,
NI-202.3G12, and NI-202.3D8 are disclosed in PCT Publication No.
W0 2012/177972
Disclosed herein is human monoclonal antibody NI-202.22Dll. Recombinant
expression of NI-202.22Dll in HEK293 or CHO cells and subsequent characterization of
its binding specificity for human u-synuclein (Fig. 2A-B) was determined. Thus, one
aspect of the present invention relates to the isolated human monoclonal anti-(x-synuclein
antibody NI-202.22Dll and n-binding fragments, derivatives and variants thereof.
The present invention is also drawn to a binding molecule such as an antibody, or
antigen-binding fragment, variant or derivatives thereof, where the antibody comprises a
VH with the amino acid sequence of SEQ ID N014 or SEQ ID N020, and a VL with the
amino acid sequence of SEQ ID N022 or SEQ ID NO:26, or n-binding fragments,
ts or derivatives f. In one embodiment, NI-202.22Dl l, as well as variants,
fragments, or derivatives thereof are terized as specifically binding human 0L-
synuclein compared to human B-synuclein and human y-synuclein, and to human (1-
synuclein as compared to murine u-synuclein. .22D11 preferentially binds to usynuclein
in the oligomeric or aggregated form.
In one ment, the present invention is directed to an anti-(x-synuclein
antibody, or antigen-binding fragment, variant or derivatives thereof, where the antibody
specifically binds to the same epitope of u-synuclein as the reference antibody NI-
202.22Dl 1. As illustrated in the Examples, antibody .22Dll binds to u-synuclein
truncations containing the inal acidic region (amino acids 96-140) in a direct
ELISA assay, e.g., within amino acids 113 to 123 of SEQ ID NO:l, and specifically binds
to an epitope within the amino acids PVDPDNE (amino acids 117-123 of SEQ ID NO: 1).
Antibody NI-202.22Dll preferentially binds to u-synuclein aggregates or fibrils
over the monomeric form of u-synuclein as shown in Example 2. Furthermore, antibody
NI-202.22Dll binds to pathological forms of u-synuclein in brain, e.g. pathological
aggregates of u-synuclein as exemplified by histochemical staining described in
Example 3. Hence, the present ion provides a new human anti-(x-synuclein
antibody useful for diagnostic and therapeutic purposes.
In one embodiment, the present invention provides binding les, e.g.,
antibodies or antigen-binding nts, variants, or derivatives thereof which exhibit the
precise binding ties of the ary NI-202.l2F4 antibody as described in PCT
Publication No. Al. The present invention provides binding molecules
which bind to an epitope at the N-terminus of u-synuclein, e. g., binding molecules which
PCT/U82012/043701
_ 31 _
bind within amino acids 4 to 15 of SEQ ID NO:1. n embodiments provide binding
molecules, e.g., dies or antigen-binding fragments, variants or derivatives thereof,
which bind within amino acids 4 to 15 of SEQ ID NO:1, but excluding antibodies
comprising a VH (SEQ ID N05 or SEQ ID NO:9), VL (SEQ ID NO:lO or SEQ ID
NO:14), VHCDRI (SEQ ID NO:6), VHCDR2 (SEQ ID NO:7), VHCDR3 (SEQ
IDNO:8), VLCDRI (SEQ ID NO:11), VLCDR2 (SEQ ID NO:12) and/or VLCDR3 (SEQ
ID NO: 13) ofNI-202. 12F4, or fragments, ts, or derivatives thereof
The present invention fithher provides binding molecules, e. g. antibodies and
antigen-binding fragments, variants, or derivatives thereof, which ses at least one,
two, three, four, five, or six mentarity determining regions (CDRs) of a NI-
202.22Dll VH and/or VL variable region comprising any one of the amino acid
sequences depicted in Fig. l. The corresponding nucleotide ces encoding the
above-identified variable regions are set forth in the attached sequence listing. An
exemplary set of CDRs of the above amino acid sequences of the VH and/or VL region as
depicted in Fig. 1 is also indicated in the appended sequence listing. However, as
discussed in the following the person skilled in the art is well aware of the fact that in
addition or alternatively CDRs can be used, which differ in their amino acid sequence
from those set forth in Fig. l by one, two, three, four, five, or more amino acids. The VH
of NI-202.22D11 is ented by amino acid sequence SEQ ID NO:15 and DNA
ce SEQ ID NO:l9, and its GL-corrected form is represented as amino acid
sequence SEQ ID N020 and DNA sequence SEQ ID NO:21. The VL of NI-202.22D11
is represented by amino acid ce SEQ ID N022 and DNA sequence SEQ ID
N028, and its GL-corrected form is represented as amino acid sequence SEQ ID N026
and DNA sequence SEQ ID NO:27. The heavy chain CDR amino acid sequences of VH-
CDRl, VH-CDR2 and VH-CDR3 are represented by SEQ ID NOl6, SEQ ID NO:l7, and
SEQ ID NO:18, respectively. The light chain CDR amino acid sequences of VL-CDRl,
VL-CDR2 and VL-CDR3 are represented by SEQ ID N023, SEQ ID N024, and SEQ
ID NO:25, respectively.
In one embodiment, a binding molecule, e.g., an antibody or antigen g
nt, variant, or derivative thereof of the present invention is any one of the
antibodies comprising an amino acid sequence of the VH and/or VL region as depicted in
Fig. 1. Alternatively, the antibody of the present invention is a binding molecule, e.g., an
antibody or antigen binding fragment, variant, or derivative thereof which competes for
binding to (x-synuclein with an antibody having a VH and/or VL region as depicted in
Fig. 1. Those antibodies can be human as well, in particular for therapeutic applications.
Alternatively, the antibody is a murine, murinized and chimeric murine-human antibody,
which are particularly useful for diagnostic methods and studies in animals.
As mentioned above, due to its generation upon a human immune response the
human monoclonal antibody of the present invention will recognize epitopes which are of
particular physiological relevance and which might not be accessible or less
genic in case of immunization processes for the generation of for example mouse
monoclonal antibodies and in in vitro screening of phage display libraries, respectively.
Accordingly, an epitope of a human anti-(x-synuclein antibody of the present invention
can be unique. Therefore, the present invention also extends lly to anti-(x-synuclein
antibodies and (x-synuclein binding molecules which compete with the human
monoclonal dy of the present invention for specific binding to (x-synuclein. The
present invention is more specifically directed to a binding molecule, e.g., an dy, or
antigen-binding fragment, variant or tives f, where the antibody specifically
binds to the same e of (x-synuclein as the reference antibody NI-202.22G11.
ition between antibodies can be determined, for e, by an assay in
which the immunoglobulin under test ts specific binding of a reference antibody to
a common antigen, such as u-synuclein. Numerous types of competitive binding assays
are known, for example: solid phase direct or indirect radioimmunoassay (RIA), solid
phase direct or indirect enzyme immunoassay (EIA), sandwich ition assay; see
Stahli et al., Methods in Enzymology 9 (1983), 242-253; solid phase direct biotin-avidin
EIA; see Kirkland et al., J. Immunol. 137 , 3614-3619 and Cheung et al., Virology
176 , 546-552; solid phase direct labeled assay, solid phase direct labeled sandwich
assay; see Harlow and Lane, dies, A Laboratory Manual, Cold Spring Harbor Press
(1988); solid phase direct label RIA using I125 label; see Morel et a], Molec. Immunol. 25
(1988), 7-15 and Moldenhauer et al., Scand. J. Immunol. 32 , 77-82. Typically,
such an assay es the use of purified (x-synuclein or aggregates thereof bound to a
solid surface or cells bearing either of these, an unlabelled test immunoglobulin and a
labeled reference immunoglobulin, z'.e. a human monoclonal antibody of the present
invention. Competitive inhibition is measured by determining the amount of label bound
PCT/U82012/043701
_ 33 _
to the solid surface or cells in the presence of the test immunoglobulin. Usually the test
immunoglobulin is present in excess. A itive binding assay can be performed
under conditions as described for the ELISA assay in the appended es. Antibodies
identified by competition assay (competing antibodies) include antibodies binding to the
same epitope as the reference antibody and antibodies binding to an adjacent epitope
sufficiently proximal to the epitope bound by the reference antibody for steric hindrance
to occur. Usually, when a competing antibody is present in excess, it will inhibit specific
g of a reference antibody to a common antigen by at least 50% or 75%. Hence, the
present invention is further drawn to a binding molecule, e.g., an antibody, or antigen-
binding fragment, variant or derivatives f, where the antibody competitively
inhibits the reference antibody NI-202.22Gll from binding to (x-synuclein.
Also disclosed is an isolated binding molecule, e.g., an dy or antigen-
binding fragment thereof which specifically binds to human (x-synuclein, comprising an
immunoglobulin heavy chain variable region (VH) amino acid ce at least 80%,
85%, 90% 95% or 100% identical to SEQ ID NO: 15 or SEQ ID NO:20.
Further disclosed is an isolated binding molecule, e. g., an antibody or antigen-
binding fragment thereof which specifically binds to human uclein, comprising a
VH amino acid sequence identical to, or identical except for one, two, three, four, five, or
more amino acid substitutions to SEQ ID NO: 15 or SEQ ID NO:20..
Also disclosed is an isolated binding molecule, e.g., an dy or antigen-
binding fragment thereof which specifically binds to human (x-synuclein, comprising an
immunoglobulin light chain variable region (VL) amino acid sequence at least 80%, 85%,
90% 95% or 100% identical to SEQ ID NO:22 or SEQ ID NO:26.
Some embodiments disclose an isolated binding molecule, e. g., an antibody or
antigen-binding fragment thereof which specifically binds to human (x-synuclein,
comprising a VL amino acid sequence identical to, or cal except for one, two, three,
four, five, or more amino acid tutions, to SEQ ID NO:22 or SEQ ID NO:26.
In other embodiments, an isolated antibody or n-binding fragment thereof
which specifically binds to human u-synuclein comprises, consists ially of, or
ts of VH and VL amino acid sequences at least 80%, 85%, 90% 95% or 100%
identical to: (a) SEQ ID NO:15 and SEQ ID NO:22, respectively,(b) SEQ ID NO:15 and
PCT/U82012/043701
SEQ ID NO:26, respectively,(c) SEQ ID N020 and SEQ ID NO:22 , respectively,(d)
SEQ ID N020 and SEQ ID NO:26, respectively. .
Also disclosed is an isolated binding molecule, e.g., an antibody or antigen-
g fragment thereof which specifically binds to human clein, comprising,
consisting essentially of, or consisting of an immunoglobulin heavy chain variable region
(VH), where at least one, two or all three VH-CDRs of the heavy chain variable region
are at least 80%, 85%, 90% or 95% identical to reference heavy chain VH-CDRl, VH-
CDR2 or VH-CDR3 amino acid sequences in Fig. l, and represented by SEQ ID N016,
SEQ ID NO:l7, and SEQ ID NO:18, respectively. Thus, according to this embodiment a
heavy chain variable region of the invention has VH-CDRl, VH-CDR2 and VH-CDR3
polypeptide sequences related to the VH-CDRl, VH-CDR2 and VH-CDR3 amino acid
ces represented by SEQ ID N016, SEQ ID NO:l7, and SEQ ID NO:18,
respectively. While Fig. 1 shows VH-CDRs defined by the Kabat system, other CDR
definitions, e.g., s defined by the Chothia system, are also included in the
present invention, and can be easily fied by a person of ordinary skill in the art
using the sequence data presented.
Also disclosed is an isolated binding molecule, e.g., an antibody or antigen-
binding fragment f which specifically binds to human clein, comprising,
consisting essentially of, or consisting of an immunoglobulin heavy chain variable region
(VH) in which the VH-CDRl, VH-CDR2 and VH-CDR3 s have ptide
sequences which are cal to the VH-CDRl, VH-CDR2 and VH-CDR3 amino acid
sequences represented by SEQ ID N016, SEQ ID NO:l7, and SEQ ID NO:18,
respectively.
Also disclosed is an ed binding molecule, e.g., an antibody or antigen-
binding fragment thereof which specifically binds to human u-synuclein, comprising,
ting essentially of, or consisting of an immunoglobulin heavy chain variable region
(VH) in which the VH-CDRl, VH-CDR2 and VH-CDR3 regions have polypeptide
sequences which are identical to, or identical except for one, two, three, four, five, or six
amino acid substitutions in any one VH-CDR, to the VH-CDRl, VH-CDR2 or VH-CDR3
amino acid sequences represented by SEQ ID NO:l6, SEQ ID NO:l7, and SEQ ID
NO:18, respectively. Also provided is an immunoglobulin heavy chain variable region
(VH) in which the VH-CDRl, VH-CDR2 and 3 regions have polypeptide
W0 2012/177972 PCT/U82012/043701
_ 35 _
sequences which are identical to, or identical except for five, six, seven, eight, nine, ten,
eleven, twelve, thirteen, fourteen, fifteen, or twenty total CDR substitutions to amino acid
sequences represented by SEQ ID NO:l6, SEQ ID NO:l7, and SEQ ID NO:18,
respectively. In certain embodiments the amino acid substitutions are conservative.
Also disclosed is an isolated binding molecule, e.g., an antibody or antigenbinding
fragment thereof which specifically binds to human u-synuclein, comprising,
consisting essentially of, or ting of an immunoglobulin light chain variable region
(VL), where at least one, two, or all three of the VL-CDRs of the light chain variable
region are at least 80%, 85%, 90% or 95% identical to nce light chain VL-CDRl,
VL-CDR2 or VL-CDR3 amino acid sequences ented by SEQ ID NO:23, SEQ ID
N024, and SEQ ID NO:25, tively. Thus, according to this embodiment a light
chain variable region of the invention has VL-CDRl, VL-CDR2 and VL-CDR3
polypeptide sequences d to the polypeptides shown in Fig. l and represented by
SEQ ID NO:23, SEQ ID N024, and SEQ ID NO:25, respectively. While Fig. 1 shows
VL-CDRs defined by the Kabat system, other CDR definitions, e. g., VL-CDRs defined
by the Chothia system, are also included in the present invention.
Also disclosed is an isolated binding molecule, e.g., an antibody or antigen-
binding fragment thereof which specifically binds to human u-synuclein, comprising,
consisting essentially of, or consisting of an immunoglobulin light chain variable region
(VL) in which the VL-CDRl, VL-CDR2 and VL-CDR3 regions have polypeptide
sequences which are cal to the VL-CDRl, VL-CDR2 and VL-CDR3 groups shown
in Fig. l and ented by SEQ ID NO:23, SEQ ID N024, and SEQ ID NO:25,
respectively.
Also sed is an isolated binding molecule, e.g., an antibody or antigen-
binding fragment thereof which specifically binds to human u-synuclein, comprising,
consisting essentially of, or consisting of an immunoglobulin light chain variable region
(VL) in which the VL-CDRl, VL-CDR2 and VL-CDR3 regions have polypeptide
sequences which are identical to, or cal except for one, two, three, four, five, or six
amino acid substitutions in any one VL-CDR, to the VL-CDRl, VL-CDR2 or VL-CDR3
amino acid sequences represented by SEQ ID NO:23, SEQ ID N024, and SEQ ID
NO:25, respectively. Also provided is an globulin light chain le region
(VL) in which the VL-CDRl, VL-CDR2 and VL-CDR3 regions have polypeptide
PCT/U82012/043701
sequences which are identical to, or identical except for five, six, seven, eight, nine, ten,
eleven, twelve, thirteen, fourteen or fifteen total CDR substitutions to amino acid
sequences represented by SEQ ID NO:23, SEQ ID N024, and SEQ ID NO:25,
tively. In certain embodiments the amino acid substitutions are conservative.
An immunoglobulin or its encoding cDNA can be further modified. Thus, in a
r embodiment the method of the present invention comprises any one of the step(s)
of producing a chimeric antibody, murinized antibody, single-chain antibody, Fab-
fragment, bi-specific antibody, fusion antibody, labeled antibody or an analog of any one
of those. Corresponding methods are known to the person skilled in the art and are
bed, e.g., in Harlow and Lane "Antibodies, A Laboratory Manual", CSH Press,
Cold Spring Harbor (1988). When tives of said antibodies are obtained by the
phage display technique, surface plasmon resonance as employed in the e system
can be used to se the efficiency of phage antibodies which bind to the same epitope
as that of any one of the antibodies described herein (Schier, Human Antibodies
Hybridomas 7 (1996), 97-105; Malmborg, J. Immunol. Methods 183 (1995), 7-13). The
production of chimeric antibodies is described, for e, in international application
WO89/09622. Methods for the production of humanized antibodies are described in, e.g.,
European application EP-Al 0 239 400 and international application WO90/0786l. A
further source of antibodies to be utilized in ance with the present invention are so-
called xenogeneic antibodies. The general principle for the tion of xenogeneic
antibodies such as human-like antibodies in mice is described in, e.g., international
applications WO91/10741, WO94/02602, WO96/34096 and WO 96/33735. As sed
above, an antibody of the invention can exist in a variety of forms besides complete
dies; including, for example, Fv, Fab and , as well as in single chains, such
as scFvs; see e.g. international application 9344.
The antibodies of the present invention or their corresponding immunoglobulin
chain(s) can be further modified using conventional ques known in the art, for
example, by using amino acid deletion(s), insertion(s), substitution(s), addition(s), and/or
recombination(s) and/or any other modification(s) known in the art either alone or in
combination. Methods for introducing such modifications in the DNA sequence
underlying the amino acid sequence of an immunoglobulin chain are well known to the
person skilled in the art; see, e.g., Sambrook, Molecular Cloning A tory Manual,
W0 2012/177972 PCT/U82012/043701
Cold Spring Harbor tory (1989) NY. and Ausubel, Current Protocols in
Molecular Biology, Green Publishing Associates and Wiley Interscience, NY. (1994).
Modifications of the antibody of the invention include chemical and/or enzymatic
derivatizations at one or more constituent amino acids, including side chain
modifications, backbone modifications, and N- and C-terminal modifications including
acetylation, hydroxylation, methylation, amidation, and the attachment of carbohydrate or
lipid moieties, cofactors, and the like. Likewise, the present invention encompasses the
tion of ic proteins which comprise the described antibody or some fragment
thereof at the amino terminus fused to heterologous molecule such as an
immunostimulatory ligand at the carboxyl terminus; see, e.g., international application
WOOD/30680 for corresponding technical details.
Additionally, the present invention encompasses peptides including those
containing a g molecule as described above, for example containing the CDR3
region of the variable region of any one of the mentioned antibodies, in particular CDR3
of the heavy chain since it has frequently been observed that heavy chain CDR3
(HCDR3) is the region having a greater degree of variability and a predominant
participation in antigen-antibody interaction. Such peptides can easily be synthesized or
ed by recombinant means to produce a binding agent useful ing to the
invention. Such methods are well known to those of ordinary skill in the art. Peptides can
be synthesized for example, using ted peptide synthesizers which are
commercially available. The peptides can also be produced by recombinant techniques by
incorporating the DNA expressing the e into an expression vector and transforming
cells with the expression vector to produce the peptide.
Hence, the t invention relates to any binding molecule, e.g., an antibody or
g fragment thereof which is oriented s the human anti-(x-synuclein
antibodies of the present invention and display the mentioned ties, z'.e. which
specifically recognize u-synuclein. Such dies and binding molecules can be tested
for their binding specificity and affinity by ELISA and Western Blot and
immunohistochemistry as described herein, see, e.g., the Examples. Furthermore,
preliminary results of uent experiments performed in accordance with the present
ion revealed that the human anti-(x-synuclein antibody of the present invention, in
particular antibody NI-202.22D11 recognizes clein inclusion bodies present on
PCT/U82012/043701
human brain sections of patients who suffered from dementia with Lewy bodies (DLB) or
son’s disease (PD). Thus, in one embodiment of the present invention, the human
antibody or binding fragment, tive or variant thereof izes (x-synuclein on
human DLB or PD brain sections (see, e.g., Fig. 4c).
Immortalized B cells or B memory cells can be used as a source of rearranged
heavy chain and light chain loci for subsequent expression and/or genetic manipulation.
Rearranged dy genes can be reverse transcribed from appropriate mRNAs to
produce cDNA. If desired, the heavy chain constant region can be exchanged for that of a
different isotype or eliminated altogether. Nucleotide sequences can be engineered to
remove undesired motifs (such as splice sites or restriction sties), and the codon usage can
be optimized for the cell in which the antibody or fragment thereof is to be expressed. In
addition, one or more mutations which alter amino acids in the variable regions can be
made, e.g., to increase affinity or improve stability. The variable regions can be linked to
encode single chain Fv s. Multiple Fv regions can be linked to confer g
ability to more than one target or chimeric heavy and light chain combinations can be
employed. Once the genetic material is available, design of analogs as described above
which retain both their ability to bind the desired target is straightforward. Methods for
the cloning of antibody variable regions and generation of recombinant antibodies are
known to the person d in the art and are described, for example, Gilliland et al.,
Tissue ns 47 (1996), 1-20; Doenecke et al., ia ll (1997), 1787-1792.
Once the appropriate genetic material is obtained and, if desired, modified to
encode an analog, the coding sequences, including those that encode, at a minimum, the
le regions of the heavy and light chain, can be inserted into expression systems
contained on vectors which can be transfected into standard recombinant host cells. A
variety of such host cells can be used; for efficient processing. Typical mammalian cell
lines useful for this purpose include, but are not limited to, CHO cells, HEK 293 cells, or
NSO cells.
The tion of the antibody or analog is then aken by culturing the
modified inant host under culture conditions appropriate for the growth of the host
cells and the expression of the coding sequences. The antibodies are then recovered by
isolating them from the culture. Expression systems can be designed to include signal
peptides so that the resulting antibodies are secreted into the medium; however,
intracellular production is also possible.
In accordance with the above, the present invention also relates to a
polynucleotide ng the antibody or equivalent binding molecule of the t
invention, one or more CDRs, one or more of a heavy chain or light chain variable
regions or variants thereof, of an immunoglobulin chain of the anti- u-synuclein
antibodies described above.
The person skilled in the art will y appreciate that the le domain of an
antibody, or any portion thereof can be used for the construction of other polypeptides or
antibodies of desired specificity and biological on. Thus, the present invention also
es polypeptides and antibodies comprising at least one heavy chain or light chain
CDR, or such CDR with 1, 2, 3, 4, or more amino acid substitutions, of antibody NI-
202.22D11, which can have substantially the same or similar binding properties as NI-
202.22D11, described in the appended examples. The person skilled in the art knows that
binding affinity can be enhanced by making amino acid substitutions within the CDRs or
within the hypervariable loops (Chothia and Lesk, J. Mol. Biol. 196 (1987), 901-917)
which partially p with the CDRs as defined by Kabat; see, e.g., Riechmann, et al,
Nature 332 (1988), 323-327. Thus, the present invention also relates to antibodies
wherein one or more of the mentioned CDRs comprise one or more amino acid
substitutions. In certain embodiments, an antibody of the invention comprises in one or
both of its immunoglobulin chains two or all three CDRs of the variable s (original
or ted) as set forth in Fig. 1.
Binding molecules, e.g., dies, or antigen-binding fragments, variants, or
derivatives thereof of the invention, as known by those of ordinary skill in the art, can
comprise a constant region which mediates one or more effector filnctions. For example,
binding of the C1 ent of complement to an antibody constant region can activate
the complement system. Activation of complement is important in the opsonization and
lysis of cell ens. The activation of complement also stimulates the inflammatory
response and can also be involved in autoimmune ensitivity. Further, antibodies
bind to receptors on various cells via the Fc region, with a PC receptor binding site on the
antibody Fc region binding to a PC receptor (FcR) on a cell. There are a number of Fc
receptors which are specific for different classes of antibody, including IgG (gamma
receptors), IgE (epsilon receptors), IgA (alpha receptors) and IgM (mu receptors).
Binding of antibody to Fc receptors on cell surfaces triggers a number of important and
diverse biological responses including engulfrnent and destruction of dy-coated
particles, clearance of immune xes, lysis of dy-coated target cells by killer
cells (called antibody-dependent cell-mediated cytotoxicity, or ADCC), release of
inflammatory mediators, placental transfer and control of immunoglobulin production.
Accordingly, certain embodiments of the present invention include an antibody, or
antigen-binding nt, variant, or derivative thereof, in which at least a fraction of one
or more of the constant region domains has been deleted or ise altered so as to
provide desired biochemical characteristics such as reduced effector functions, the ability
to non-covalently dimerize, increased ability to localize at the site of u-synuclein
aggregation and deposition, d serum half-life, or increased serum half-life when
compared with a whole, unaltered antibody of approximately the same immunogenicity.
For example, certain antibodies for use in the diagnostic and treatment methods described
herein are domain deleted antibodies which comprise a polypeptide chain similar to an
immunoglobulin heavy chain, but which lack at least a portion of one or more heavy
chain domains. For instance, in certain antibodies, one entire domain of the constant
region of the ed antibody will be deleted, for e, all or part of the CH2
domain will be deleted. In other embodiments, certain antibodies for use in the diagnostic
and treatment methods described herein have a constant region, e.g., an IgG heavy chain
constant region, which is d to eliminate glycosylation, referred to elsewhere herein
as aglycosylated or "agly" dies. Such 'agly" antibodies can be ed
enzymatically as well as by engineering the consensus glycosylation site(s) in the
constant region. While not being bound by theory, it is believed that "agly" antibodies
may have an improved safety and stability profile in viva. Methods of producing
sylated antibodies, having desired effector function are found for example in
ational application W02005/018572, which is incorporated by reference in its
entirety.
In n antibodies, or antigen-binding fragments, variants, or derivatives thereof
described herein, the Fc portion can be mutated to decrease effector function using
ques known in the art. For example, the on or inactivation (through point
mutations or other means) of a constant region domain can reduce Fc receptor binding of
the circulating modified antibody thereby increasing u-synuclein localization. In other
cases, nt region modifications consistent with the instant invention moderate
complement binding and thus reduce the serum half life and nonspecific association of a
conjugated cytotoxin. Yet other ations of the constant region can be used to
modify disulfide linkages or oligosaccharide moieties that allow for enhanced localization
due to increased antigen city or antibody flexibility. The resulting physiological
profile, bioavailability and other biochemical effects of the modifications, such as 0L-
synuclein localization, biodistribution and serum ife, can easily be measured and
quantified using well know immunological techniques without undue mentation.
In n antibodies, or antigen-binding fragments, variants, or derivatives thereof
described herein, the Fc portion can be mutated or exchanged for alternative protein
sequences to increase the cellular uptake of antibodies by way of example by enhancing
receptor-mediated endocytosis of antibodies via Fcy receptors, LRP, or Thyl receptors or
by 'SuperAntibody Technology', which is said to enable antibodies to be shuttled into
living cells without harming them (Expert Opin. Biol. Ther. (2005), 237-241). For
example, the generation of filSlOI‘l proteins of the antibody g region and the cognate
protein ligands of cell surface receptors or bi- or multi-specific dies with a specific
sequences biding to u-synuclein as well as a cell e receptor can be engineered using
techniques known in the art.
In certain antibodies, or antigen-binding fragments, variants, or derivatives thereof
bed herein, the Fc portion can be d or exchanged for alternative protein
ces or the antibody can be chemically modified to increase its blood brain r
penetration.
Modified forms of antibodies, or antigen-binding fragments, variants, or
derivatives thereof of the invention can be made from whole precursor or parent
antibodies using techniques known in the art. Exemplary techniques are sed in
more detail herein. Antibodies, or antigen-binding fragments, variants, or derivatives
f of the invention can be made or ctured using techniques that are known in
the art. In certain embodiments, antibody molecules or fragments thereof are
binantly produced," z'.e., are produced using recombinant DNA technology.
Exemplary techniques for making antibody molecules or fragments thereof are discussed
in more detail elsewhere herein.
Antibodies, or antigen-binding fragments, variants, or derivatives thereof of the
invention also e tives that are modified, e.g., by the covalent attachment of
any type of molecule to the antibody such that covalent attachment does not prevent the
antibody from specifically g to its cognate e. For example, but not by way of
limitation, the antibody derivatives include antibodies that have been modified, e. g., by
glycosylation, acetylation, pegylation, orylation, amidation, derivatization by
known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or
other protein, etc. Any of numerous chemical ations can be carried out by known
techniques, including, but not d to specific chemical cleavage, acetylation,
formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative can
contain one or more non-classical amino acids.
In certain embodiments, antibodies, or antigen-binding fragments, variants, or
derivatives thereof of the invention will not elicit a deleterious immune se in the
animal to be treated, e. g., in a human. In certain embodiments, binding molecules, e.g.,
antibodies, or antigen-binding fragments thereof of the invention are derived from a
patient, e. g., a human patient, and are subsequently used in the same species from which
they are derived, e.g., human, alleviating or minimizing the occurrence of deleterious
immune responses.
De-immunization can also be used to decrease the immunogenicity of an antibody.
As used herein, the term munization" includes alteration of an antibody to modify
T cell es; see, e.g., international ations WO98/52976 and WOOD/34317. For
example, VH and VL sequences from the starting antibody are analyzed and a human T
cell epitope "map" from each V region showing the location of epitopes in relation to
complementarity determining regions (CDRs) and other key residues within the sequence.
Individual T cell epitopes from the T cell epitope map are analyzed in order to identify
alternative amino acid substitutions with a low risk of altering activity of the final
antibody. A range of ative VH and VL sequences are designed comprising
combinations of amino acid substitutions and these sequences are subsequently
incorporated into a range of g polypeptides, e.g., u-synuclein-specific dies or
immunospecific fragments thereof for use in the diagnostic and ent methods
disclosed herein, which are then tested for fianction. Typically, between 12 and 24 variant
antibodies are generated and tested. Complete heavy and light chain genes comprising
PCT/U82012/043701
_ 43 _
modified V and human C regions are then cloned into sion vectors and the
subsequent plasmids introduced into cell lines for the production of whole antibody. The
antibodies are then compared in appropriate biochemical and biological assays, and the
optimal variant is identified.
Monoclonal antibodies can be ed using a wide variety of techniques known
in the art including the use of hybridoma, recombinant, and phage display technologies,
or a combination thereof. For example, monoclonal antibodies can be produced using
hybridoma techniques including those known in the art and taught, for e, in
Harlow et al., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press,
2nd ed. (1988); ling et al., in: Monoclonal Antibodies and T-Cell Hybridomas
Elsevier, N.Y., 563-681 (1981), said references incorporated by reference in their
entireties. The term lonal antibody" as used herein is not limited to antibodies
produced through hybridoma technology. The term "monoclonal antibody" refers to an
antibody that is derived from a single clone, ing any eukaryotic, prokaryotic, or
phage clone, and not the method by which it is produced. Thus, the term "monoclonal
antibody" is not limited to antibodies produced through hybridoma logy. In certain
embodiments, antibodies of the present invention are derived from human B cells which
have been immortalized via transformation with Epstein-Barr virus, as described .
In the well known hybridoma process (Kohler et al., Nature 256 (1975), 495) the
relatively short-lived, or mortal, lymphocytes from a , e.g., B cells d from
a human subject as bed herein, are fused with an immortal tumor cell line (e. g.,. a
myeloma cell line), thus, producing hybrid cells or "hybridomas" which are both
immortal and e of producing the genetically coded antibody of the B cell. The
resulting hybrids are segregated into single genetic strains by selection, dilution, and re-
growth with each individual strain comprising specific genes for the ion of a single
antibody. They produce antibodies, which are homogeneous against a desired antigen
and, in reference to their pure genetic parentage, are termed "monoclonal".
Hybridoma cells thus prepared are seeded and grown in a suitable culture medium
that can contain one or more substances that inhibit the growth or survival of the unfilsed,
parental myeloma cells. Those skilled in the art will appreciate that reagents, cell lines
and media for the formation, selection and growth of hybridomas are commercially
available from a number of sources and standardized protocols are well established.
PCT/U82012/043701
_ 44 _
Generally, culture medium in which the oma cells are growing is assayed for
production of onal antibodies against the desired antigen. The binding specificity
of the monoclonal antibodies produced by hybridoma cells is determined by in vitro
assays such as immunoprecipitation, radioimmunoassay (RIA) or enzyme-linked
immunoabsorbent assay (ELISA) as described herein. After hybridoma cells are
identified that produce antibodies of the desired specificity, affinity and/or activity, the
clones can be subcloned by limiting dilution ures and grown by standard methods;
see, e.g., Goding, Monoclonal Antibodies: Principles and Practice, Academic Press, pp
59-103 (1986). It will fiarther be appreciated that the onal antibodies secreted by
the subclones can be separated from culture medium, ascites fluid or serum by
conventional purification ures such as, for example, protein-A, hydroxylapatite
chromatography, gel electrophoresis, dialysis or affinity tography.
In another embodiment, lymphocytes can be ed by micromanipulation and
the variable genes isolated. For example, peripheral blood clear cells can be
isolated from an immunized or naturally immune mammal, e.g, a human, and cultured
for about 7 days in vitro. The cultures can be screened for specific IgGs that meet the
ing criteria. Cells from positive wells can be isolated. Individual Ig-producing B
cells can be isolated by FACS or by identifying them in a complement-mediated
hemolytic plaque assay. Ig-producing B cells can be micromanipulated into a tube and the
VH and VL genes can be amplified using, e. g., . The VH and VL genes can be
cloned into an antibody expression vector and transfected into cells (e. g., eukaryotic or
prokaryotic cells) for expression.
Alternatively, antibody-producing cell lines can be selected and cultured using
techniques well known to the skilled artisan. Such techniques are described in a y of
laboratory manuals and primary publications. In this respect, techniques suitable for use
in the invention as described below are described in Current Protocols in Immunology,
Coligan et al., Eds., Green Publishing Associates and Wiley-Interscience, John Wiley and
Sons, New York (1991) which is herein incorporated by reference in its entirety,
including ments.
Antibody fragments that recognize specific es can be generated by known
techniques. For example, Fab and F(ab')2 fragments can be produced recombinantly or by
proteolytic cleavage of immunoglobulin les, using enzymes such as papain (to
W0 2012/177972 PCT/U82012/043701
_ 45 _
produce Fab fragments) or pepsin (to produce F(ab')2 fragments). F(ab')2 fragments
contain the variable region, the light chain constant region and the CH1 domain of the
heavy chain. Such nts are sufficient for use, for example, in immunodiagnostic
procedures involving coupling the immunospecific portions of globulins to
detecting reagents such as radioisotopes.
Completely human antibodies, such as described herein, are ularly desirable
for therapeutic treatment of human patients. Human antibodies of the t invention
are isolated, e.g, from elderly ts who because of their age may be suspected to be
at risk of developing a disorder, e.g., Parkinson’s disease, or a patient with the disorder
but with an unusually stable e course. However, though it is prudent to expect that
elderly healthy and symptom-free subjects, respectively, more regularly will have
developed protective anti-u-synuclein antibodies than younger subjects, the latter can be
used as well as source for obtaining a human antibody of the present ion. This is
particularly true for younger patients who are predisposed to develop a familial form of a
einopathic disease but remain symptom-free since their immune system and
response functions more efficiently than that in older adults.
In one embodiment, an antibody of the invention comprises at least one heavy or
light chain CDR of an antibody molecule. In another embodiment, an antibody of the
invention comprises at least two CDRs from one or more antibody molecules. In r
embodiment, an antibody of the invention comprises at least three CDRs from one or
more antibody molecules. In r embodiment, an antibody of the invention comprises
at least four CDRs from one or more antibody molecules. In r embodiment, an
antibody of the invention comprises at least five CDRs from one or more antibody
molecules. In another embodiment, an antibody of the invention ses at least six
CDRs from one or more antibody molecules. ary antibody molecules comprising
at least one CDR that can be ed in the subject dies are described herein.
Antibodies of the present invention can be produced by any method known in the
art for the synthesis of antibodies, in particular, by chemical synthesis or by recombinant
expression techniques as described herein.
In one embodiment, an antibody, or antigen-binding fragment, variant, or
derivative thereof of the invention comprises a synthetic constant region wherein one or
more domains are partially or entirely d ("domain-deleted antibodies"). In certain
PCT/U82012/043701
_ 46 _
ments compatible modified antibodies will comprise domain deleted constructs or
variants n the entire CH2 domain has been removed (ACHZ constructs). For other
embodiments a short connecting peptide can be substituted for the deleted domain to
provide flexibility and freedom of movement for the variable . Domain deleted
constructs can be derived using a vector ng an IgG1 human constant domain, see,
e.g., international applications W002/060955 and W002/096948A2. This vector is
engineered to delete the CH2 domain and provide a synthetic vector expressing a domain
deleted IgG1 constant region.
In certain embodiments, antibodies, or antigen-binding fragments, variants, or
derivatives thereof of the present invention are minibodies. Minibodies can be made using
methods described in the art, see, e.g., US patent 5,837,821 or ational application
WO 94/09817.
In one embodiment, an antibody, or n-binding fragment, t, or
derivative thereof of the invention comprises an immunoglobulin heavy chain having
deletion or substitution of a few or even a single amino acid as long as it s
association between the monomeric subunits. For example, the on of a single amino
acid in selected areas of the CH2 domain can be enough to ntially reduce Fc
binding and thereby increase u-synuclein localization. Similarly, one or more constant
region domains that control the effector function (e. g. complement binding) can be
deleted. Such partial ons of the constant regions can improve selected characteristics
of the antibody (serum half-life) while leaving other desirable functions associated with
the subject nt region domain intact. Moreover, as alluded to above, the constant
s of the disclosed antibodies can be synthetic through the mutation or substitution
of one or more amino acids that enhances the profile of the resulting construct. In this
respect it can be possible to disrupt the activity provided by a conserved binding site (e.g.
Fc binding) while substantially maintaining the configuration and immunogenic profile of
the modified antibody. Yet other embodiments comprise the addition of one or more
amino acids to the constant region to enhance ble characteristics such as effector
function or provide for more cytotoxin or carbohydrate attachment. In such embodiments
it can be desirable to insert or replicate specific sequences derived from selected constant
region domains.
The present invention also provides antibodies that comprise, consist essentially
of, or consist of, variants (including derivatives) of antibody molecules (e. g., the VH
regions and/or VL regions) described herein, which antibodies or fragments f
immunospecifically bind to u-synuclein. Standard techniques known to those of skill in
the art can be used to introduce ons in the nucleotide sequence encoding an
antibody, including, but not limited to, site-directed mutagenesis and diated
mutagenesis which result in amino acid substitutions. Variants (including derivatives) can
encode less than 50 amino acid tutions, less than 40 amino acid substitutions, less
than 30 amino acid tutions, less than 25 amino acid substitutions, less than 20 amino
acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid
substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions,
less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the
reference VH , VH-CDRl, VH-CDRZ, VH-CDR3, VL region, VL-CDRl, VL-
CDR2, or VL-CDR3. A "conservative amino acid substitution" is one in which the amino
acid e is replaced with an amino acid residue having a side chain with a similar
charge. Families of amino acid es having side chains with similar charges have
been defined in the art. These families include amino acids with basic side chains (e.g.,
, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid),
uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine,
tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, e,
phenylalanine, methionine, phan), beta-branched side chains ( e.g., threonine,
valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan,
histidine). Alternatively, mutations can be introduced randomly along all or part of the
coding sequence, such as by saturation mutagenesis, and the resultant s can be
screened for biological activity to identify mutants that retain activity (e.g., the ability to
bind u-synuclein).
For example, it is possible to introduce mutations only in framework regions or
only in CDR regions of an antibody molecule. Introduced mutations can be silent or
neutral missense mutations, e.g., have no, or little, effect on an antibody’s y to bind
antigen, indeed some such ons do not alter the amino acid sequence ever.
These types of mutations can be useful to optimize codon usage, or improve a
hybridoma’s antibody production. Codon-optimized coding regions encoding antibodies
WO 77972 PCT/U82012/043701
_ 48 _
of the t invention are disclosed elsewhere herein. Alternatively, non-neutral
missense mutations can alter an antibody’s ability to bind antigen. The location of most
silent and neutral missense mutations is likely to be in the framework regions, while the
location of most non-neutral missense mutations is likely to be in CDR, though this is not
an absolute requirement. One of skill in the art would be able to design and test mutant
molecules with desired properties such as no alteration in n-binding activity or
alteration in binding activity (e.g., improvements in antigen-binding activity or change in
antibody specificity). Following mutagenesis, the encoded protein can routinely be
expressed and the fianctional and/or biological activity of the d n, (e. g.,
ability to immunospecifically bind at least one epitope of u-synuclein) can be determined
using ques described herein or by routinely modifying techniques known in the art.
111. Polynucleotides Encoding Antibodies
A polynucleotide encoding an antibody, or antigen-binding fragment, variant, or
derivative thereof can be composed of any polyribonucleotide or polydeoxribonucleotide,
which can be unmodified RNA or DNA or modified RNA or DNA. For example, a
polynucleotide encoding an antibody, or n-binding fragment, t, or derivative
thereof can be composed of single- and double-stranded DNA, DNA that is a mixture of
- and -stranded regions, single- and double-stranded RNA, and RNA that is
mixture of single- and double-stranded s, hybrid molecules comprising DNA and
RNA that can be single-stranded or, more typically, double-stranded or a mixture of
single- and double-stranded regions. In addition, a polynucleotide encoding an antibody,
or antigen-binding fragment, variant, or derivative thereof can be ed of triple-
stranded regions comprising RNA or DNA or both RNA and DNA. A polynucleotide
encoding an antibody, or antigen-binding fragment, variant, or derivative thereof can also
contain one or more modified bases or DNA or RNA backbones modified for ity or
for other reasons. "Modified" bases include, for example, tritylated bases and unusual
bases such as inosine. A variety of modifications can be made to DNA and RNA; thus,
"polynucleotide" es chemically, enzymatically, or metabolically modified forms.
An isolated polynucleotide encoding a non-natural variant of a polypeptide
derived from an immunoglobulin (e.g., an immunoglobulin heavy chain portion or light
chain portion) can be d by introducing one or more nucleotide substitutions,
additions or ons into the nucleotide sequence of the immunoglobulin such that one
PCT/U82012/043701
_ 49 _
or more amino acid substitutions, additions or deletions are uced into the encoded
protein. Mutations can be introduced by standard techniques, such as site-directed
mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions can
be made at one or more non-essential amino acid residues.
As is well known, RNA can be isolated from the al B cells, hybridoma cells
or from other transformed cells by standard techniques, such as guanidinium
isothiocyanate extraction and precipitation followed by centrifilgation or chromatography.
Where desirable, mRNA can be isolated from total RNA by rd techniques such as
chromatography on oligo dT cellulose. Suitable techniques are familiar in the art. In one
embodiment, cDNAs that encode the light and the heavy chains of the antibody can be
made, either simultaneously or separately, using reverse riptase and DNA
polymerase in accordance with well known methods. PCR can be initiated by consensus
constant region primers or by more specific primers based on the published heavy and
light chain DNA and amino acid sequences. As discussed above, PCR also can be used to
e DNA clones encoding the antibody light and heavy chains. In this case the
libraries can be screened by consensus primers or larger homologous probes, such as
human constant region .
DNA, typically d DNA, can be ed from the cells using techniques
known in the art, restriction mapped and sequenced in accordance with standard, well
known techniques set forth in detail, e.g., in the foregoing references relating to
recombinant DNA techniques. Of course, the DNA can be synthetic according to the
present invention at any point during the isolation process or subsequent analysis.
One embodiment provides an isolated polynucleotide sing, ting
ially of, or consisting of a nucleic acid encoding an immunoglobulin heavy chain
variable region (VH) amino acid sequence at least 80%, 85%, 90% 95% or 100%
identical to SEQ ID NO:lS or SEQ ID NO:20.
Another embodiment es an isolated polynucleotide comprising, consisting
essentially of, or consisting of a nucleic acid encoding a VH amino acid sequence
identical to, or identical except for one, two, three, four, five, or more amino acid
substitutions to SEQ ID NO:15 or SEQ ID NO:20.
Another ment provides an isolated polynucleotide comprising, consisting
essentially of, or consisting of a nucleic acid encoding an immunoglobulin heavy chain
W0 2012/177972 2012/043701
variable region (VH), where at least one, two or all three of the CDRs of the heavy chain
variable region are at least 80%, 85%, 90% or 95% identical to reference heavy chain
VH-CDRl, VH-CDR2 or VH-CDR3 amino acid sequences represented by SEQ ID
N016, SEQ ID NO:17, and SEQ ID NO:18, respectively. Thus, this embodiment
provides an ed polynucleotide encoding a heavy chain variable region of the
invention which has VH-CDRl, VH-CDR2 and VH-CDR3 amino acid sequences related
to those represented by SEQ ID N016, SEQ ID NO:17, and SEQ ID NO:18, respectively,
as shown in Fig. 1.
In another ment, the present invention provides an isolated polynucleotide
comprising, consisting ially of, or consisting of a nucleic acid encoding an
immunoglobulin heavy chain variable region (VH) in which the VH-CDRl, VH-CDRZ,
and VH-CDR3 regions have polypeptide sequences which are identical to the VH-CDRl,
VH-CDRZ, and VH-CDR3 groups represented by SEQ ID N016, SEQ ID NO:17, and
SEQ ID NO:18, respectively, as shown in Fig. 1.
A fiarther embodiment provides an ed binding molecule e. g., an dy or
antigen-binding nt comprising the VH d by the polynucleotide which
specifically or preferentially binds to human (x-synuclein.
Another embodiment provides an isolated polynucleotide comprising, consisting
essentially of, or consisting of a nucleic acid encoding an immunoglobulin light chain
variable region (VL) amino acid sequence at least 80%, 85%, 90% 95% or 100% identical
toSEQ ID NO:22 or SEQ ID NO:26.
A fiarther embodiment provides an isolated polynucleotide comprising, consisting
essentially of, or consisting of a nucleic acid ng a VL amino acid sequence
identical to, or identical except for one, two, three, four, five, or more amino acid
substitutions to SEQ ID NO:22 or SEQ ID NO:26.
Another embodiment provides an isolated polynucleotide comprising, consisting
essentially of, or consisting of a c acid encoding an immunoglobulin light chain
variable region (VL), where at least one, two, or all three of the VL-CDRs of the light
chain variable region are at least 80%, 85%, 90% or 95% identical to reference light chain
VL-CDRl, 2 or VL-CDR3 amino acid sequences represented by SEQ ID N023,
SEQ ID N024, and SEQ ID NO:25, respectively. Thus, this embodiment provides an
isolated polynucleotide ng a light chain variable region of the invention which has
VL-CDRl, VL-CDR2 and VL-CDR3 amino acid sequences d to those represented
by SEQ ID N023, SEQ ID N024, and SEQ ID NO:25, respectively, as shown in Fig. 1.
In another embodiment, the present invention provides an isolated polynucleotide
comprising, consisting essentially of, or consisting of a nucleic acid encoding an
immunoglobulin light chain variable region (VL) in which the VL-CDRl , VL-CDRZ, and
VL-CDR3 regions have polypeptide sequences which are identical to the VH-CDRl, VH-
CDR2, and 3 groups represented by SEQ ID N016, SEQ ID NO:17, and SEQ
ID NO:18, respectively, as shown in Fig. 1.
A fiarther ment provides an isolated binding molecule e. g., an antibody or
antigen-binding fragment comprising the VL encoded by the polynucleotide which
specifically or preferentially binds to human (x-synuclein.
As known in the art, "sequence identity" between two polypeptides or two
polynucleotides is determined by comparing the amino acid or nucleic acid sequence of
one ptide or polynucleotide to the sequence of a second ptide or
cleotide. When discussed herein, whether any particular polypeptide is at least
about 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95% identical to
another polypeptide can be determined using methods and computer programs/software
known in the art such as, but not limited to, the BESTFIT program (Wisconsin Sequence
is Package, Version 8 for Unix, Genetics Computer Group, sity Research
Park, 575 Science Drive, Madison, WI 53711). BESTFIT uses the local homology
algorithm of Smith and Waterman, Advances in d Mathematics 2 (1981), 482-489,
to find the best segment of homology between two sequences. When using BESTFIT or
any other sequence alignment program to determine whether a particular sequence is, for
example, 95% identical to a reference sequence according to the present ion, the
parameters are set, of course, such that the percentage of identity is calculated over the
full length of the reference polypeptide sequence and that gaps in homology of up to 5%
of the total number of amino acids in the reference sequence are d.
In one embodiment of the present invention, the cleotide comprises,
consists essentially of, or consists of a nucleic acid having a polynucleotide sequence of
the VH set forth in SEQ ID NO:19 or SEQ ID NO:21, or the VL set forth in SEQ ID
NO:27 or SEQ ID NO:28. In this respect, the person skilled in the art will readily
appreciate that the cleotides encoding at least the variable domain of the light
W0 2012/177972 PCT/U82012/043701
and/or heavy chain can encode the le domain of both immunoglobulin chains or
only one.
The present ion also includes fragments of the polynucleotides of the
ion, as described elsewhere. Additionally polynucleotides which encode fusion
polynucleotides, Fab fragments, and other tives, as described herein, are also
plated by the invention.
The polynucleotides can be produced or manufactured by any method known in
the art. For example, if the nucleotide ce of the antibody is known, a
polynucleotide encoding the dy can be assembled from chemically synthesized
oligonucleotides, e.g., as described in Kutmeier et al., BioTechniques 17 (1994), 242,
which, briefly, involves the synthesis of overlapping oligonucleotides containing portions
of the sequence encoding the antibody, annealing and ligating of those oligonucleotides,
and then amplification of the ligated oligonucleotides by PCR.
Alternatively, a polynucleotide ng an antibody, or antigen-binding
fragment, variant, or derivative thereof can be generated from nucleic acid from a le
. If a clone containing a nucleic acid encoding a particular antibody is not ble,
but the sequence of the antibody le is known, a nucleic acid encoding the antibody
can be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA
library, or a cDNA library generated from, or nucleic acid, such as polyA+ RNA, isolated
from, any tissue or cells expressing the a-synuclein-specific antibody, such as hybridoma
cells selected to express an antibody) by PCR amplification using synthetic primers
hybridizable to the 3' and 5' ends of the sequence or by cloning using an oligonucleotide
probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a
cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR can
then be cloned into replicable cloning vectors using any method well known in the art.
Once the nucleotide sequence and ponding amino acid sequence of the
dy, or antigen-binding fragment, variant, or derivative thereof is determined, its
nucleotide sequence can be manipulated using methods well known in the art for the
manipulation of tide sequences, e. g., recombinant DNA techniques, site directed
mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al.,
Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold
Spring Harbor, NY. (1990) and Ausubel et al., eds., Current Protocols in Molecular
W0 2012/177972 PCT/U82012/043701
_ 53 _
y, John Wiley & Sons, NY (1998), which are both incorporated by reference herein
in their entireties), to generate antibodies having a different amino acid ce, for
example to create amino acid substitutions, deletions, and/or insertions.
IV. Expression of Antibody Polypeptides
ing manipulation of the isolated genetic material to provide antibodies, or
antigen-binding fragments, variants, or derivatives thereof of the invention, the
polynucleotides encoding the antibodies are typically inserted in an expression vector for
introduction into host cells that can be used to produce the desired ty of antibody.
Recombinant expression of an antibody, or fragment, derivative or analog thereof, e.g., a
heavy or light chain of an dy which binds to a target molecule is described herein.
Once a polynucleotide ng an antibody molecule or a heavy or light chain of an
antibody, or n thereof (e.g., containing the heavy or light chain variable domain), of
the invention has been obtained, the vector for the tion of the antibody molecule
can be ed by recombinant DNA technology using techniques well known in the art.
Thus, methods for ing a protein by expressing a polynucleotide containing an
antibody encoding nucleotide ce are described herein. Methods which are well
known to those skilled in the art can be used to construct expression vectors containing
dy coding sequences and appropriate transcriptional and ational control
signals. These methods include, for example, in vitro recombinant DNA techniques,
synthetic techniques, and in viva genetic recombination. The invention, thus, provides
replicable vectors comprising a nucleotide sequence encoding an antibody molecule of
the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain,
operably linked to a promoter. Such vectors can include the nucleotide sequence encoding
the constant region of the antibody molecule (see, e.g., international applications WO
07 and WO 89/01036; and US patent no. 5,122,464) and the variable domain of
the dy can be cloned into such a vector for expression of the entire heavy or light
chain.
The term "vector" or "expression vector" is used herein to mean vectors used in
accordance with the present invention as a vehicle for introducing into and expressing a
desired gene in a host cell. As known to those skilled in the art, such vectors can easily be
selected from the group consisting of plasmids, phages, Viruses and iruses. In
general, vectors compatible with the instant invention will comprise a selection marker,
W0 2012/177972 PCT/U82012/043701
appropriate restriction sites to facilitate cloning of the desired gene and the ability to enter
and/or replicate in eukaryotic or prokaryotic cells. For the purposes of this invention,
numerous expression vector systems can be employed. For example, one class of vector
utilizes DNA elements which are derived from animal viruses such as bovine papilloma
virus, polyoma virus, adenovirus, ia virus, baculovirus, iruses (RSV, MMTV
or MOMLV) or SV40 virus. Others involve the use of polycistronic systems with internal
ribosome binding sites. Additionally, cells which have integrated the DNA into their
chromosomes can be selected by introducing one or more markers which allow selection
of transfected host cells. The marker can provide for prototrophy to an ophic host,
biocide resistance (e. g., antibiotics) or resistance to heavy metals such as copper. The
selectable marker gene can either be directly linked to the DNA sequences to be
expressed, or introduced into the same cell by nsformation. Additional ts can
also be added for optimal sis of mRNA. These elements can e signal
sequences, splice signals, as well as transcriptional promoters, enhancers, and termination
signals.
In certain embodiments the cloned variable region genes are inserted into an
expression vector along with the heavy and light chain constant region genes (e.g.,
human) as discussed above. In one embodiment, this is effected using a proprietary
expression vector of Biogen IDEC, Inc., ed to as NEOSPLA, disclosed in US patent
no. 6,159,730. This vector contains the cytomegalovirus promoter/enhancer, the mouse
beta globin major promoter, the SV40 origin of replication, the bovine grth hormone
polyadenylation sequence, neomycin phosphotransferase exon 1 and exon 2, the
dihydrofolate reductase gene and leader sequence. This vector has been found to result in
very high level expression of antibodies upon incorporation of variable and constant
region genes, transfection in CHO cells, followed by selection in G418 containing
medium and methotrexate amplification. Of course, any expression vector which is
capable of eliciting expression in eukaryotic cells can be used in the present invention.
Examples of suitable vectors include, but are not limited to plasmids pcDNA3,
Zeo, pCR3.l, pEFl/His, pIND/GS, MVZ, pSV4O/Ze02, pTRACER-
HCMV, 5-His, pVAXl, and pZeoSV2 (available from Invitrogen, San Diego,
CA), and plasmid pCI (available from Promega, Madison, WI). In general, screening
large numbers of ormed cells for those which s suitably high levels if
_ 55 _
globulin heavy and light chains is routine experimentation which can be carried
out, for example, by robotic systems. Vector systems are also taught in US patent nos.
,736,137 and 5,658,570, each of which is incorporated by reference in its entirety herein.
This system provides for high expression levels, e. g., > 30 pg/cell/day. Other exemplary
vector systems are disclosed e.g., in US patent no. 6,413,777.
In other embodiments the antibodies, or antigen-binding fragments, variants, or
derivatives thereof of the ion can be expressed using polycistronic constructs such
as those disclosed in US patent application publication no. 2003-0157641 A1 and
incorporated herein in its entirety. In these expression s, multiple gene products of
interest such as heavy and light chains of antibodies can be produced from a single
polycistronic construct. These systems advantageously use an internal ribosome entry site
(IRES) to provide relatively high levels of antibodies. Compatible IRES sequences are
disclosed in US patent no. 6,193,980 which is also orated herein. Those skilled in
the art will iate that such expression systems can be used to effectively produce the
full range of antibodies disclosed in the instant ation.
More generally, once the vector or DNA sequence encoding a monomeric subunit
of the antibody has been prepared, the expression vector can be introduced into an
appropriate host cell. uction of the plasmid into the host cell can be accomplished
by various techniques well known to those of skill in the art. These include, but are not
limited to, transfection including lipotransfection using, e.g., Fugene or lipofectamine,
protoplast fusion, calcium phosphate precipitation, cell fusion with enveloped DNA,
microinjection, and infection with intact Virus. Typically, d introduction into the
host is Via standard calcium phosphate co-precipitation method. The host cells harboring
the expression construct are grown under conditions appropriate to the production of the
light chains and heavy chains, and d for heavy and/or light chain n synthesis.
Exemplary assay techniques e enzyme-linked immunosorbent assay ),
radioimmunoassay (RIA), or fluorescence-activated cell sorter analysis (FACS),
immunohistochemistry and the like.
The expression vector is transferred to a host cell by conventional techniques and
the transfected cells are then cultured by conventional techniques to produce an antibody
for use in the s described . Thus, the invention includes host cells containing
a polynucleotide encoding an antibody of the ion, or a heavy or light chain thereof,
W0 2012/177972 2012/043701
operably linked to a heterologous promoter. For the expression of double-chained
antibodies, s encoding both the heavy and light chains can be inserted into a host
cell for expression of the entire immunoglobulin molecule, as detailed below.
The host cell can be nsfected with two expression vectors of the invention,
the first vector encoding a heavy chain derived ptide and the second vector
encoding a light chain d polypeptide. The two vectors can contain identical
selectable markers which enable equal expression of heavy and light chain ptides.
Alternatively, a single vector can be used which encodes both heavy and light chain
polypeptides. In such situations, the light chain is advantageously placed before the heavy
chain to avoid an excess of toxic free heavy chain; see Proudfoot, Nature 322 (1986), 52;
Kohler, Proc. Natl. Acad. Sci. USA 77 (1980), 2197. The coding sequences for the heavy
and light chains can comprise cDNA or c DNA.
As used herein, "host cells" refers to cells which harbor vectors constructed using
recombinant DNA techniques and encoding at least one heterologous gene. In
descriptions of processes for isolation of antibodies from recombinant hosts, the terms
"cell" and "cell culture" are used interchangeably to denote the source of antibody unless
it is clearly specified otherwise. In other words, recovery of polypeptide from the "cells"
can mean either from spun down whole cells, or from the cell culture containing both the
medium and the suspended cells.
A variety of xpression vector systems can be utilized to express antibody
molecules for use in the methods described . Such host-expression systems
represent vehicles by which the coding sequences of interest can be produced and
subsequently purified, but also represent cells which can, when transformed or transfected
with the riate nucleotide coding sequences, express an antibody molecule of the
invention in situ. These include but are not limited to microorganisms such as bacteria
(e.g., E. 0011', B. subtilis) transformed with recombinant bacteriophage DNA, plasmid
DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast
(e.g., Saccharomyces, ) transformed with recombinant yeast expression vectors
containing antibody coding sequences; insect cell systems ed with recombinant
virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant
cell systems ed with recombinant virus expression vectors (e.g., cauliflower mosaic
virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid
expression s (e.g., Ti plasmid) containing antibody coding sequences; or
mammalian cell systems (e. g., COS, CHO, NSO, BLK, 293, 3T3 cells) harboring
recombinant expression constructs containing ers derived from the genome of
mammalian cells (e.g., othionein promoter) or from ian viruses (e.g., the
adenovirus late promoter; the vaccinia virus 7.5K promoter). Bacterial cells such as
Escherichia coli, or eukaryotic cells, ally for the expression of whole recombinant
antibody molecule, are used for the expression of a recombinant antibody molecule. For
example, mammalian cells such as Chinese Hamster Ovary (CHO) cells, in conjunction
with a vector such as the major intermediate early gene promoter element from human
cytomegalovirus is an effective sion system for antibodies; see, e. g., Foecking et
al., Gene 45 (1986), 101; Cockett et al., Bio/Technology 8 (1990), 2.
The host cell line used for protein expression is often of ian origin; those
skilled in the art are credited with ability to determine particular host cell lines which are
best suited for the desired gene product to be expressed therein. Exemplary host cell lines
e, but are not limited to, CH0 (Chinese Hamster Ovary), DG44 and DUXBll
(Chinese Hamster Ovary lines, DHFR minus), HELA (human al carcinoma), CV1
(monkey kidney line), COS (a derivative of CV1 with SV40 T antigen), VERY, BHK
(baby hamster kidney), MDCK, W138, R1610 (Chinese hamster fibroblast) BALBC/3T3
(mouse fibroblast), HAK (hamster kidney line), SP2/O (mouse myeloma), P3x63-
Ag3.653 (mouse myeloma), BFA-lclBPT (bovine endothelial cells), RAJI (human
lymphocyte) and 293 (human kidney). Host cell lines are typically available from
commercial services, the an Tissue Culture Collection or from published
literature.
In addition, a host cell strain can be chosen which modulates the expression of the
inserted sequences, or modifies and processes the gene product in the specific fashion
desired. Such modifications (e. g., ylation) and processing (e. g., cleavage) of
protein products can be ant for the function of the protein. Different host cells have
characteristic and specific mechanisms for the post-translational sing and
modification of proteins and gene products. Appropriate cell lines or host systems can be
chosen to ensure the correct modification and processing of the foreign protein expressed.
To this end, eukaryotic host cells which possess the cellular machinery for proper
processing of the primary transcript, glycosylation, and phosphorylation of the gene
product can be used.
For long-term, high-yield production of recombinant ns, stable expression is
used. For example, cell lines which stably express the dy molecule can be
engineered. Rather than using expression vectors which contain viral s of
replication, host cells can be transformed with DNA controlled by appropriate expression
control elements (e.g, promoter, enhancer, sequences, transcription terminators,
polyadenylation sites, etc.), and a selectable marker. Following the introduction of the
n DNA, engineered cells are allowed to grow for, e.g, 1-2 days in an enriched
media, and then are ed to a selective media. The selectable marker in the
recombinant plasmid confers ance to the selection and allows cells to stably
integrate the plasmid into their chromosomes and grow to form foci which in turn can be
cloned and expanded into cell lines. This method can advantageously be used to engineer
cell lines which stably express the antibody molecule.
A number of selection s can be used, including but not limited to the herpes
simplex virus thymidine kinase (Wigler et al., Cell 11 (1977), 223), nthine-
guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA
48 (1992), 202), and adenine phosphoribosyltransferase (Lowy et al., Cell 22 (1980),
817) genes can be employed in tk-, hgprt- or aprt-cells, respectively. Also, etabolite
resistance can be used as the basis of selection for the following genes: dhfr, which
confers ance to methotrexate r et al., Natl. Acad. Sci. USA 77 (1980), 357;
O'Hare et al., Proc. Natl. Acad. Sci. USA 78 (1981), 1527); gpt, which confers resistance
to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA 78 (1981), 2072);
neo, which confers resistance to the aminoglycoside G-418 Goldspiel et al., Clinical
Pharmacy 12 , 488-505; Wu and Wu, Biotherapy 3 (1991), 87-95; Tolstoshev,
Ann. Rev. col. Toxicol. 32 (1993), 573-596; Mulligan, Science 260 (1993), 926-
932; and Morgan and Anderson, Ann. Rev. Biochem. 62 (1993), 191-217; TIB TECH 11
(1993), 155-215; and hygro, which confers resistance to hygromycin (Santerre et al.,
Gene 30 (1984), 147. Methods commonly known in the art of recombinant DNA
technology which can be used are described in Ausubel et al. (eds.), Current Protocols in
Molecular Biology, John Wiley & Sons, NY ; Kriegler, Gene Transfer and
Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and
W0 2012/177972
13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY
(1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981), which are incorporated by
reference herein in their entireties.
The expression levels of an antibody le can be sed by vector
amplification, for a review, see Bebbington and Hentschel, The use of vectors based on
gene amplification for the expression of cloned genes in mammalian cells in DNA
cloning, Academic Press, New York, Vol. 3. (1987). When a marker in the vector system
expressing antibody is amplifiable, increase in the level of inhibitor present in culture of
host cell will increase the number of copies of the marker gene. Since the amplified
region is associated with the antibody gene, production of the dy will also increase;
see Crouse et al., Mol. Cell. Biol. 3 , 257.
In vitro production allows scale-up to give large amounts of the desired
polypeptides. Techniques for mammalian cell cultivation under tissue e conditions
are known in the art and include homogeneous suspension culture, e.g. in an airlift reactor
or in a continuous stirrer reactor, or lized or entrapped cell culture, e. g. in hollow
fibers, microcapsules, on agarose eads or ceramic cartridges. If necessary and/or
d, the solutions of polypeptides can be purified by the customary chromatography
methods, for example gel filtration, ion-exchange chromatography, chromatography over
DEAE-cellulose or immuno-affinity chromatography, e. g., after preferential biosynthesis
of a synthetic hinge region polypeptide or prior to or subsequent to the HIC
chromatography step described herein.
Genes encoding antibodies, or n-binding fragments, variants, or derivatives
thereof of the invention can also be expressed in mmalian cells such as bacteria or
insect or yeast or plant cells. Bacteria which readily take up nucleic acids include
members of the enterobacteriaceae, such as strains of Escherichia coli or Salmonella;
aceae, such as Bacillus subtilis; Pneumococcus; Streptococcus, and Haemophilus
influenzae. It will filrther be appreciated that, when expressed in bacteria, the
heterologous ptides typically become part of inclusion bodies. The heterologous
polypeptides must be isolated, purified and then led into onal molecules.
Where tetravalent forms of antibodies are desired, the subunits will then self-assemble
into tetravalent antibodies; see, e.g., international application W002/096948.
In bacterial systems, a number of expression vectors can be advantageously
selected depending upon the use intended for the antibody molecule being sed. For
example, when a large quantity of such a protein is to be produced, for the generation of
ceutical itions of an antibody molecule, vectors which direct the
expression of high levels of fusion protein products that are readily purified can be used.
Such s include, but are not limited, to the E. coli expression vector pUR278 (Ruther
et al., EMBO J. 2 (1983), 1791), in which the antibody coding ce can be ligated
individually into the vector in frame with the lacZ coding region so that a filSlOI‘l protein is
produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13 (1985), 3101-3109; Van
Heeke & Schuster, J. Biol. Chem. 24 (1989), 5503-5509); and the like. pGEX vectors can
also be used to express foreign polypeptides as fusion proteins with glutathione S-
transferase (GST). In general, such fusion ns are soluble and can easily be purified
from lysed cells by adsorption and binding to a matrix of glutathione-agarose beads
followed by elution in the presence of free glutathione. The pGEX vectors are designed to
include thrombin or factor Xa protease cleavage sites so that the cloned target gene
t can be released from the GST moiety.
In addition to prokaryotes, eukaryotic microbes can also be used. Saccharomyces
cerevisiae, or common baker's yeast, is the most commonly used among otic
microorganisms although a number of other strains are commonly available, e.g., Pichia
pastoris. For expression in Saccharomyces, the d YRp7, for example, hcomb
et al., Nature 282 (1979), 39; Kingsman et al., Gene 7 (1979), 141; Tschemper et al.,
Gene 10 (1980), 157) is commonly used. This plasmid already contains the TRPl gene
which provides a selection marker for a mutant strain of yeast lacking the ability to grow
in tryptophan, for example ATCC No. 44076 or PEP4-l (Jones, Genetics 85 , 12).
The presence of the trpl lesion as a characteristic of the yeast host cell genome then
provides an effective environment for detecting transformation by grth in the absence
of tryptophan.
In an insect system, Autographa calz’form’ca nuclear polyhedrosis virus )
is typically used as a vector to s foreign genes. The virus grows in Spodoptera
frugz'perda cells. The antibody coding ce can be cloned individually into non-
essential regions (for example the polyhedrin gene) of the virus and placed under control
of an AcNPV promoter (for example the polyhedrin promoter).
PCT/U82012/043701
_ 61 _
Once an antibody molecule of the invention has been recombinantly expressed,
the whole antibodies, their dimers, dual light and heavy chains, or other
immunoglobulin forms of the present invention, can be purified according to standard
procedures of the art, including for example, by chromatography (e.g., ion exchange,
affinity, particularly by affinity for the specific antigen after Protein A, and sizing column
chromatography), centrifugation, differential solubility, e. g. ammonium sulfate
precipitation, or by any other standard technique for the purification of proteins; see, e.g.,
Scopes, "Protein Purification", Springer Verlag, NY. . Alternatively, a method for
increasing the affinity of antibodies of the invention is disclosed in US patent publication
2002-0123057 Al.
V. Fusion Proteins and Conjugates
In n embodiments, the antibody polypeptide comprises an amino acid
sequence or one or more moieties not normally associated with an antibody. ary
modifications are described in more detail below. For example, in some embodiments a
-chain fv antibody nt of the invention can comprise a flexible linker
sequence, or can be d to add a functional moiety (e.g., PEG, a drug, a toxin, or a
label such as a cent, radioactive, enzyme, nuclear magnetic, heavy metal and the
like)
In certain embodiments, an antibody polypeptide of the invention comprises,
ts essentially of, or consists of a fusion protein. Fusion proteins are chimeric
molecules which comprise, for example, an immunoglobulin clein-binding domain
with at least one target binding site, and at least one heterologous portion, z'.e., a portion
with which it is not naturally linked in nature. The amino acid sequences can normally
exist in separate proteins that are brought together in the fusion polypeptide or they can
normally exist in the same protein but are placed in a new arrangement in the fusion
polypeptide. Fusion proteins can be created, for example, by chemical synthesis, or by
ng and translating a polynucleotide in which the peptide regions are encoded in the
desired relationship.
The term "heterologous" as applied to a polynucleotide or a polypeptide, means
that the cleotide or polypeptide is derived from a ct entity from that of the
rest of the entity to which it is being compared. For ce, as used herein, a
"heterologous polypeptide" to be fused to an antibody, or an antigen-binding fragment,
WO 77972 PCT/U82012/043701
_ 62 _
variant, or analog thereof is d from a non-immunoglobulin polypeptide of the same
species, or an immunoglobulin or non-immunoglobulin polypeptide of a different species.
As sed in more detail elsewhere herein, antibodies, or antigen-binding
fragments, variants, or derivatives thereof of the invention can r be recombinantly
fused to a logous polypeptide at the N- or C-terminus or ally conjugated
(including nt and non-covalent conjugations) to polypeptides or other
compositions. For example, antibodies can be recombinantly fused or conjugated to
molecules useful as labels in detection assays and effector molecules such as heterologous
polypeptides, drugs, radionuclides, or toxins; see, e. g., international applications
8495; WO9l/l4438; 2624; US patent no. 5,314,995; and European
patent application EP 0 396 387.
Antibodies, or antigen-binding fragments, variants, or derivatives f of the
invention can be composed of amino acids joined to each other by peptide bonds or
modified e bonds, z'.e., peptide isosteres, and can contain amino acids other than the
gene-encoded amino acids. Antibodies can be modified by natural processes, such as
posttranslational processing, or by chemical modification techniques which are well
known in the art. Such modifications are well described in basic texts and in more
detailed monographs, as well as in a voluminous ch literature. Modifications can
occur anywhere in the antibody, including the peptide backbone, the amino acid side-
chains and the amino or carboxyl termini, or on moieties such as carbohydrates. It will be
appreciated that the same type of modification can be t in the same or varying
degrees at several sites in a given antibody. Also, a given antibody can contain many
types of modifications. Antibodies can be branched, for example, as a result of
ubiquitination, and they can be cyclic, with or without ing. Cyclic, branched, and
branched cyclic antibodies can result from posttranslation natural processes or can be
made by synthetic methods. Modifications include acetylation, acylation, ADP-
lation, amidation, covalent attachment of flaVin, covalent attachment of a heme
moiety, covalent attachment of a tide or nucleotide derivative, covalent attachment
of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking,
cyclization, disulfide bond formation, demethylation, formation of covalent cross-links,
formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation,
glycosylation, GPI anchor formation, hydroxylation, iodination, methylation,
WO 77972 PCT/U82012/043701
_ 63 _
myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation,
prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated on of
amino acids to proteins such as arginylation, and tination; see, e.g, ns -
Structure And Molecular Properties, T. E. Creighton, W. H. Freeman and Company, New
York 2nd Ed., (1993); Posttranslational Covalent Modification 0f Proteins, B. C.
Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth.
Enzymol. 182 (1990), 626-646; Rattan et al., Ann. NY Acad. Sci. 663 (1992), 48-62).
The present invention also provides for 11 proteins comprising an antibody, or
antigen-binding fragment, variant, or derivative thereof, and a heterologous polypeptide.
In one embodiment, a filSlOI‘l protein of the ion comprises, consists essentially of, or
consists of, a polypeptide having the amino acid sequence of any one or more of the VH
s of an antibody of the ion or the amino acid ce of any one or more of
the VL regions of an antibody of the invention or fragments or ts f, and a
heterologous polypeptide sequence. In another ment, a fusion protein for use in
the diagnostic and treatment methods disclosed herein comprises, consists essentially of,
or consists of a polypeptide having the amino acid sequence of any one, two, three of the
VH-CDRs of an antibody, or fragments, variants, or derivatives thereof, or the amino acid
sequence of any one, two, three of the VL-CDRs of an antibody, or fragments, variants,
or tives thereof, and a heterologous polypeptide sequence. In one embodiment, the
fusion n comprises a polypeptide having the amino acid sequence of a VH-CDR3 of
an antibody of the present invention, or fragment, derivative, or variant thereof, and a
heterologous polypeptide sequence, which fusion protein specifically binds to a-
synuclein. In another embodiment, a filSlOI‘l protein comprises a polypeptide having the
amino acid sequence of at least one VH region of an antibody of the invention and the
amino acid sequence of at least one VL region of an antibody of the invention or
fragments, derivatives or variants thereof, and a heterologous ptide sequence. In
some embodiments, the VH and VL regions of the fusion n correspond to a single
source antibody (or scFv or Fab fragment) which specifically binds a-synuclein. In yet
another embodiment, a fusion protein for use in the diagnostic and treatment methods
disclosed herein comprises a polypeptide having the amino acid sequence of any one,
two, three or more of the VH CDRs of an antibody and the amino acid sequence of any
one, two, three or more of the VL CDRs of an antibody, or fragments or variants thereof,
PCT/U82012/043701
_ 64 _
and a heterologous polypeptide sequence. In certain embodiments, two, three, four, five,
six, or more of the VH-CDR(s) or VL-CDR(s) correspond to single source antibody (or
scFv or Fab nt) of the invention. Nucleic acid molecules encoding these fusion
proteins are also encompassed by the invention.
Exemplary filSlOIl ns reported in the literature include filSlOIlS of the T cell
receptor (Gascoigne et al., Proc. Natl. Acad. Sci. USA 84 , 2936-2940; CD4
(Capon et al., Nature 337 (1989), 525-531; Traunecker et al., Nature 339 (1989), 68-70;
Zettmeissl et al., DNA Cell Biol. USA 9 (1990), 347-353; and Bym et al., Nature 344
, 667-670); L-selectin g receptor) (Watson et al., J. Cell. Biol. 110 (1990),
2221-2229; and Watson et al., Nature 349 (1991), 164-167); CD44 (Aruffo et al., Cell 61
(1990), 1303-1313); CD28 and B7 (Linsley et al., J. Exp. Med. 173 (1991),721-730);
CTLA-4 (Lisley et al., J. Exp. Med. 174 (1991), 9); CD22 (Stamenkovic et al.,
Cell 66 (1991), 1133-1144); TNF receptor (Ashkenazi et al., Proc. Natl. Acad. Sci. USA
88 (1991), 10535-10539; Lesslauer et al., Eur. J. Immunol. 27 (1991), 886; and
Peppel et al., J. Exp. Med. 174 (1991), 1483-1489 (1991); and IgE receptor a (Ridgway
and Gorman, J. Cell. Biol. 115 (1991), Abstract No. 1448).
As discussed elsewhere herein, antibodies, or antigen-binding fragments, variants,
or derivatives thereof of the invention can be fused to logous polypeptides to
increase the in viva half life of the polypeptides or for use in immunoassays using
methods known in the art. For e, in one embodiment, PEG can be conjugated to
the dies of the invention to increase their half-life in viva; see, e. g., Leong et al.,
Cytokine 16 , 106-119; Adv. in Drug Deliv. Rev. 54 (2002), 531; or Weir et al.,
m. Soc. Transactions 30 , 512.
Moreover, antibodies, or antigen-binding fragments, variants, or derivatives
thereof of the invention can be fused to marker sequences, such as a peptide to facilitate
their purification or detection. In certain embodiments, the marker amino acid sequence is
a hexa-histidine peptide (HIS), such as the tag provided in a pQE vector (QIAGEN, Inc.,
9259 Eton Avenue, Chatsworth, Calif, , among others, many of which are
commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci. USA 86
(1989), 821-824, for instance, hexa-histidine provides for convenient purification of the
fusion protein. Other peptide tags useful for purification include, but are not limited to,
the "HA" tag, which corresponds to an epitope derived from the influenza hemagglutinin
protein (Wilson et al., Cell 37 (1984), 767) and the "flag" tag.
Fusion proteins can be prepared using s that are well known in the art; see
for example US patent nos. 5,116,964 and 5,225,538. The e site at which the fusion
is made can be selected empirically to optimize the secretion or binding characteristics of
the fiJsion protein. DNA encoding the fusion protein is then transfected into a host cell for
expression.
Antibodies of the present invention can be used in non-conjugated form or can be
conjugated to at least one of a y of molecules, e. g., to improve the therapeutic
ties of the le, to facilitate target detection, or for imaging or y of the
patient. Antibodies, or antigen-binding fragments, ts, or derivatives thereof of the
invention can be d or conjugated either before or after purification, when
purification is performed. In particular, antibodies, or antigen-binding fragments, variants,
or derivatives thereof of the invention can be conjugated to therapeutic agents, prodrugs,
peptides, ns, enzymes, viruses, lipids, biological response modifiers, pharmaceutical
agents, or PEG.
Conjugates that are toxins including tional antibodies have been
widely described in the art. The toxins can be coupled to the antibodies by conventional
coupling techniques or immunotoxins containing protein toxin portions can be produced
as fusion proteins. The antibodies of the present invention can be used in a corresponding
way to obtain such toxins. Illustrative of such immunotoxins are those described
by Byers, Seminars Cell. Biol. 2 (1991), 59-70 and by Fanger, Immunol. Today 12
(1991), 51-54.
Those skilled in the art will appreciate that conjugates can also be assembled
using a variety of techniques depending on the selected agent to be conjugated. For
example, conjugates with biotin are prepared 6. g. by reacting an (x-synuclein binding
polypeptide with an activated ester of biotin such as the biotin N-hydroxysuccinimide
ester. Similarly, conjugates with a fluorescent marker can be prepared in the presence of a
coupling agent, 6.g. those listed herein, or by reaction with an isothiocyanate, such as
fluorescein-isothiocyanate. Conjugates of the antibodies, or antigen-binding fragments,
variants, or derivatives thereof of the invention are prepared in an ous manner.
PCT/U82012/043701
_ 66 _
The present invention r encompasses antibodies, or antigen-binding
nts, variants, or derivatives thereof of the invention conjugated to a diagnostic or
therapeutic agent. The antibodies can be used diagnostically to, for example, demonstrate
presence of a neurological disease, to indicate the risk of getting a neurological disease, to
monitor the development or progression of a neurological e, z'.e. synucleinopathic
disease as part of a clinical testing procedure to, e.g., ine the efficacy of a given
treatment and/or prevention regimen. Detection can be facilitated by coupling the
antibody, or antigen-binding fragment, variant, or derivative f to a detectable
substance. Examples of detectable substances include s enzymes, etic groups,
fluorescent materials, luminescent materials, bioluminescent materials, radioactive
materials, positron emitting metals using various positron emission aphies, and
nonradioactive paramagnetic metal ions; see, e.g., US patent no. 4,741,900 for metal ions
which can be conjugated to antibodies for use as diagnostics according to the present
invention. Examples of suitable enzymes include horseradish peroxidase, ne
phosphatase, B-galactosidase, or acetylcholinesterase; examples of le prosthetic
group complexes include streptavidin/biotin and avidin/biotin; examples of suitable
fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate,
rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an
example of a luminescent material includes luminol; es of bioluminescent
als include luciferase, luciferin, and aequorin; and examples of suitable radioactive
material include 125I, 131I, 111In or 99Tc.
An antibody, or antigen-binding fragment, variant, or tive thereof also can
be detectably labeled by coupling it to a chemiluminescent compound. The presence of
the uminescent-tagged antibody is then ined by detecting the presence of
luminescence that arises during the course of a chemical reaction. Examples of
particularly useful chemiluminescent ng compounds are luminol, isoluminol,
theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
One of the ways in which an antibody, or n-binding fragment, variant, or
derivative thereof can be ably labeled is by linking the same to an enzyme and
using the linked product in an enzyme immunoassay (EIA) (Voller, A., "The Enzyme
Linked Immunosorbent Assay (ELISA)" Microbiological Associates Quarterly
Publication, Walkersville, Md., Diagnostic Horizons 2 (1978), 1-7); Voller et al., J. Clin.
W0 2012/177972 PCT/U82012/043701
_ 67 _
Pathol. 31 (1978), 507-520; Butler, Meth. Enzymol. 73 (1981), 482-523; Maggio, E.
(ed.), Enzyme Immunoassay, CRC Press, Boca Raton, Fla., (1980); Ishikawa, E. et al.,
(eds.), Enzyme Immunoassay, Kgaku Shoin, Tokyo (1981). The enzyme, which is bound
to the dy will react with an appropriate substrate, such as a chromogenic substrate,
in such a manner as to produce a chemical moiety which can be detected, for example, by
spectrophotometric, fluorimetric or by Visual means. Enzymes which can be used to
detectably label the antibody include, but are not limited to, malate dehydrogenase,
staphylococcal nuclease, delta-S-steroid isomerase, yeast alcohol ogenase, alpha-
glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase,
alkaline atase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease,
urease, catalase, glucosephosphate dehydrogenase, glucoamylase and
acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric
methods which employ a chromogenic ate for the enzyme. Detection can also be
accomplished by Visual comparison of the extent of enzymatic reaction of a substrate in
comparison with similarly prepared standards.
Detection can also be accomplished using any of a variety of other immunoassays.
For example, by radioactively labeling the antibody, or antigen-binding fragment, variant,
or derivative thereof, it is possible to detect the antibody h the use of a
radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of
Radioimmanoassays, Seventh Training Course on Radioligand Assay Techniques, The
Endocrine Society, (March, , which is incorporated by reference herein). The
radioactive isotope can be detected by means including, but not limited to, a gamma
counter, a scintillation counter, or autoradiography.
An antibody, or antigen-binding fragment, variant, or derivative thereof can also
be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the
lanthanide series. These metals can be ed to the dy using such metal
ing groups as diethylenetriaminepentacetic acid (DTPA) or
nediaminetetraacetic acid (EDTA).
Techniques for conjugating various moieties to an antibody, or antigen-binding
fragment, variant, or derivative f are well known, see, e.g., Amon et al.,
"Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer y", in
Monoclonal Antibodies And Cancer Therapy, ld et al. (eds.), pp. 243-56 (Alan R.
PCT/U82012/043701
_ 68 _
Liss, Inc. (1985); Hellstrom et al., "Antibodies For Drug ry", in Controlled Drug
ry (2nd Ed.), Robinson et al. (eds.), Marcel Dekker, Inc., pp. 623-53 (1987);
Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review", in
Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds),
pp. 6 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use
Of Radiolabeled Antibody In Cancer Therapy", in Monoclonal Antibodies For Cancer
Detection And Therapy, Baldwin et al. (eds.), Academic Press pp. 303-16 (1985), and
Thorpe et al., "The Preparation And Cytotoxic Properties Of Antibody-Toxin
Conjugates", Imrnunol. Rev. 62 (1982), 119-158.
As mentioned, in certain embodiments, a moiety that enhances the stability or
efficacy of a binding molecule, e.g., a binding polypeptide, e. g., an antibody or
immunospecif1c nt thereof can be conjugated. For example, in one embodiment,
PEG can be conjugated to the binding molecules of the invention to increase their half-
life in vivo. Leong et al., Cytokine 16 (2001), 106; Adv. in Drug Deliv. Rev. 54 ,
531; or Weir et al., Biochem. Soc. ctions 30 (2002), 512.
VI. Compositions and Methods ofUse
The present invention relates to compositions comprising the aforementioned 0L-
ein binding molecule, e.g, antibody or antigen-binding fragment thereof of the
present invention or derivative or t thereof, or the cleotide, vector or cell of
the invention. The composition of the present invention can further comprise a
pharmaceutically acceptable carrier. Furthermore, the pharmaceutical composition of the
present invention can comprise further agents such as interleukins or interferons
depending on the intended use of the pharmaceutical composition. For example, for use in
the treatment of Parkinson’s disease the additional agent can be selected from the group
consisting of small organic molecules, x-synuclein antibodies, and combinations
thereof. Hence, in one ment the present invention relates to the use of the 0L-
synuclein binding molecule, e.g, dy or antigen-binding fragment thereof of the
present ion or of a binding molecule having substantially the same binding
specificities of any one thereof, the cleotide, the vector or the cell of the present
invention for the ation of a pharmaceutical or diagnostic composition for
prophylactic and therapeutic treatment of a synucleinopathic e, monitoring the
progression of a synucleinopathic disease or a response to a synucleinopathic disease
PCT/U82012/043701
_ 69 _
treatment in a subject or for determining a subject's risk for developing a synucleinopathic
disease.
Hence, in one embodiment the present invention s to a method of treating a
neurological disorder characterized by abnormal accumulation and/or deposition of 0L-
synuclein in the brain and the central nervous system, respectively, which method
comprises administering to a subject in need thereof a therapeutically effective amount of
an anti-u-synuclein binding molecule, antibody, cleotide, vector or cell of the
instant invention. In certain embodiments NI-202.22Dll or a fragment, variant or
derivative thereof is delivered. The term "neurological disorder" includes but is not
limited to synucleinopathic diseases such as Parkinson's disease (PD), dementia with
Lewy bodies (DLB), and multiple systems atrophy (MSA). Unless stated otherwise, the
terms neurodegenerative, neurological or neuropsychiatric are used interchangeably
A particular advantage of the therapeutic approach of the present invention lies in
the fact that the antibodies of the present invention are derived from B cells or B memory
cells from elderly subjects with no signs of Parkinsonism and thus are, with a certain
probability, capable of preventing a clinically manifest synucleinopathic disease, or of
diminishing the risk of the occurrence of the ally manifest disease, or of delaying
the onset or progression of the clinically manifest disease. Typically, the antibodies of the
t invention also have already successfully gone through c maturation, z'.e. the
optimization with respect to selectivity and effectiveness in the high affinity binding to
the target u-synuclein molecule by means of somatic ion of the variable regions of
the dy.
The knowledge that such cells in viva, e. g. in a human, have not been activated by
means of related or other logical ns or cell structures in the sense of an
autoimmunological or allergic reaction is also of great medical importance since this
es a considerably increased chance of successfully living through the clinical test
. So to speak, efficiency, acceptability and tolerability have already been
demonstrated before the preclinical and al development of the prophylactic or
therapeutic antibody in at least one human subject. It can thus be expected that the human
anti-(x-synuclein dies of the present ion, both its target structure-specific
W0 2012/177972 PCT/U82012/043701
_ 70 _
efficiency as therapeutic agent and its decreased probability of side effects significantly
increase its clinical probability of success.
The present invention also provides a pharmaceutical and diagnostic, respectively,
pack or kit comprising one or more containers filled with one or more of the above
described ingredients, e.g. anti-u-synuclein dy, binding fragment, derivative or
t thereof, polynucleotide, vector or cell of the present invention. Associated with
such container(s) can be a notice in the form prescribed by a governmental agency
regulating the manufacture, use or sale of pharmaceuticals or biological products, which
notice reflects al by the agency of manufacture, use or sale for human
administration. In addition or alternatively the kit comprises reagents and/or instructions
for use in appropriate diagnostic assays. The composition, 6.g. kit of the t invention
is of course particularly le for the risk assessment, diagnosis, prevention and
treatment of a disorder which is accompanied with the ce of u-synuclein, and in
particular applicable for the treatment of Parkinson’s disease (PD), dementia with Lewy
bodies (DLB) and multiple systems atrophy (MSA).
The pharmaceutical itions of the t ion can be formulated
according to s well known in the art; see for example Remington: The Science and
Practice of Pharmacy (2000) by the University of Sciences in Philadelphia, ISBN 0
306472. es of suitable pharmaceutical rs are well known in the art and
include phosphate buffered saline solutions, water, emulsions, such as oil/water
emulsions, various types of wetting agents, sterile solutions etc. Compositions comprising
such carriers can be formulated by well known conventional methods. These
pharmaceutical compositions can be administered to the subject at a suitable dose.
Administration of the suitable itions can be effected by different ways, e.g., by
intravenous, intraperitoneal, subcutaneous, intramuscular, intranasal, topical or
intradermal administration or spinal or brain delivery. Aerosol formulations such as nasal
spray formulations include purified aqueous or other solutions of the active agent with
preservative agents and isotonic agents. Such ations can be adjusted to a pH and
isotonic state compatible with the nasal mucous nes. Formulations for rectal or
vaginal administration can be presented as a suppository with a suitable carrier.
Furthermore, whereas the present invention includes the now standard (though
fortunately infrequent) procedure of drilling a small hole in the skull to administer a drug
W0 2012/177972 PCT/U82012/043701
_ 71 _
of the t invention, in a one aspect, the binding molecule, especially antibody or
antibody based drug of the present invention can cross the blood-brain barrier, which
allows for intravenous or oral administration.
The dosage regimen will be determined by the attending ian and clinical
factors. As is well known in the medical arts, dosages for any one patient depends upon
many factors, including the patient's size, body surface area, age, the particular compound
to be administered, sex, time and route of administration, l health, and other drugs
being administered concurrently. Preparations for parenteral administration include
sterile aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of non-
aqueous solvents are propylene glycol, hylene , vegetable oils such as olive
oil, and able organic esters such as ethyl oleate. Aqueous carriers include water,
alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered
media. Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose
and sodium chloride, lactated Ringer's, or fixed oils. Intravenous es e fluid
and nutrient replenishers, electrolyte replenishers (such as those based on Ringer's
dextrose), and the like. vatives and other additives can also be present such as, for
example, antimicrobials, anti-oxidants, chelating agents, and inert gases and the like.
Furthermore, the pharmaceutical ition of the invention can comprise fiarther
agents such as dopamine or psychopharmacologic drugs, depending on the intended use
of the pharmaceutical composition.
Furthermore, a pharmaceutical composition can be formulated as a vaccine, for
example, if the pharmaceutical ition of the invention comprises an anti-0L-
synuclein antibody or g nt, derivative or variant thereof for passive
immunization. As mentioned in the background section, oligomeric species of 0L-
synuclein have been reported extracellularly in plasma and CSF (El-Agnaf et al., FASEB
J. 20 (2006), 419—425) and passive immunization studies in mouse models of son’s
disease show that ellular mouse monoclonal antibodies against u-synuclein can
reduce accumulation of intracellular u-synuclein aggregates (Masliah et al., Neuron, 46
(2005), 857—868). Accordingly it is prudent to expect that the human anti-u-synuclein
antibodies and equivalent u-synuclein binding molecules of the present invention are
particularly useful as a vaccine for the tion or amelioration of synucleinopathic
diseases such as PD, DLB and MSA.
In one embodiment, it is ial to use recombinant Fab (rFab) and single chain
fragments (scFvs) of the antibody of the present invention, which might more readily
penetrate a cell ne. For example, Robert et al., Protein Eng. Des. Sel. (2008) Oct
16; 81741-0134, published online ahead, be the use of ic recombinant Fab
(rFab) and single chain fragments (scFvs) of monoclonal antibody WO-2 which
recognizes an epitope in the N-terminal region of AB. The engineered fragments were
able to (i) prevent amyloid fibrillization, (ii) disaggregate preformed ABl-42 fibrils and
(iii) t ABl-42 oligomer-mediated neurotoxicity in vitro as efficiently as the whole
IgG molecule. The perceived ages of using small Fab and scFv engineered
antibody formats which lack the effector fianction include more efficient passage across
the blood-brain barrier and minimizing the risk of triggering inflammatory side reactions.
Furthermore, besides scFv and single-domain antibodies retain the binding city of
full-length antibodies, they can be expressed as single genes and intracellularly in
mammalian cells as intrabodies, with the potential for alteration of the folding,
interactions, modifications, or subcellular localization of their targets; see for review, e.g.,
Miller and Messer, Molecular Therapy 12 (2005), 394—401.
In a different approach Muller et al., Expert Opin. Biol. Ther. (2005), 237-241,
describe a technology platform, so-called 'SuperAntibody Technology', which is said to
enable antibodies to be shuttled into living cells without harming them. Such cell-
penetrating antibodies open new diagnostic and therapeutic windows. The term
'TransMabs' has been coined for these antibodies.
A r embodiment es co-administration or sequential administration of
other neuroprotective agents useful for ng a synucleinopathic disease. In one
embodiment, the additional agent is comprised in the pharmaceutical ition of the
t invention. es of neuroprotective agents which can be used to treat a
subject include, but are not limited to, an acetylcholinesterase inhibitor, a glutamatergic
receptor antagonist, kinase inhibitors, HDAC inhibitors, anti-inflammatory agents,
divalproex sodium, or any combination thereof es of other rotective agents
that can be used concomitant with pharmaceutical composition of the present invention
are described in the art; see, e.g. international application WO2007/Oll907. In one
embodiment, the additional agent is dopamine or a dopamine receptor agonist.
W0 2012/177972 PCT/U82012/043701
_ 73 _
In a filrther embodiment of the present invention the u-synuclein binding
molecules, in particular antibodies of the present invention can also be co-administered or
administered before or after transplantation y with neural transplants or stem cell
y useful for treating a synucleinopathic disease. Such ches with transplants
of embryonic mesencephalic neurons have been performed in patients with Parkinson’s
disease with the aim of replacing the neurons that are lost in the disease and reinstating
dopaminergic neurotransmission in the striatum. After 11-16 years post transplantation,
the d neurons were found to contain Lewy bodies and Lewy neurites. This spread of
u-synuclein pathology from the host to the grated tissues can be prevented by co-
administration of u-synuclein binding molecules, in particular antibodies of the present
ion.
A therapeutically effective dose or amount refers to that amount of the active
ingredient sufficient to ameliorate the symptoms or condition. Therapeutic efficacy and
toxicity of such compounds can be ined by standard pharmaceutical procedures in
cell cultures or experimental animals, e.g., ED50 (the dose therapeutically effective in
50% of the population) and LD50 (the dose lethal to 50% of the population). The dose
ratio between eutic and toxic effects is the therapeutic index, and it can be
expressed as the ratio, LDso/EDSO. In n embodiments, the therapeutic agent in the
composition is present in an amount sufficient to restore or preserve normal behavior
and/or cognitive ties in case of PD, DLB or other synucleinopathic diseases.
From the foregoing, it is evident that the present invention encompasses any use
of an clein binding molecule comprising at least one CDR of NI-202.22Dll or
fragments, variants, or derivatives thereof, in particular for diagnosing and/or treatment of
a synucleinopathic disease as mentioned above. The g molecule can be an antibody
of the present invention or an immunoglobulin chain thereof. In addition, the present
invention relates to anti-idiotypic antibodies of any one of the mentioned antibodies
described herein. These are antibodies or other binding molecules which bind to the
unique antigenic peptide sequence located on an antibody's le region near the
antigen-binding site and are useful, e.g., for the ion of anti-(x-synuclein antibodies in
sample of a t.
In another embodiment the present invention relates to a diagnostic composition
comprising any one of the above described clein binding molecules, antibodies,
W0 2012/177972 PCT/U82012/043701
_ 74 _
antigen-binding fragments, polynucleotides, s or cells of the invention and
optionally suitable means for detection such as reagents conventionally used in immuno
or nucleic acid based diagnostic methods. The antibodies of the invention are, for
example, suited for use in immunoassays in which they can be utilized in liquid phase or
bound to a solid phase carrier. Examples of assays which can utilize the antibody
of the invention are competitive and non-competitive immunoassays in either a direct or
indirect format. Examples of such immunoassays are the radioimmunoassay (RIA), the
sandwich (immunometric , flow cytometry and the Western blot assay. The
antigens and antibodies of the invention can be bound to many different carriers and used
to isolate cells specif1cally bound thereto. Examples of well known carriers include glass,
polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, n,
nylon, amyloses, natural and d celluloses, polyacrylamides, agaroses, and
magnetite. The nature of the carrier can be either soluble or insoluble for the purposes of
the invention. There are many different labels and methods of labeling known to those of
ordinary skill in the art. es of the types of labels which can be used in the t
invention include enzymes, radioisotopes, colloidal metals, fluorescent compounds,
chemiluminescent compounds, and bioluminescent compounds; see also the embodiments
discussed hereinabove.
By a filrther embodiment, the u-synuclein binding molecules, in particular
dies of the present invention are used in a method for the diagnosis of a disorder in
an individual by obtaining a body fluid sample from the tested individual which can be a
blood sample, a lymph sample or any other body fluid sample and ting the body
fluid sample with an antibody of the instant invention under conditions enabling the
formation of antibody-antigen complexes. The level of such xes is then
determined by methods known in the art, a level significantly higher than that formed in a
control sample indicating the disease in the tested individual. In the same manner, the
specific n bound by the antibodies of the invention can also be used. Thus, the
present invention relates to an in vitro immunoassay comprising the binding molecule,
e.g., antibody or antigen-binding fragment thereof of the invention.
In this context, the present invention also s to means specifically designed
for this purpose. For e, an antibody-based array can be used, which is for example
loaded with antibodies or equivalent antigen-binding molecules of the present invention
which specifically ize u-synuclein. Design of microarray immunoassays is
summarized in Kusnezow et al., Mol. Cell Proteomics 5 (2006), 696. Accordingly,
the present invention also relates to microarrays loaded with u-synuclein binding
molecules fied in accordance with the present invention.
In one embodiment, the t invention relates to a method of diagnosing a
synucleinopathic disease in a subject, the method comprising:
(a) assessing the level, localization, conformation or a combination thereof of
u-synuclein in a subject to be diagnosed with the antibody or fragment thereof of any one
of the invention and
(b) comparing the level, localization, conformation or combination thereof of
u-synuclein in the subject to one or more reference rds derived from one or more
l samples,
wherein a difference or similarity between the level, localization, conformation or
ation thereof of u-synuclein in the subject and the nce standard indicates
whether the subject has a synucleinopathic disease.
The subject to be diagnosed can be asymptomatic or nical for the disease.
The reference rd can be from a patient with a synucleinopathic disease, for example
PD, DLB or MSA, where a similarity n the level, localization, conformation or
combination thereof of u-synuclein in the subject to be diagnosed and the reference
standard indicates that the subject to be diagnosed has a synucleinopathic disease.
Alternatively, or in addition a reference standard is derived from a subject does not have a
einopathic disease. In n embodiments, the subject to be diagnosed and the
reference standard(s) are age-matched. The analysis can be done in vivo, or via a sample
isolated from the subject to be diagnosed, e. g., any body fluid suspected to contain 0L-
synuclein, for example a blood, CSF, or urine sample
The level, localization, and/or conformation of u-synuclein can be assessed by any
le method known in the art comprising, e.g., analyzing u-synuclein by one or more
techniques chosen from Western blot, immunoprecipitation, -linked
immunosorbent assay (ELISA), radioimmunoassay (RIA), fluorescent activated cell
sorting (FACS), two-dimensional gel electrophoresis, mass spectroscopy (MS), matrix-
assisted laser desorption/ionization-time of flight-MS (MALDI-TOF), surface-enhanced
laser desorption ionization-time of flight (SELDI-TOF), high performance liquid
W0 77972 2012/043701
_ 76 _
chromatography (HPLC), fast n liquid chromatography (FPLC), multidimensional
liquid chromatography (LC) followed by tandem mass spectrometry (MS/MS), and laser
densitometry. In vivo imaging of (x-synuclein can ses positron emission
tomography (PET), single photon emission tomography (SPECT), near infrared (NIR)
optical imaging or magnetic nce imaging (MRI).
Methods of diagnosing a synucleinopathic disease such as PD, DLB, or MSA,
monitoring a synucleinopathic disease progression, and monitoring a synucleinopathic
disease treatment using antibodies and related means which can be adapted in accordance
with the t invention are also described in international application
/011907. Similarly, antibody based detection methods for (x-synuclein are
described in international applications WO99/50300, W02005/047860, /021255
and W02008/103472, the disclosure content of all being incorporated herein by
reference. Those methods can be applied as described but with an (x-synuclein specific
antibody, binding fragment, derivative or t of the present invention.
These and other embodiments are sed and encompassed by the description
and examples of the present invention. Further ture concerning any one of the
materials, methods, uses and compounds to be employed in accordance with the present
invention can be retrieved from public libraries and databases, using for example
electronic devices. For example the public database "Medline" can be utilized, which is
hosted by the National Center for Biotechnology Information and/or the National y
of ne at the National Institutes of Health. Further databases and web addresses,
such as those of the European Bioinformatics ute (EBI), which is part of the
European Molecular Biology Laboratory (EMBL) are known to the person skilled in the
art and can also be obtained using intemet search engines. An ew of patent
information in biotechnology and a survey of relevant sources of patent information
useful for pective searching and for current awareness is given in Berks, TIBTECH
12 (1994), 352-364.
The above disclosure generally describes the present invention. Unless otherwise
stated, a term as used herein is given the definition as provided in the Oxford Dictionary
of Biochemistry and Molecular Biology, Oxford University Press, 1997, revised 2000 and
reprinted 2003, ISBN 0 19 850673 2. Several documents are cited throughout the text of
this specification. The contents of all cited references (including literature references,
2012/043701
_ 77 _
issued s, published patent applications as cited throughout this application and
manufacturer's specifications, instructions, etc) are hereby expressly incorporated by
reference; however, there is no ion that any document cited is indeed prior art as to
the present invention.
A more complete understanding can be obtained by nce to the ing
specific examples which are provided herein for purposes of illustration only and are not
intended to limit the scope of the invention.
EXAMPLES
The examples which follow fiarther illustrate the invention, but should not be
construed to limit the scope of the invention in any way. The ing experiments in
Examples 1 and 2 are illustrated and described with respect to antibody NI-202.3G12, NI-
F4, and NI-202.3D8 as cloned, z'.e. containing primer induced mutations at the very
N—termini of the framework 1 Ig-variable regions and not being adjusted to the germ line
(GL) sequences of human variable heavy and light chains; see Figure 1. However, the
other antibodies of the NI-202 series, in particular those with the adjusted GL sequences
are structurally similar and thus can be expected to provide comparable results. These
antibodies were expressed as human IgG1 molecules. The experiments in examples 3 and
4 are illustrated and described with respect to dy NI-202.12F4 with primer d
mutations at the N—termini of the Ig-variable regions being adjusted to the germ line (GL)
sequences of human variable heavy and light chains; see Figure 1. This antibody was
sed as a chimeric le where the adjusted human variable domains were fused
to mouse IgG2a constant regions to allow for chronic dosing studies in transgenic mouse
models without to induce a mouse anti-human immune response.
Material and methods
ed descriptions of conventional methods, such as those employed herein can
be found in the cited literature. Unless indicated otherwise below, identification of (X-
synuclein-speciflc B cells and molecular g of u-synuclein antibodies displaying
specificity of interest as well as their recombinant expression and functional
characterization has been or can be performed as described in the Examples and
Supplementary Methods section of international application
W0 2012/177972 PCT/U82012/043701
_ 78 _
published as W02008/081008 and of l86 published as
W02010/069603, the disclosure content of which are incorporated herein by reference in
their entireties.
Purification of antigen
Recombinant His-u-synuclein was obtained by recombinant expression in
Escherichia coli and subsequent purification using heat induced precipitation, Nickel
y-, anion exchange- and size exclusion-chromatography.
For example, a DNA uct comprising the cDNA encoding u-synuclein under
the control of the T7 promotor was used to transform an appropriate Escherichia coli
strain such as BL21(DE3) and expression of 200 ml cell culture was induced by the
addition of lmM isopropyl iogalactopyranoside (IPTG). Cells were ted after
4 hrs induction at 37°C and then resuspended in 20 ml 50mM Tris, 150 mM NaCl pH 8,
followed by sonification. After boiling for 15 min, the heat resistant l7000g supernatant
was collected. Similar, heat-resistant l7000g supernatant from mock Escherichia coli was
collected. After heat resistant l7000g supernatant (20 ml) from Escherichia coli
expressing His-tagged u-synuclein was adjusted to 50 mM Tris, 300 mM NaCl, 20 mM
Imidazole, pH 8, it was loaded onto a HisTrap HP lml (GE Life e) column and
HIS-(x-synuclein was eluted with an 30-500mM ole gradient. Fractions ning
HIS-u-synuclein were pooled and then diluted 1:10 with 50 mM Tris pH 8. Diluted
pooled fractions were applied to a HiTrap Q HP lml (GE Life Science) column and
bound proteins were eluted in a 30-1000 mM NaCl gradient. Finally, eluates containing
HIS-(x-synuclein were further purified using high performance gel ion (Superdex
200 10/300 GL). This purification procedure yields HIS-u-synuclein with a purity grade
of around 99% as estimated by SDS-PAGE and Coomassie staining. Concentration of
purified n has been determined using a BCA assay (Pierce).
u-symuclein antibody ing
96 well half area Microplates (Corning) were coated with purified HIS-0t-
synuclein or d-synuclein (rPeptide) at a standard concentration of 2ug/ml in coating
buffer (PBS pH 9.6) ght at 4°C. Plates were washed in PBS-T pH 7.6 and non-
specific binding sites were blocked for 1 hr at RT with PBS-T containing 2% BSA
(Sigma, Buchs, Switzerland). B cell conditioned medium was preabsorbed for 1 hr at RT
with 10% Heat-resistant E. coli proteins in 1% BSA. This preabsorption step had been
developed after several previous attempts of ELISA screening were unsuccessful in
identifying human (x-synuclein c antibodies. Thus, fortunately it turned out that
orption of the ELISA plate with heat-resistant E. coli proteins excludes screening
for false positive hits such as sticky antibodies and antibodies directed against E. coli
protein inations probably present in purified recombinant (x-synuclein samples.
Preabsorbed medium was then transferred from memory B cell culture plates to ELISA
plates and incubated for 2 hrs at RT. ELISA plates were washed in PBS-T and then
incubated with horse radish peroxidase conjugated donkey anti-human IgG (Fcy
fragment specific) polyclonal antibodies. After washing with PBS-T, binding of human
antibodies was determined by measurement of HRP activity in a standard colorimetric
assay.
Molecular cloning of u-symuclein antibodies
Samples containing memory B cells were obtained from eers >60 years of
age. All eers had in common to lack any sign of Parkinsonism. Living B cells of
selected memory B cell cultures are ted and mRNA is prepared. Immunoglobulin
heavy and light chain sequences are then obtained using Ig-framework 1 specific primers
for all human variable heavy and light chain families as 5’ primers in combination with
primers specific for all human I segments (heavy and kappa light chain) and C segments
(lambda light chain) as 3’primers (Marks et (1]., Mol. Biol. 222 (1991), 581-597; de Haard
et al., J. Biol. Chem. 26 (1999), 18218-18230).
Identification of the antibody clone with the desired specificity is performed by re-
screening on ELISA upon recombinant expression of te dies. inant
expression of complete human IgGl dies or chimeric IgG2a antibodies is achieved
upon insertion of the variable heavy and light chain sequences "in the correct reading
frame" into expression vectors that complement the variable region ce with a
sequence encoding a leader peptide at the 5’-end and at the 3’-end with a sequence
encoding the appropriate constant domain(s). To that end the primers contained restriction
sites designed to tate cloning of the variable heavy and light chain sequences into
antibody expression vectors. Heavy chain immunoglobulin are expressed by inserting the
immunoglobulin heavy chain RT-PCR product in frame into a heavy chain expression
vector bearing a signal peptide and the nt domains of human immunoglobulin
gamma 1 or mouse immunoglobulin gamma 2a. Kappa light chain immunoglobulin is
sed by inserting the kappa light chain RT-PCR-product of NI-202.3D8 in frame
into a light chain expression vector providing a signal peptide and the constant domain of
human kappa light chain immunoglobulin. NI-202.12F4 and NI-202.3G12 lambda light
chain immunoglobulins are expressed by inserting the lambda light chain RT-PCR-
product in frame into a lambda light chain expression vector providing a signal peptide
and the nt domain of human or mouse lambda light chain immunoglobulin.
Functional inant monoclonal antibodies were obtained upon co-
ection into HEK293 or CHO cells (or any other appropriate recipient cell line of
human or mouse origin) of an Ig- heavy—chain expression vector and a kappa or lambda
Ig-light—chain expression . Recombinant human monoclonal antibody was
subsequently purified from the conditioned medium using a rd Protein A column
purification.
Antibodies
Pan synuclein antibody Syn211 (Sigma) was used according to manufacturer’s
protocol. Recombinant human u-synuclein antibodies N1202.22Gll and N1202.12F4 are
antibodies of this invention. They were expressed in HEK293 or CHO cells and then
conditioned media was directly used in subsequent applications unless otherwise stated.
Direct ELISA
Antigens were coated at ted concentration in PBS pH 9.6 onto 96 well half
area microplates (Corning) overnight at 4°C. Plates were washed in PBS-T pH 7.6 and
non-specific binding sites were blocked for 1 hr at RT with PBS-T ning 2% BSA
(Sigma). Probes (Primary antibodies) were then transferred to wells and incubated for 2
hrs at RT. After washing in PBS-T pH 7.6, wells were incubated with horse radish
peroxidase (HRP)-conjugated polyclonal anti-human (for recombinant human antibodies),
anti-rabbit (for pan synuclein antibody) or anti-mouse (for LB509 or Syn2l l) secondary
antibodies for 1 hr at RT. After rigorous washing in PBS-T, binding of probes was
ined by measurement of HRP ty in a standard colorimetric assay using
,5'-tetramethylbiphenyl-4,4'-diamine (Sigma) as chromogenic substrate.
_ 81 _
Peptide scan for epitope mapping
The entire sequence of human u-synuclein was synthesized as pping
peptides, with lengths of 15 amino acids (aa) and an overlap of ll aa, coupled via a
flexible linker to cellulose membrane (JPT, Berlin, Germany). A membrane that
comprises a total of 33 peptides was rinsed in methanol and then blocked with Rotiblock
(Roth, Karlsruhe, y). The membrane was ted with indicated antibodies
diluted in blocking solution and then with horse radish peroxidase (HRP)-labeled
secondary dy for 1 hr. Between incubations the membrane was washed 3x with
PBS-T for 5 min. The membrane was then developed using ECL plus Western Blotting
Detection Reagents (GE Healthcare).
In-solution ELISA
NI-202.12F4 (2 ug/ml) diluted in sodium bicarbonate buffer (pH 9.6) was coated
at 4°C overnight onto ELISA plates. Then the plate was blocked with 2% BSA PBS-T
and subsequently washed with PBS-T. Indicated biotinylated u-synuclein peptides were
added and after 2 hrs incubation the plates were washed with PBS-T. After incubation
with HRP labeled streptavidin for 1 hr, binding was determined by measurement of HRP
activity in a standard colorimetric assay.
Example 1: Human derived u-synuclein antibody NI-202.21Dll is selective for human 0L-
synuclein
u-,B- and y-synuclein are highly homologous proteins that are predominantly
expressed in the nervous system, skeletal muscle and heart. clein is ly linked
to a broad spectrum of CNS diseases whereas B-synuclein can be a neuroprotective
n. Thus the ion provides therapeutic antibodies against ogical 0L-
synuclein variants which do not cross react with B- and y-synuclein. In order to t
potential therapeutic use of NI-202.21Dl l, the antibody was tested for binding to (1-, B-
and y-synuclein in a direct ELISA. Recombinant u-,B and y-synuclein was coated onto
ELISA plates at equal concentration and then either incubated with recombinant NI-
202.21Dll or a control pan synuclein antibody. The pan-synuclein dy detects all
three synuclein proteins but NI-202.21Dll ys selective binding for u-synuclein (Fig
2a).
WO 77972 PCT/U82012/043701
_ 82 _
Human and mouse u-synuclein are highly conserved proteins. To probe if
recombinant NI-202.21Dll preferentially binds human vs murine (x-synuclein,
recombinant His-tagged human or murine (x-synuclein were coated onto ELISA plates at
equal tration and then tested for NI-202.21Dll and NI-202.12F4 g (Fig 2b).
NI-202.21Dll detects only human (x-synuclein s NI-202.12F4 detects both human
and murine (x-synuclein in this direct ELISA (see PCT Publication No.
Al). Together these findings demonstrate that NI-202.21Dll is highly selective for
human u-synuclein.
Example 2: NI-202.21Dll shows preferential binding to human a-synuclein at high
coating concentrations pointing to a conformational epitope
The half maximal effective concentration (ECSO) ting the potency of NI-
202.21Dll was determined for low and high coating concentrations of recombinant 0t-
synuclein using a direct (x-synuclein ELISA. High affinity binding of recombinant NI-
202.21Dll with an ECSO of ~200 pM was observed for high coating concentrations of 0L-
synuclein protein (20 ug/ml). At lower concentrations of (x-synuclein, a sharp decrease in
affinity was observed (Fig 3). These characteristics are in strong contrast to commercially
available dy synle that is also detecting an epitope in the C-terminal domain of 0t-
synuclein. This finding suggests that NI-202.21Dll prefers an epitope that is formed or
exposed under high density conditions such as found in high molecular weight species of
u-synuclein.
e 3: Recombinant NI-202.22Dll binds to ogical (x-synuclein species in the
brain.
Binding of .21Dll to human (x-synuclein was filrther characterized by
immunohistochemical ng of brain sections from uclein transgenic mice and
from a patient with a neuropathologically confirmed synucleinopathy (Dementia with
Lewy ). .21Dll shows prominent staining of Lewy Body and Lewy Neurite
like inclusions on Proteinase K treated paraffin sections from brain tissue of transgenic
mice overexpressing human u-synuclein A53T (Fig 4a). No N1202-21Dll staining was
detected in brain sections from wild-type mice supporting that NI-202.21Dll is specific
for human (x-synuclein (Fig 4b). NI-202.21Dll also detected pathological (x-synuclein in
human brain tissue of a patient with Dementia with Lewy Body (Fig 4c). These results
PCT/U82012/043701
_ 83 _
show that human-derived antibody NI-202.21D11 detects pathological u-synuclein in
brain.
Example 4: Mapping the epitope of human derived clein-specific antibody NI-
D11 to an epitope within C-terminal domain of human clein.
u-synuclein is a 140 amino acids (aa) long natively unfolded protein that is
ed of three domains. These are the N-terminal amphipathic repeat region (aa 1-
60), the center region (aa 61-95) and the acidic C-terminal region (aa 96-140). (A) In
order to get an initial understanding for the .21D11 binding domain, recombinant
u-synuclein truncations were tested for NI-202.21D11 binding in a direct ELISA.
Recombinant clein truncations from residues 1-60, 1-95, 61-140 and 96-140 were
coated onto ELISA plates and then incubated with recombinant NI-202.21D11. Binding
of NI-202.21D11 was only observed to u-synuclein truncations 61-140 and 96-140
demonstrating that NI-202.21D11 binds to the C-terminal acidic domain of u-synuclein
(Fig. 5a).
In order to understand the recognition sequence of NI-202.21D11 in more detail,
NI-202.21D11 was tested for binding to overlapping linear 15-mer peptides that cover the
entire human u-synuclein amino acid sequence. Adjacent peptides share an overlap of 11
residues and peptides are C-terminally spotted to a cellulose support membrane. NI-
202.21D11 bound to three pping peptides namely es 109-123 (B08), 113-127
(B09) and 117-131 (B10) of human u-synuclein (Fig. 5b). This result suggests the
minimal ition sequence within the C-terminus of u-synuclein required for NI-
202.21D11 binding is PVDPDNE (117-123). Notably, NI-202.21D11 bound peptide B10
slightly less than peptides B08 and B09. Thus es 113-117 within a-synuclein may
influence on NI-202.21D11 g.
Almost no binding of NI-202.21D11 to mouse u-synuclein was observed in a
direct ELISA (Fig. 2B). Sequence alignment of the determined epitope sequence of NI-
202.21D11 (PVDPEE) to the corresponding murine sequence E) suggest that
D121 and N122 are key amino acids for selectivity of NI-202.21D11 for human vs.
murine a-synuclein. In order to confirm the key role of D121/N 122, recombinant d
human a-synuclein D121G/N122S was produced and tested for NI202.21D11 binding in
a direct ELISA. As shown in Figure 5c NI-202.21D11 showed almost no binding to
human u-synuclein D121G/N122S compared to wt human u-synuclein. A control pan-
PCT/U82012/043701
_ 84 _
synuclein antibody was used as normalization l for equal coating of synuclein
proteins.
These results show that NI-202.21D11 is a human-derived uclein antibody
detecting a C-terminal epitope (residues 117-123) within human (x-synuclein and that
amino acids D12 1/N 122 contribute to human vs. murine (x-synuclein the selectivity.
Example 5: NI-202.12F4 detects epitope within (x-synuclein 4-15 and K10 in (x-synuclein
is key amino acid for (x-synuclein selectivity ofNI-202. 12F4
In order to understand the recognition sequence (epitope) of NI-202.12F4 in more
, overlapping linear 15-mer peptides that cover the entire human (x-synuclein amino
acid sequence were tested for NI-202.12F4 binding by immunoblotting. Adjacent
peptides share an overlap of 11 es and peptides were C-terminally spotted to a
cellulose support ne. NI-202.12F4 only bound to the very N-terminal peptide
(A01) g that epitope is within residues 1-15 (Fig. 6a). Since NI-202.12F4 does not
bind to peptide (A02) residues 5-20, the epitope starts between residues 1 and 5. To
determine the exact start e of the epitope, synthetic (x-synuclein peptides were tested
for .12F4 binding in an in-solutz'on binding ELISA. First in order to validate in-
solution binding ELISA, synthetic peptides (x-synuclein 1-30 and 5-30 were tested for NI-
202.12F4 binding. NI-202.12F4 bound (x-synuclein 1-30 but not 5-30 validating the assay
by confirming the epitope starts between residue 1 and 5 (Fig. 6b). Next, (x-synuclein 4-30
was tested for NI-202.12F4 binding. As show in Figure 6b NI-202.12F4 bound to 0L-
synuclein 4-30. These results show that NI-202.12F4 epitope starts at residue 4.
.12F4 ively bound to (x-synuclein but not B- and y-synuclein.
Sequence alignment of the NI-202.12F4 epitope containing sequence (u-synuclein 4-15)
with the ponding B-synuclein sequence showed that these sequences only differed
in one amino acid. Lysine at position 10 in (x-synuclein is replaced by methionine in B-
synuclein. Thus 12F4 should bind to clein M10K but not (x-synuclein
K10M. For experimental confirmation, recombinant wt and K10M (x-synuclein, and wt
and M10K B-synuclein were tested for NI-202.12F4 binding in a direct ELISA. As
predicted N1202.12F4 only bound to wt (x-synuclein and B-synuclein M10K but not to wt
B-synuclein and (x-synuclein K10M (Fig 6c). A pan-synuclein antibody bound to all four
recombinant proteins equally well demonstrating equal g onto ELISA plates.
W0 2012/177972 PCT/U82012/043701
_ 85 _
All er these experiments show that the epitope for N1202.12F4 is localized
within residues 4-15 of u-synuclein. The epitope starts at residue 4 and ends between
residue 11-15. Lysine at position 10 in (x-synuclein accounts for the specificity of NI-
202.12F4 for u- versus B-synuclein.
Claims (54)
1. An isolated antibody or n-binding fragment thereof that specifically binds to human α-synuclein (SEQ ID NO:1), said antibody or n-binding nt comprising: a VH CDR1 region comprising the amino acid ce of SEQ ID NO:16 or a ce that differs from SEQ ID NO:16 by one amino acid; a VH CDR2 region comprising the amino acid sequence of SEQ ID NO:17 or a sequence that differs from SEQ ID NO:17 by one amino acid; a VH CDR3 region comprising the amino acid sequence of SEQ ID NO:18 or a sequence that differs from SEQ ID NO:18 by one amino acid; a VL CDR1 region comprising the amino acid sequence of SEQ ID NO:23 or a sequence that differs from SEQ ID NO:23 by one amino acid; a VL CDR2 region comprising the amino acid sequence of SEQ ID NO:24 or a sequence that differs from SEQ ID NO:24 by one amino acid; and a VL CDR3 region comprising the amino acid sequence of SEQ ID NO:25 or a sequence that differs from SEQ ID NO:25 by one amino acid.
2. The antibody or antigen-binding nt of claim 1, comprising a VH and a VL that are respectively at least 95% identical to SEQ ID NO:15 and SEQ ID NO:22; SEQ ID NO:15 and SEQ ID NO:26; SEQ ID NO:20 and SEQ ID NO:22; or SEQ ID NO:20 and SEQ ID NO:26.
3. The antibody or antigen-binding fragment of claim 1 or claim 2, wherein the VH ses a polypeptide sequence at least 95% identical to SEQ ID NO:15 or SEQ ID NO:20.
4. The antibody or antigen-binding fragment of any one of claims 1 to 3, wherein the VH comprises SEQ ID NO:15 or SEQ ID NO:20.
5. The antibody or antigen-binding fragment of claim 1 or claim 2, wherein the VL comprises a polypeptide sequence at least 95% identical to SEQ ID NO:22 or SEQ ID NO:26.
6. The antibody or antigen-binding fragment of claim 1 or claim 2, wherein the VL comprises SEQ ID NO:22 or SEQ ID NO:26.
7. The antibody or antigen-binding fragment of claim 1 or claim 2, wherein the VH and VL respectively comprise: SEQ ID NO:15 and SEQ ID NO:22; SEQ ID NO:15 and SEQ ID NO:26; SEQ ID NO:20 and SEQ ID NO:22; or SEQ ID NO:20 and SEQ ID NO:26.
8. The antibody or antigen-binding fragment of claim 1, wherein: the CDR1, CDR2 and CDR3 regions of the VH comprise amino acid sequences SEQ ID NO:16, SEQ ID NO:17 and SEQ ID NO:18, respectively; and the CDR1, CDR2 and CDR3 regions of the VL se amino acid sequences SEQ ID NO:23, SEQ ID NO:24 and SEQ ID NO:25, respectively.
9. An isolated dy or n-binding fragment thereof that specifically binds to human α-synuclein (SEQ ID NO:1) comprising: a VH CDR1 region comprising the amino acid sequence of SEQ ID NO:16; a VH CDR2 region comprising the amino acid sequence of SEQ ID NO:17; a VH CDR3 region comprising the amino acid sequence of SEQ ID NO:18; a VL CDR1 region comprising the amino acid sequence of SEQ ID NO:23; a VL CDR2 region comprising the amino acid sequence of SEQ ID NO:24; and a VL CDR3 region comprising the amino acid sequence of SEQ ID NO:25.
10. The antibody or antigen-binding fragment thereof of claim 9, comprising: SEQ ID NO:15 and SEQ ID NO:22; SEQ ID NO:15 and SEQ ID NO:26; SEQ ID NO:20 and SEQ ID NO:22; or SEQ ID NO:20 and SEQ ID NO:26.
11. The antibody or antigen-binding fragment of any one of claims 1 to 10, further comprising a signal peptide fused to the VH.
12. The antibody or antigen-binding nt of any one of claims 1 to 11, further comprising a signal peptide fused to the VL.
13. The antibody or antigen-binding fragment of any one of claims 1 to 12, further comprising a CH1 domain fused to the VH.
14. The antibody or antigen-binding fragment of any one of claims 1 to 13, further comprising a CH2 domain fused to the VH.
15. The antibody or antigen-binding nt of any one of claims 1 to 14, further comprising a CH3 domain fused to the VH.
16. The antibody or antigen-binding fragment of any one of claims 1 to 15, further comprising a hinge region fused to the VH.
17. The antibody or antigen-binding fragment of any one of claims 1 to 16, further comprising a CK domain fused to the VL.
18. The antibody or antigen-binding fragment of any one of claims 1 to 16, further comprising a CL domain fused to the VL.
19. The antibody or antigen-binding fragment of any one of claims 1, 8, 9 and 11 to 18, wherein said antibody or antigen-binding fragment is ic.
20. The antibody or antigen-binding fragment of any one of claims 1 to 18, wherein said antibody or antigen-binding nt is fully human.
21. The antibody or antigen-binding nt of any one of claims 1 to 20, wherein said antibody or antigen-binding fragment is a Fab fragment, a Fab' fragment or a F(ab)2 fragment.
22. The antibody or n-binding fragment of any one of claims 1 to 20, wherein said antibody or antigen-binding fragment is a single chain antibody.
23. The antibody or antigen-binding fragment of any one of claims 1 to 20, wherein said antibody or antigen-binding fragment is a single-chain Fv fragment.
24. The antibody or n-binding fragment of any one of claims 1 to 23, further comprising a heterologous polypeptide fused thereto.
25. The antibody or antigen-binding fragment of any one of claims 1 to 24 conjugated to an agent selected from the group consisting of a eutic agent, a g, a peptide, a protein, an enzyme, a virus, a lipid, a biological response modifier, a pharmaceutical agent and PEG.
26. A composition comprising the antibody or antigen-binding fragment of any one of claims 1 to 25 and a carrier.
27. An isolated polynucleotide comprising a nucleic acid encoding the antibody or antigenbinding fragment thereof of any one of claims 1 to 25.
28. A composition comprising the polynucleotide of claim 27 and a carrier.
29. A vector comprising the polynucleotide of claim 27.
30. The vector of claim 29, wherein said polynucleotide is ly associated with a promoter.
31. The vector of claim 30, wherein the polynucleotide includes a polynucleotide encoding a VH and a polynucleotide encoding a VL and wherein the polynucleotide encoding the VH and the polynucleotide encoding the VL are co-transcribed from a single promoter operably associated ith, but are separately translated.
32. The vector of claim 31, r comprising an IRES sequence disposed between the polynucleotide encoding the VH and the cleotide encoding the VL.
33. The vector of claim 31, wherein the polynucleotide encoding the VH and the polynucleotide encoding the VL are tely transcribed, each being operably ated with a separate promoter.
34. The vector of claim 33, wherein each said separate promoter is ent.
35. An isolated host cell comprising the polynucleotide of claim 27 or the vector of any one of claims 29 to 34.
36. A method of producing an anti-human α-synuclein antibody or antigen-binding fragment thereof, said method comprising culturing the host cell of claim 35 and recovering the antibody or antigen-binding fragment thereof.
37. An anti-human clein antibody or antigen-binding fragment thereof produced by the method of claim 36.
38. Use of the antibody of antigen-binding fragment of any one of claims 1 to 25 and 37, the composition of claim 26 or claim 28, the polynucleotide of claim 27, the vector of any one of claims 29 to 34 or the host cell of claim 35 in the manufacture of a medicament for treating or preventing a synucleinopathic disease in a subject.
39. Use of the antibody or antigen-binding fragment of any one of claims 1 to 25 and 37 in the manufacture of a medicament for diagnosing whether a subject has a synucleinopathic disease, wherein said sing comprises: (a) assessing a level, localization, conformation or a combination thereof of lein in the t to be diagnosed with the medicament; and (b) comparing the level, localization, conformation or combination thereof of said αsynuclein in the subject to one or more reference standards derived from one or more control samples, wherein a difference or similarity between the level, localization, conformation or combination thereof of said α-synuclein in the subject and the one or more reference standards indicates whether the subject has a einopathic disease.
40. The use of claim 38 or claim 39, wherein the synucleinopathic disease is Parkinson's disease (PD), dementia with Lewy bodies (DLB), multiple systems y (MSA) or a combination thereof.
41. The use of any one of claims 38 to 40, n the subject is a mammal.
42. The use of claim 41, wherein the mammal is a human.
43. The use of claim 42, wherein the level, localization, conformation or combination f of α-synuclein in the subject is measured by in vivo imaging.
44. The use of claim 43, wherein said in vivo imaging comprises positron emission tomography (PET), single photon emission tomography (SPECT), near infrared (NIR) optical imaging or magnetic resonance imaging (MRI).
45. An isolated antibody or antigen-binding fragment thereof as defined in any one of claims 1 to 10 and as substantially described herein with reference to the section entitled “DETAILED DESCRIPTION OF THE INVENTION”.
46. A composition comprising the dy or antigen-binding nt of claim 45 and a
47. An isolated polynucleotide comprising a nucleic acid encoding the antibody or antigenbinding fragment of claim 45.
48. A ition comprising the polynucleotide of claim 47 and a carrier.
49. A vector comprising the polynucleotide of claim 47.
50. An isolated host cell comprising the polynucleotide of claim 47 or the vector of claim
51. A method of producing an anti-human α-synuclein antibody or antigen-binding fragment, said method comprising culturing the host cell of claim 50 and recovering the antibody or antigen-binding fragment thereof.
52. An anti-human α-synuclein antibody or antigen-binding fragment thereof produced by the method of claim 51.
53. Use of the antibody of n-binding fragment of claim 45 or claim 52, the composition of claim 46 or claim 48, the polynucleotide of claim 47, the vector of claim 49 or the host cell of claim 50 in the manufacture of a medicament for treating or preventing a synucleinopathic disease in a subject.
54. Use of the antibody or antigen-binding fragment of claim 45 or claim 52 in the manufacture of a medicament for diagnosing whether a subject has a synucleinopathic disease, wherein said sing comprises: (a) assessing a level, localization, conformation or a ation f of αsynuclein in the subject to be diagnosed with the medicament; and (b) comparing the level, localization, conformation or combination thereof of said lein in the subject to one or more nce standards derived from one or more control wherein a difference or similarity between the level, localization, mation or combination thereof of said α-synuclein in the subject and the one or more reference standards indicates whether the subject has a synucleinopathic disease.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161500580P | 2011-06-23 | 2011-06-23 | |
US61/500,580 | 2011-06-23 | ||
PCT/US2012/043701 WO2012177972A1 (en) | 2011-06-23 | 2012-06-22 | Anti-alpha synuclein binding molecules |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ618914A NZ618914A (en) | 2015-12-24 |
NZ618914B2 true NZ618914B2 (en) | 2016-03-30 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10301381B2 (en) | Anti-alpha synuclein binding molecules | |
US10703808B2 (en) | Human anti-alpha-synuclein antibodies | |
US11542322B2 (en) | Nano-theranostics for Parkinson's disease | |
NZ618914B2 (en) | Anti-alpha synuclein binding molecules | |
NZ709976B2 (en) | Human anti-tau antibodies | |
NZ709976A (en) | Human anti-tau antibodies |