NZ623275B2 - Treatment of ocular disease - Google Patents
Treatment of ocular disease Download PDFInfo
- Publication number
- NZ623275B2 NZ623275B2 NZ623275A NZ62327512A NZ623275B2 NZ 623275 B2 NZ623275 B2 NZ 623275B2 NZ 623275 A NZ623275 A NZ 623275A NZ 62327512 A NZ62327512 A NZ 62327512A NZ 623275 B2 NZ623275 B2 NZ 623275B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- ecd
- antibody
- hptpb
- use according
- ηρτρβ
- Prior art date
Links
- 201000010099 disease Diseases 0.000 title description 36
- 239000011230 binding agent Substances 0.000 claims abstract description 88
- 239000011780 sodium chloride Substances 0.000 claims abstract description 55
- 150000003839 salts Chemical class 0.000 claims abstract description 48
- 206010030113 Oedema Diseases 0.000 claims abstract description 40
- 239000003814 drug Substances 0.000 claims abstract description 11
- 238000004519 manufacturing process Methods 0.000 claims abstract description 9
- 102000004965 antibodies Human genes 0.000 claims description 131
- 108090001123 antibodies Proteins 0.000 claims description 131
- 239000003795 chemical substances by application Substances 0.000 claims description 94
- 102000004169 proteins and genes Human genes 0.000 claims description 44
- 108090000623 proteins and genes Proteins 0.000 claims description 44
- 230000027455 binding Effects 0.000 claims description 33
- 239000000427 antigen Substances 0.000 claims description 27
- 102000038129 antigens Human genes 0.000 claims description 27
- 108091007172 antigens Proteins 0.000 claims description 27
- 206010012688 Diabetic retinal oedema Diseases 0.000 claims description 21
- 201000011190 diabetic macular edema Diseases 0.000 claims description 21
- 108010045030 monoclonal antibodies Proteins 0.000 claims description 21
- 102000005614 monoclonal antibodies Human genes 0.000 claims description 20
- 239000003981 vehicle Substances 0.000 claims description 20
- 230000035693 Fab Effects 0.000 claims description 18
- 238000001356 surgical procedure Methods 0.000 claims description 18
- 239000000539 dimer Substances 0.000 claims description 16
- 229920001223 polyethylene glycol Polymers 0.000 claims description 13
- 210000004408 Hybridomas Anatomy 0.000 claims description 11
- 150000002500 ions Chemical class 0.000 claims description 11
- 229920002395 Aptamer Polymers 0.000 claims description 8
- 206010058202 Cystoid macular oedema Diseases 0.000 claims description 8
- 201000010206 cystoid macular edema Diseases 0.000 claims description 8
- 206010038886 Retinal oedema Diseases 0.000 claims description 7
- 239000003937 drug carrier Substances 0.000 claims description 7
- 108020004707 nucleic acids Proteins 0.000 claims description 7
- 150000007523 nucleic acids Chemical class 0.000 claims description 7
- 201000011195 retinal edema Diseases 0.000 claims description 7
- 108091008117 polyclonal antibodies Proteins 0.000 claims description 6
- PQEJXGNZBLONLG-XJDOXCRVSA-N (2R)-2-amino-3-[1-[3-[2-[4-[1,3-bis(2-methoxyethylcarbamoyloxy)propan-2-yloxy]butanoylamino]ethylamino]-3-oxopropyl]-2,5-dioxopyrrolidin-3-yl]sulfanylpropanoic acid Chemical compound COCCNC(=O)OCC(COC(=O)NCCOC)OCCCC(=O)NCCNC(=O)CCN1C(=O)CC(SC[C@H](N)C(O)=O)C1=O PQEJXGNZBLONLG-XJDOXCRVSA-N 0.000 claims description 4
- 238000010253 intravenous injection Methods 0.000 claims 1
- 238000010254 subcutaneous injection Methods 0.000 claims 1
- 239000007929 subcutaneous injection Substances 0.000 claims 1
- 239000000203 mixture Substances 0.000 description 58
- 206010029113 Neovascularisation Diseases 0.000 description 33
- 210000001508 Eye Anatomy 0.000 description 31
- 210000004027 cells Anatomy 0.000 description 31
- 101710037124 TEK Proteins 0.000 description 26
- 206010012689 Diabetic retinopathy Diseases 0.000 description 25
- 230000000694 effects Effects 0.000 description 23
- 239000000969 carrier Substances 0.000 description 22
- 208000004644 Retinal Vein Occlusion Diseases 0.000 description 21
- 238000000034 method Methods 0.000 description 21
- 150000001413 amino acids Chemical class 0.000 description 18
- 239000007924 injection Substances 0.000 description 17
- 208000002780 Macular Degeneration Diseases 0.000 description 15
- 206010038934 Retinopathy proliferative Diseases 0.000 description 15
- 206010012601 Diabetes mellitus Diseases 0.000 description 14
- 201000004569 blindness Diseases 0.000 description 14
- 210000004204 Blood Vessels Anatomy 0.000 description 13
- 210000001525 Retina Anatomy 0.000 description 13
- 208000007135 Retinal Neovascularization Diseases 0.000 description 13
- 208000005590 Choroidal Neovascularization Diseases 0.000 description 12
- 206010060823 Choroidal neovascularisation Diseases 0.000 description 12
- 206010022114 Injury Diseases 0.000 description 12
- 239000002202 Polyethylene glycol Substances 0.000 description 12
- 206010038932 Retinopathy Diseases 0.000 description 12
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 12
- 102100001937 PTPRB Human genes 0.000 description 11
- 101700082725 PTPRB Proteins 0.000 description 11
- 206010038923 Retinopathy Diseases 0.000 description 11
- 206010046851 Uveitis Diseases 0.000 description 11
- 238000002347 injection Methods 0.000 description 11
- 102000018358 Immunoglobulins Human genes 0.000 description 10
- 108060003951 Immunoglobulins Proteins 0.000 description 10
- 210000004379 Membranes Anatomy 0.000 description 10
- 239000012528 membrane Substances 0.000 description 10
- MYMOFIZGZYHOMD-UHFFFAOYSA-N oxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 10
- 229910052760 oxygen Inorganic materials 0.000 description 10
- 239000001301 oxygen Substances 0.000 description 10
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 10
- 125000003275 alpha amino acid group Chemical group 0.000 description 9
- 201000005667 central retinal vein occlusion Diseases 0.000 description 9
- PEDCQBHIVMGVHV-UHFFFAOYSA-N glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 9
- 229920000642 polymer Polymers 0.000 description 9
- -1 sine Polymers 0.000 description 9
- 239000000243 solution Substances 0.000 description 9
- 206010064930 Age-related macular degeneration Diseases 0.000 description 8
- 206010030032 Ocular disorder Diseases 0.000 description 8
- 229960000070 antineoplastic Monoclonal antibodies Drugs 0.000 description 8
- 230000004438 eyesight Effects 0.000 description 8
- 229960000060 monoclonal antibodies Drugs 0.000 description 8
- 239000008194 pharmaceutical composition Substances 0.000 description 8
- 230000004393 visual impairment Effects 0.000 description 8
- 241001465754 Metazoa Species 0.000 description 7
- 238000006366 phosphorylation reaction Methods 0.000 description 7
- 230000000865 phosphorylative Effects 0.000 description 7
- 229920001469 poly(aryloxy)thionylphosphazene Polymers 0.000 description 7
- 230000002207 retinal Effects 0.000 description 7
- 235000020945 retinal Nutrition 0.000 description 7
- 210000001519 tissues Anatomy 0.000 description 7
- 101700023370 Ang3 Proteins 0.000 description 6
- 229920002676 Complementary DNA Polymers 0.000 description 6
- 210000002889 Endothelial Cells Anatomy 0.000 description 6
- 102000004877 Insulin Human genes 0.000 description 6
- 108090001061 Insulin Proteins 0.000 description 6
- 206010061255 Ischaemia Diseases 0.000 description 6
- 208000001344 Macular Edema Diseases 0.000 description 6
- 206010025415 Macular oedema Diseases 0.000 description 6
- 229930002945 all-trans-retinaldehyde Natural products 0.000 description 6
- 125000000539 amino acid group Chemical group 0.000 description 6
- 239000002299 complementary DNA Substances 0.000 description 6
- 230000001276 controlling effect Effects 0.000 description 6
- 239000012530 fluid Substances 0.000 description 6
- 230000002401 inhibitory effect Effects 0.000 description 6
- 150000002632 lipids Chemical class 0.000 description 6
- 239000007788 liquid Substances 0.000 description 6
- 201000010230 macular retinal edema Diseases 0.000 description 6
- 102000004196 processed proteins & peptides Human genes 0.000 description 6
- 108090000765 processed proteins & peptides Proteins 0.000 description 6
- 239000011604 retinal Substances 0.000 description 6
- 230000002792 vascular Effects 0.000 description 6
- 229920002307 Dextran Polymers 0.000 description 5
- 229940072221 IMMUNOGLOBULINS Drugs 0.000 description 5
- 102000004851 Immunoglobulin G Human genes 0.000 description 5
- 108090001095 Immunoglobulin G Proteins 0.000 description 5
- 229920001850 Nucleic acid sequence Polymers 0.000 description 5
- 239000002033 PVDF binder Substances 0.000 description 5
- 241000700159 Rattus Species 0.000 description 5
- 208000001072 Type 2 Diabetes Mellitus Diseases 0.000 description 5
- 230000004913 activation Effects 0.000 description 5
- 230000015572 biosynthetic process Effects 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 230000004927 fusion Effects 0.000 description 5
- 108020001507 fusion proteins Proteins 0.000 description 5
- 102000037240 fusion proteins Human genes 0.000 description 5
- 238000001114 immunoprecipitation Methods 0.000 description 5
- 239000000463 material Substances 0.000 description 5
- 239000002609 media Substances 0.000 description 5
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 5
- 238000001262 western blot Methods 0.000 description 5
- 210000004369 Blood Anatomy 0.000 description 4
- 102100002977 CDR1 Human genes 0.000 description 4
- 239000004471 Glycine Substances 0.000 description 4
- 239000012593 Hanks’ Balanced Salt Solution Substances 0.000 description 4
- 206010025421 Macule Diseases 0.000 description 4
- 206010043189 Telangiectasia Diseases 0.000 description 4
- 208000009056 Telangiectasis Diseases 0.000 description 4
- 239000004480 active ingredient Substances 0.000 description 4
- 238000004458 analytical method Methods 0.000 description 4
- 239000012298 atmosphere Substances 0.000 description 4
- 239000008280 blood Substances 0.000 description 4
- 230000000875 corresponding Effects 0.000 description 4
- 230000002708 enhancing Effects 0.000 description 4
- 238000001943 fluorescence-activated cell sorting Methods 0.000 description 4
- 239000000499 gel Substances 0.000 description 4
- 238000000338 in vitro Methods 0.000 description 4
- 230000000302 ischemic Effects 0.000 description 4
- 239000006166 lysate Substances 0.000 description 4
- 229920000023 polynucleotide Polymers 0.000 description 4
- 239000002157 polynucleotide Substances 0.000 description 4
- 229920001184 polypeptide Polymers 0.000 description 4
- 238000007920 subcutaneous administration Methods 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 231100001005 telangiectasia Toxicity 0.000 description 4
- 230000001225 therapeutic Effects 0.000 description 4
- 230000003442 weekly Effects 0.000 description 4
- HBOCNGZYIAZFSU-UHFFFAOYSA-N 3-(4-hydroxyphenyl)-2-(phosphonoamino)propanoic acid Chemical group OP(=O)(O)NC(C(=O)O)CC1=CC=C(O)C=C1 HBOCNGZYIAZFSU-UHFFFAOYSA-N 0.000 description 3
- 229920000936 Agarose Polymers 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 230000036499 Half live Effects 0.000 description 3
- 108010001336 Horseradish Peroxidase Proteins 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 210000001957 Retinal Vein Anatomy 0.000 description 3
- 102000005632 Single-Chain Antibodies Human genes 0.000 description 3
- 108010070144 Single-Chain Antibodies Proteins 0.000 description 3
- 210000000952 Spleen Anatomy 0.000 description 3
- 229940035295 Ting Drugs 0.000 description 3
- 102000004142 Trypsin Human genes 0.000 description 3
- 108090000631 Trypsin Proteins 0.000 description 3
- 206010067584 Type 1 diabetes mellitus Diseases 0.000 description 3
- 210000001745 Uvea Anatomy 0.000 description 3
- 102100015249 VEGFA Human genes 0.000 description 3
- 108010073929 Vascular Endothelial Growth Factor A Proteins 0.000 description 3
- 238000010521 absorption reaction Methods 0.000 description 3
- 238000004166 bioassay Methods 0.000 description 3
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 description 3
- 230000015556 catabolic process Effects 0.000 description 3
- 150000001768 cations Chemical class 0.000 description 3
- 230000004059 degradation Effects 0.000 description 3
- 238000006731 degradation reaction Methods 0.000 description 3
- 239000008121 dextrose Substances 0.000 description 3
- 230000002255 enzymatic Effects 0.000 description 3
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 3
- 238000005755 formation reaction Methods 0.000 description 3
- DHMQDGOQFOQNFH-UHFFFAOYSA-N glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 3
- 230000003053 immunization Effects 0.000 description 3
- 239000007943 implant Substances 0.000 description 3
- 238000002647 laser therapy Methods 0.000 description 3
- 239000003446 ligand Substances 0.000 description 3
- 239000012139 lysis buffer Substances 0.000 description 3
- 230000001404 mediated Effects 0.000 description 3
- 230000003472 neutralizing Effects 0.000 description 3
- 238000002360 preparation method Methods 0.000 description 3
- 230000000750 progressive Effects 0.000 description 3
- 230000002062 proliferating Effects 0.000 description 3
- HEMHJVSKTPXQMS-UHFFFAOYSA-M sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 241000894007 species Species 0.000 description 3
- 238000006467 substitution reaction Methods 0.000 description 3
- 238000003786 synthesis reaction Methods 0.000 description 3
- 230000002194 synthesizing Effects 0.000 description 3
- 238000001890 transfection Methods 0.000 description 3
- 230000001131 transforming Effects 0.000 description 3
- 239000012588 trypsin Substances 0.000 description 3
- 229960001322 trypsin Drugs 0.000 description 3
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 3
- 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 3
- 101710027066 ALB Proteins 0.000 description 2
- 229940035674 ANESTHETICS Drugs 0.000 description 2
- 229940116904 ANTIINFLAMMATORY THERAPEUTIC RADIOPHARMACEUTICALS Drugs 0.000 description 2
- 206010002329 Aneurysm Diseases 0.000 description 2
- 244000303258 Annona diversifolia Species 0.000 description 2
- 235000002198 Annona diversifolia Nutrition 0.000 description 2
- 210000001775 Bruch Membrane Anatomy 0.000 description 2
- 241000220450 Cajanus cajan Species 0.000 description 2
- 241000282836 Camelus dromedarius Species 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 210000004087 Cornea Anatomy 0.000 description 2
- IQFYYKKMVGJFEH-XLPZGREQSA-N DEOXYTHYMIDINE Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 2
- 241000196324 Embryophyta Species 0.000 description 2
- 208000009745 Eye Disease Diseases 0.000 description 2
- 108050007372 Fibroblast growth factor family Proteins 0.000 description 2
- 102000018233 Fibroblast growth factor family Human genes 0.000 description 2
- 206010018987 Haemorrhage Diseases 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 102000003839 Human Proteins Human genes 0.000 description 2
- 108090000144 Human Proteins Proteins 0.000 description 2
- 108010064750 Humanized Monoclonal Antibodies Proteins 0.000 description 2
- 102000015434 Humanized Monoclonal Antibodies Human genes 0.000 description 2
- 241000282619 Hylobates lar Species 0.000 description 2
- RAXXELZNTBOGNW-UHFFFAOYSA-N Imidazole Chemical compound C1=CNC=N1 RAXXELZNTBOGNW-UHFFFAOYSA-N 0.000 description 2
- 102000004854 Immunoglobulin M Human genes 0.000 description 2
- 101710026201 JCHAIN Proteins 0.000 description 2
- 102100007033 JCHAIN Human genes 0.000 description 2
- 208000009857 Microaneurysm Diseases 0.000 description 2
- 108091007229 NSP3 Papain-like protease domain Proteins 0.000 description 2
- 229940074726 OPHTHALMOLOGIC ANTIINFLAMMATORY AGENTS Drugs 0.000 description 2
- 108010039918 Polylysine Proteins 0.000 description 2
- 108020004511 Recombinant DNA Proteins 0.000 description 2
- 206010038867 Retinal haemorrhage Diseases 0.000 description 2
- 210000003786 Sclera Anatomy 0.000 description 2
- 210000002966 Serum Anatomy 0.000 description 2
- 210000003491 Skin Anatomy 0.000 description 2
- 241000700605 Viruses Species 0.000 description 2
- 239000002671 adjuvant Substances 0.000 description 2
- 230000000240 adjuvant Effects 0.000 description 2
- 230000001058 adult Effects 0.000 description 2
- 230000003444 anaesthetic Effects 0.000 description 2
- 239000002260 anti-inflammatory agent Substances 0.000 description 2
- 230000000845 anti-microbial Effects 0.000 description 2
- 230000010056 antibody-dependent cellular cytotoxicity Effects 0.000 description 2
- 239000004599 antimicrobial Substances 0.000 description 2
- 230000001580 bacterial Effects 0.000 description 2
- 150000001720 carbohydrates Chemical class 0.000 description 2
- 235000014633 carbohydrates Nutrition 0.000 description 2
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 2
- 108091006028 chimera Proteins 0.000 description 2
- HVYWMOMLDIMFJA-DPAQBDIFSA-N cholesterol group Chemical group [C@@H]1(CC[C@H]2[C@@H]3CC=C4C[C@@H](O)CC[C@]4(C)[C@H]3CC[C@]12C)[C@H](C)CCCC(C)C HVYWMOMLDIMFJA-DPAQBDIFSA-N 0.000 description 2
- 238000003776 cleavage reaction Methods 0.000 description 2
- 230000002354 daily Effects 0.000 description 2
- 229940079593 drugs Drugs 0.000 description 2
- 238000004520 electroporation Methods 0.000 description 2
- 238000005516 engineering process Methods 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
- 238000001502 gel electrophoresis Methods 0.000 description 2
- 239000003193 general anesthetic agent Substances 0.000 description 2
- 238000010191 image analysis Methods 0.000 description 2
- 238000010348 incorporation Methods 0.000 description 2
- 239000003112 inhibitor Substances 0.000 description 2
- 238000003780 insertion Methods 0.000 description 2
- 239000002608 ionic liquid Substances 0.000 description 2
- 230000000670 limiting Effects 0.000 description 2
- 239000002502 liposome Substances 0.000 description 2
- 238000000520 microinjection Methods 0.000 description 2
- 239000011859 microparticle Substances 0.000 description 2
- 239000003607 modifier Substances 0.000 description 2
- 229920001542 oligosaccharide Polymers 0.000 description 2
- 150000002482 oligosaccharides Polymers 0.000 description 2
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 2
- 229920000656 polylysine Polymers 0.000 description 2
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 2
- 230000035755 proliferation Effects 0.000 description 2
- DNIAPMSPPWPWGF-UHFFFAOYSA-N propylene glycol Chemical group CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 2
- 238000000746 purification Methods 0.000 description 2
- 108020003175 receptors Proteins 0.000 description 2
- 102000005962 receptors Human genes 0.000 description 2
- 239000004017 serum-free culture media Substances 0.000 description 2
- 231100000486 side effect Toxicity 0.000 description 2
- BQCADISMDOOEFD-UHFFFAOYSA-N silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 2
- 229910052709 silver Inorganic materials 0.000 description 2
- 239000004332 silver Substances 0.000 description 2
- VMHLLURERBWHNL-UHFFFAOYSA-M sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 2
- 239000001632 sodium acetate Substances 0.000 description 2
- 235000017281 sodium acetate Nutrition 0.000 description 2
- 150000003431 steroids Chemical class 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 230000002522 swelling Effects 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 231100000419 toxicity Toxicity 0.000 description 2
- 230000001988 toxicity Effects 0.000 description 2
- GDBQQVLCIARPGH-ULQDDVLXSA-N (2S)-2-acetamido-N-[(2S)-1-[[(2S)-5-(diaminomethylideneamino)-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]-4-methylpentanamide Chemical compound CC(C)C[C@H](NC(C)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@H](C=O)CCCN=C(N)N GDBQQVLCIARPGH-ULQDDVLXSA-N 0.000 description 1
- LRJUYAVTHIEHAI-WRENIRACSA-N (2S,3R,5S,10R,11R,13R,14S)-17-[(2R,3R)-2,3-dihydroxy-6-methylheptan-2-yl]-2,3,5,11,14-pentahydroxy-10,13-dimethyl-1,2,3,4,9,11,12,15,16,17-decahydrocyclopenta[a]phenanthren-6-one Chemical compound C1[C@@H](O)[C@@H](O)C[C@]2(C)C([C@H](O)C[C@@]3(C([C@@](C)(O)[C@H](O)CCC(C)C)CC[C@]33O)C)C3=CC(=O)[C@@]21O LRJUYAVTHIEHAI-WRENIRACSA-N 0.000 description 1
- 125000004169 (C1-C6) alkyl group Chemical group 0.000 description 1
- YFDSDPIBEUFTMI-UHFFFAOYSA-N 2,2,2-tribromoethanol Chemical compound OCC(Br)(Br)Br YFDSDPIBEUFTMI-UHFFFAOYSA-N 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- TVZGACDUOSZQKY-LBPRGKRZSA-N 4-aminofolic acid Chemical compound C1=NC2=NC(N)=NC(N)=C2N=C1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 TVZGACDUOSZQKY-LBPRGKRZSA-N 0.000 description 1
- ZTOJFFHGPLIVKC-CLFAGFIQSA-N ABTS Chemical compound S/1C2=CC(S(O)(=O)=O)=CC=C2N(CC)C\1=N\N=C1/SC2=CC(S(O)(=O)=O)=CC=C2N1CC ZTOJFFHGPLIVKC-CLFAGFIQSA-N 0.000 description 1
- 229940074728 ANTIINFECTIVE OPHTHALMOLOGICS Drugs 0.000 description 1
- 229960003896 Aminopterin Drugs 0.000 description 1
- 206010059245 Angiopathy Diseases 0.000 description 1
- 102000009088 Angiopoietin-1 Human genes 0.000 description 1
- 108010048154 Angiopoietin-1 Proteins 0.000 description 1
- 229940006211 Anticholinergic mydriatics and cycloplegics Drugs 0.000 description 1
- 229940065524 Anticholinergics inhalants for obstructive airway diseases Drugs 0.000 description 1
- 229940021383 Antiinfective irrigating solutions Drugs 0.000 description 1
- 229960005475 Antiinfectives Drugs 0.000 description 1
- 241000271566 Aves Species 0.000 description 1
- 210000003719 B-Lymphocytes Anatomy 0.000 description 1
- 108010071919 Bispecific Antibodies Proteins 0.000 description 1
- 229940098773 Bovine Serum Albumin Drugs 0.000 description 1
- 108091003117 Bovine Serum Albumin Proteins 0.000 description 1
- 241000282832 Camelidae Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 208000002177 Cataract Diseases 0.000 description 1
- 241000700199 Cavia porcellus Species 0.000 description 1
- 210000003161 Choroid Anatomy 0.000 description 1
- 241000557626 Corvus corax Species 0.000 description 1
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 description 1
- 230000036947 Dissociation constant Effects 0.000 description 1
- 108010041308 Endothelial Growth Factors Proteins 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000283074 Equus asinus Species 0.000 description 1
- 229940109526 Ery Drugs 0.000 description 1
- 229940012356 Eye Drops Drugs 0.000 description 1
- 210000000744 Eyelids Anatomy 0.000 description 1
- 102100008658 FN1 Human genes 0.000 description 1
- 241000282326 Felis catus Species 0.000 description 1
- 108010067306 Fibronectins Proteins 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 229920000209 Hexadimethrine bromide Polymers 0.000 description 1
- 229940088597 Hormone Drugs 0.000 description 1
- 206010022489 Insulin resistance Diseases 0.000 description 1
- PMVSDNDAUGGCCE-TYYBGVCCSA-L Iron(II) fumarate Chemical compound [Fe+2].[O-]C(=O)\C=C\C([O-])=O PMVSDNDAUGGCCE-TYYBGVCCSA-L 0.000 description 1
- 241000229754 Iva xanthiifolia Species 0.000 description 1
- 201000006165 Kuhnt-Junius degeneration Diseases 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-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
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 229920002521 Macromolecule Polymers 0.000 description 1
- 208000008466 Metabolic Disease Diseases 0.000 description 1
- 241000699666 Mus <mouse, genus> Species 0.000 description 1
- 206010028980 Neoplasm Diseases 0.000 description 1
- 241001182492 Nes Species 0.000 description 1
- 206010052896 Ocular vascular disease Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 241000283898 Ovis Species 0.000 description 1
- 101710027269 PECAM1 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
- 101700079089 PTPRD Proteins 0.000 description 1
- 102100005502 PTPRD Human genes 0.000 description 1
- 210000000496 Pancreas Anatomy 0.000 description 1
- FAXGPCHRFPCXOO-LXTPJMTPSA-N Pepstatin Chemical compound OC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C)NC(=O)C[C@H](O)[C@H](CC(C)C)NC(=O)[C@H](C(C)C)NC(=O)[C@H](C(C)C)NC(=O)CC(C)C FAXGPCHRFPCXOO-LXTPJMTPSA-N 0.000 description 1
- 229950000964 Pepstatin Drugs 0.000 description 1
- 108010067902 Peptide Library Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- 229920003171 Poly (ethylene oxide) Polymers 0.000 description 1
- 239000004743 Polypropylene Substances 0.000 description 1
- 229920001451 Polypropylene glycol Polymers 0.000 description 1
- 239000004372 Polyvinyl alcohol Substances 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 229960003876 Ranibizumab Drugs 0.000 description 1
- 108010062724 Ranibizumab Proteins 0.000 description 1
- 206010038862 Retinal exudate Diseases 0.000 description 1
- 206010038901 Retinal vascular disease Diseases 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 229920002684 Sepharose Polymers 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 108010090091 TIE-2 Receptor Proteins 0.000 description 1
- 102000012753 TIE-2 Receptor Human genes 0.000 description 1
- 108060008443 TPPP Proteins 0.000 description 1
- 239000004473 Threonine Substances 0.000 description 1
- 229940104230 Thymidine Drugs 0.000 description 1
- 102000009843 Thyroglobulin Human genes 0.000 description 1
- 108010034949 Thyroglobulin Proteins 0.000 description 1
- 229960002175 Thyroglobulin Drugs 0.000 description 1
- 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 1
- 229940117013 Triethanolamine oleate Drugs 0.000 description 1
- 229920004890 Triton X-100 Polymers 0.000 description 1
- 210000003606 Umbilical Veins Anatomy 0.000 description 1
- 108091007928 VEGF receptors Proteins 0.000 description 1
- 102000009484 Vascular Endothelial Growth Factor Receptors Human genes 0.000 description 1
- 210000003462 Veins Anatomy 0.000 description 1
- 206010047571 Visual impairment Diseases 0.000 description 1
- LWZFANDGMFTDAV-BURFUSLBSA-N [(2R)-2-[(2R,3R,4S)-3,4-dihydroxyoxolan-2-yl]-2-hydroxyethyl] dodecanoate Chemical compound CCCCCCCCCCCC(=O)OC[C@@H](O)[C@H]1OC[C@H](O)[C@H]1O LWZFANDGMFTDAV-BURFUSLBSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K [O-]P([O-])([O-])=O Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 239000011149 active material Substances 0.000 description 1
- 230000001154 acute Effects 0.000 description 1
- 238000007792 addition Methods 0.000 description 1
- 239000000654 additive Substances 0.000 description 1
- 238000001261 affinity purification Methods 0.000 description 1
- 229960002833 aflibercept Drugs 0.000 description 1
- 108010081667 aflibercept Proteins 0.000 description 1
- 229940037003 alum Drugs 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 230000003042 antagnostic Effects 0.000 description 1
- 239000005557 antagonist Substances 0.000 description 1
- 230000001078 anti-cholinergic Effects 0.000 description 1
- 230000002924 anti-infective Effects 0.000 description 1
- 239000003472 antidiabetic agent Substances 0.000 description 1
- 230000000890 antigenic Effects 0.000 description 1
- 239000002220 antihypertensive agent Substances 0.000 description 1
- 239000003443 antiviral agent Substances 0.000 description 1
- 239000003855 balanced salt solution Substances 0.000 description 1
- 230000002146 bilateral Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000000740 bleeding Effects 0.000 description 1
- 231100000319 bleeding Toxicity 0.000 description 1
- 230000037396 body weight Effects 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 239000008366 buffered solution Substances 0.000 description 1
- 239000001506 calcium phosphate Substances 0.000 description 1
- 229910000389 calcium phosphate Inorganic materials 0.000 description 1
- 235000011010 calcium phosphates Nutrition 0.000 description 1
- 239000002327 cardiovascular agent Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 230000001413 cellular Effects 0.000 description 1
- 239000001913 cellulose Substances 0.000 description 1
- 229920002678 cellulose Polymers 0.000 description 1
- 239000003576 central nervous system agent Substances 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000000812 cholinergic antagonist Substances 0.000 description 1
- 238000004587 chromatography analysis Methods 0.000 description 1
- 230000001684 chronic Effects 0.000 description 1
- 230000015271 coagulation Effects 0.000 description 1
- 238000005345 coagulation Methods 0.000 description 1
- 238000005037 combinatorial chemistry Methods 0.000 description 1
- 150000001875 compounds Chemical class 0.000 description 1
- 239000003636 conditioned culture media Substances 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 229920001577 copolymer Polymers 0.000 description 1
- 230000001808 coupling Effects 0.000 description 1
- 238000010168 coupling process Methods 0.000 description 1
- 238000005859 coupling reaction Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 239000003405 delayed action preparation Substances 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 229920003013 deoxyribonucleic acid Polymers 0.000 description 1
- 238000006209 dephosphorylation reaction Methods 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 238000003745 diagnosis Methods 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 201000010870 diseases of metabolism Diseases 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 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 1
- 238000001962 electrophoresis Methods 0.000 description 1
- 238000002571 electroretinography Methods 0.000 description 1
- 238000010828 elution Methods 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000008393 encapsulating agent Substances 0.000 description 1
- 238000005538 encapsulation Methods 0.000 description 1
- 238000006911 enzymatic reaction Methods 0.000 description 1
- XNWFRZJHXBZDAG-UHFFFAOYSA-N ethylene glycol monomethyl ether Chemical compound COCCO XNWFRZJHXBZDAG-UHFFFAOYSA-N 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 239000003889 eye drop Substances 0.000 description 1
- 230000001605 fetal Effects 0.000 description 1
- 230000037320 fibronectin Effects 0.000 description 1
- 239000000945 filler Substances 0.000 description 1
- 239000010408 film Substances 0.000 description 1
- 102000034387 fluorescent proteins Human genes 0.000 description 1
- 108091006031 fluorescent proteins Proteins 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 description 1
- 239000003349 gelling agent Substances 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000011521 glass Substances 0.000 description 1
- LYCAIKOWRPUZTN-UHFFFAOYSA-N glycol Chemical compound OCCO LYCAIKOWRPUZTN-UHFFFAOYSA-N 0.000 description 1
- 239000003102 growth factor Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 229920001519 homopolymer Polymers 0.000 description 1
- 239000005556 hormone Substances 0.000 description 1
- 229920001600 hydrophobic polymer Polymers 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-M hydroxyl anion Chemical compound [OH-] XLYOFNOQVPJJNP-UHFFFAOYSA-M 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 238000003365 immunocytochemistry Methods 0.000 description 1
- 230000002163 immunogen Effects 0.000 description 1
- 230000016784 immunoglobulin production Effects 0.000 description 1
- 239000003018 immunosuppressive agent Substances 0.000 description 1
- 238000002513 implantation Methods 0.000 description 1
- 238000011065 in-situ storage Methods 0.000 description 1
- 238000009540 indirect ophthalmoscopy Methods 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 230000000366 juvenile Effects 0.000 description 1
- 238000002372 labelling Methods 0.000 description 1
- 108010052968 leupeptin Proteins 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 244000144972 livestock Species 0.000 description 1
- 231100000864 loss of vision Toxicity 0.000 description 1
- 210000004962 mammalian cells Anatomy 0.000 description 1
- 238000004949 mass spectrometry Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 229910052751 metal Inorganic materials 0.000 description 1
- 239000002184 metal Substances 0.000 description 1
- 239000003094 microcapsule Substances 0.000 description 1
- 230000000116 mitigating Effects 0.000 description 1
- 238000002156 mixing Methods 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000006011 modification reaction Methods 0.000 description 1
- 102000035365 modified proteins Human genes 0.000 description 1
- 108091005569 modified proteins Proteins 0.000 description 1
- 230000000051 modifying Effects 0.000 description 1
- 239000003068 molecular probe Substances 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 239000002674 ointment Substances 0.000 description 1
- 238000002515 oligonucleotide synthesis Methods 0.000 description 1
- LFGREXWGYUGZLY-UHFFFAOYSA-N oxophosphanyl Chemical group [P]=O LFGREXWGYUGZLY-UHFFFAOYSA-N 0.000 description 1
- 238000004806 packaging method and process Methods 0.000 description 1
- 230000001575 pathological Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 108010091212 pepstatin Proteins 0.000 description 1
- 239000000546 pharmaceutic aid Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 230000000275 pharmacokinetic Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 230000000649 photocoagulation Effects 0.000 description 1
- 230000003169 placental Effects 0.000 description 1
- 229920000740 poly(D-lysine) polymer Polymers 0.000 description 1
- 229920001583 poly(oxyethylated polyols) Polymers 0.000 description 1
- 229920001155 polypropylene Polymers 0.000 description 1
- 229920002451 polyvinyl alcohol Polymers 0.000 description 1
- 239000002244 precipitate Substances 0.000 description 1
- 230000001376 precipitating Effects 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- OZAIFHULBGXAKX-UHFFFAOYSA-N precursor Substances N#CC(C)(C)N=NC(C)(C)C#N OZAIFHULBGXAKX-UHFFFAOYSA-N 0.000 description 1
- 230000002335 preservative Effects 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 238000002203 pretreatment Methods 0.000 description 1
- 239000000047 product Substances 0.000 description 1
- 201000007914 proliferative diabetic retinopathy Diseases 0.000 description 1
- 210000001938 protoplasts Anatomy 0.000 description 1
- 230000005180 public health Effects 0.000 description 1
- 239000011541 reaction mixture Substances 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 230000002829 reduced Effects 0.000 description 1
- 230000004044 response Effects 0.000 description 1
- 201000011056 retinal disease Diseases 0.000 description 1
- 230000004233 retinal vasculature Effects 0.000 description 1
- 239000011435 rock Substances 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- 235000013024 sodium fluoride Nutrition 0.000 description 1
- PUZPDOWCWNUUKD-UHFFFAOYSA-M sodium fluoride Inorganic materials [F-].[Na+] PUZPDOWCWNUUKD-UHFFFAOYSA-M 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 229940035044 sorbitan monolaurate Drugs 0.000 description 1
- 230000000087 stabilizing Effects 0.000 description 1
- 239000008223 sterile water Substances 0.000 description 1
- 231100000803 sterility Toxicity 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 125000000472 sulfonyl group Chemical group *S(*)(=O)=O 0.000 description 1
- 239000003826 tablet Substances 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- 238000003325 tomography Methods 0.000 description 1
- 230000001052 transient Effects 0.000 description 1
- 229950004616 tribromoethanol Drugs 0.000 description 1
- 125000002306 tributylsilyl group Chemical group C(CCC)[Si](CCCC)(CCCC)* 0.000 description 1
- 238000004450 types of analysis Methods 0.000 description 1
- 230000000989 vascularization Effects 0.000 description 1
- 230000003612 virological Effects 0.000 description 1
- 229920003169 water-soluble polymer Polymers 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/54—Medicinal preparations containing antigens or antibodies characterised by the route of administration
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P27/00—Drugs for disorders of the senses
- A61P27/02—Ophthalmic agents
- A61P27/06—Antiglaucoma agents or miotics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/04—Antibacterial agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/10—Antioedematous agents; Diuretics
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/40—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/75—Agonist effect on antigen
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
Abstract
Disclosed is a use of an ?????-ECD binding agent or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the alleviation of an ocular edema.
Description
TREATMENT OF OCULAR DISEASE
CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority US. Provisional ation Serial No. 61/546,708
filed October 13, 2011. The entire content of US. Provisional Application Serial No. 61/546,708
is incorporated herein by reference.
INCORPORATION BY REFERENCE OF MATERIAL SUBMITTED
ELECTRONICALLY
Incorporated by reference in its entirety is a computer-readable sequence listing submitted
concurrently herewith and identified as follows: One 92 KB ASCII (Text) file named “233106-
33l562_Seq_Listing_ST25.txt,” created on October 12, 2012, at 12:42 pm.
FIELD
Methods for treating eye diseases or conditions characterized by vascular ility,
vascular leakage, and neovacularization such as ocular edema, ocular cularization,
diabetic macular edema, age-related macular degeneration, choroidal neovascularization, diabetic
retinopathy, retinal vein occlusion (central or branch), ocular ia, ocular trauma, surgery
d edema, and uveitis.
BACKGROUND
The eye ses several structurally and functionally ct vascular beds, which
supply ocular components critical to the maintenance of vision. These include the l and
choroidal atures, which supply the inner and outer portions of the retina, respectively, and
the limbal vasculature located at the periphery of the cornea. Injuries and diseases that impair the
normal structure or function of these ar beds are among the leading causes of visual
impairment and blindness. For example, diabetic retinopathy is the most common e
affecting the retinal vasculature, and is the leading cause of vision loss among the working age
population in the United States. Vascularization of the cornea secondary to injury or disease is
yet another category of ocular vascular disease that can lead to severe impairment of vision.
“Macular degeneration” is a general medical term that s to any of several
disease syndromes, which e a gradual loss or impairment of eyesight due to cell and tissue
degeneration of the yellow macular region in the center of the retina. Macular degeneration is
often characterized as one of two types, non-exudative (dry form) or exudative (wet form).
Although both types are bilateral and progressive, each type may reflect ent pathological
processes. The wet form of age-related r degeneration (AMD) is the most common form
of choroidal neovascularization and a leading cause of blindness in the y. AMD affects
millions of Americans over the age of 60, and is the leading cause of new ess among the
elderly.
Choroidal neovascular membrane (CNVM) is a problem that is related to a wide
variety of retinal diseases, but is most commonly linked to lated macular ration.
With CNVM, abnormal blood vessels stemming from the choroid (the blood vessel-rich tissue
layer just beneath the ) grow up through the retinal layers. These new vessels are very
fragile and break easily, causing blood and fluid to pool within the layers of the retina.
Diabetes tes mellitus) is a metabolic disease caused by the inability of the
pancreas to produce insulin or to use the insulin that is produced. The most common types of
diabetes are type 1 diabetes (often referred to as Juvenile Onset Diabetes Mellitus) and type 2
diabetes (often referred to as Adult Onset Diabetes Mellitus). Type 1 diabetes results from the
body's failure to produce insulin due to loss of n producing cells, and presently requires the
person to inject insulin. Type 2 diabetes generally results from insulin resistance, a condition in
which cells fail to use insulin properly. Type 2 diabetes may have a component of insulin
deficiency as well.
Diabetes is directly responsible for a large number of disease conditions, including
conditions or es of the eye including diabetic retinopathy (DR) and diabetic macular edema
(DME) which are leading causes of vision loss and blindness in most developed ies. The
increasing number of individuals with diabetes worldwide suggests that DR and DME will
continue to be major contributors to vision loss and associated functional ment for years to
COl’IlC.
Diabetic retinopathy is a complication of diabetes that results from damage to the
blood vessels of the light-sensitive tissue at the back of the eye (retina). At first, diabetic
retinopathy may cause no ms or only mild vision problems. Eventually, however,
diabetic retinopathy can result in ess. Diabetic retinopathy can develop in anyone who has
type 1 diabetes or type 2 diabetes.
At its earliest stage, non-proliferative retinopathy, microaneurysms occur in the
’s tiny blood vessels. As the disease progresses, more of these blood vessels become
damaged or d and these areas of the retina send signals into the al tissue to grow
new blood vessels for hment. This stage is called proliferative retinopathy. The new blood
vessels grow along the retina and along the surface of the clear, vitreous gel that fills the inside of
the eye.
By themselves, these blood vessels do not cause symptoms or vision loss. However,
they have thin, fragile walls and without timely treatment, these new blood vessels can leak
blood (whole blood or a tuent thereof) which can result in severe vision loss and even
blindness.
Also, fluid can leak into the center of the macula, the part of the eye where sharp,
ht-ahead vision occurs. The fluid and the associated protein begin to deposit on or under the
macula causing the patient’s central vision to become distorted. This condition is called macular
edema. It can occur at any stage of diabetic retinopathy, although it is more likely to occur as the
disease progresses. About half of the people with proliferative retinopathy also have macular
edema.
Uveitis is a condition in which the uvea becomes inflamed. The eye is shaped much
like a tennis ball, hollow on the inside with three different layers of tissue surrounding a central
cavity. The outermost is the sclera (white coat of the eye) and the innermost is the retina. The
middle layer between the sclera and the retina is called the uvea. The uvea contains many of the
blood vessels that nourish the eye. Complications of uveitis include glaucoma, cataracts or new
blood vessel formation (neovascularization).
The currently available interventions for exudative (wet form) macular degeneration,
diabetic retinopathy, diabetic macular edema, choroidal neovascular membrane, cations
from uveitis or ocular trauma, include laser photocoagulation therapy, low dose radiation
(teletherapy) and surgical removal of neovascular membranes (vitrectomy). Laser therapy has
had limited success and selected choroidal neovascular nes which initially respond to
laser therapy have high disease ence rates. There is also a potential loss of vision resulting
from laser therapy. Low dose radiation has been applied ineffectively to induce regression of
choroidal neovascularization. Recently, vascular endothelial growth factor (VEGF) antagonists,
ranibizumab and aflibercept, have been approved for use in age-related macular degeneration,
diabetic macular edema and retinal vein occlusion (RVO).
(RVO) is the most common retinal vascular disease after diabetic retinopathy.
ing on the area of retinal venous drainage effectively occluded, it is broadly classified as
either central retinal vein occlusion (CRVO), hemispheric retinal vein occlusion (HRVO), or
branch l vein occlusion (BRVO). It has been observed that each of these has two subtypes.
Presentation of RVO in general is with variable ss visual loss with any combination of
fundal findings consisting of retinal vascular sity, retinal hemorrhages (blot and flame
shaped), cotton wool spots, optic disc swelling and macular edema. In a CRVO, retinal
hemorrhages will be found in all four quadrants of the fundus, while these are restricted to either
the superior or inferior fundal hemisphere in a HRVO. In a BRVO, hemorrhages are largely
localized to the area d by the occluded in the retinal vein.
There is therefore a long felt and substantial need for methods of treating diseases of
the eye which are characterized by ar instability, vascular leakage and neovascularization.
SUMMARY
Disclosed are agents that bind to the extracellular portion and inhibit human protein
ne phosphatase beta (HPTPB). Also disclosed are methods for treating eye diseases or
conditions characterized by ar instability, ar leakage, and neovacularization such as
ocular edema, ocular neovascularization, diabetic macular edema, age-related macular
degeneration, choroidal neovascularization, diabetic retinopathy, retinal vein occlusion (central
or branch), ocular ia, ocular trauma, surgery induced edema, and uveitis.
BRIEF DESCRIPTION OF THE FIGURES
Fig. 1. The onal antibody R15E6 recognizes Endogenous HPTPB on
elial cells. (Panel A) Endothelial cell lysates are immunoprecipitated with a control
antibody (Lane 1), with R15E6 (Lane 2) or with a mixture of anti-Tie2 and anti-VEGFR2
antibodies (Lane 3). precipitates are resolved by SDS-PAGE, transferred to a PVDF
membrane and probed by western blot with a mixture of R15E6, anti-Tie2 and anti-VEGFR2
antibodies. A single major high molecular weight band tent with HPTPB is seen with
R15E6 (Lane 2) and not with the control antibody (Lane 1) or the mixture of anti-Tie2 and anti-
VEGFR2 (Lane 3). (Panel B) Endothelial cells are subjected to FACS analysis with R15E6
(white peak) or a control with no primary antibody (black peak). The robust shift in fluorescence
indicates that R15E6 binds to HPTPB on the surface of intact endothelial cells.
Fig. 2. The monoclonal antibody R15E6 enhances Tie2 Receptor Activation in
HUVECs. Tie2 activation is measured in human elial cells as described in Example 4.
R15E6 dose dependently enhances both basal and Angl-induced Tie2 activation.
Fig. 3. Is a graphical representation of the mean area of choroidal neovascularization
in C57BL/6 mice 14 days post laser injury in eyes treated with intravitreal injection of 1 ug or
2pg of an anti-VE-PTP extracellular domain antibody in one eye versus similar treatment of the
fellow eye with control.
Fig. 4. Shows the mean area (mmz) of retinal neovascularization in C57BL/6 mice on
day P17 after containment in a 75% oxygen atmosphere from P5 to P12 and intravitreal injection
of an anti-VE-PTP extracellular domain antibody at P12 when the mice were returned to room
air.
Fig. 5. Shows representative fluorescent micrographs of mouse retinas in the -
induced pathy model after intravitreal injection of vehicle or 2 ug of an anti-VE-PTP
ellular domain antibody.
Fig. 6. Shows the mean area (mmz) of retinal neovascularization in C57BL/6 mice
on day P17 after containment in a 75% oxygen atmosphere from P5 to P12 followed by return to
room air on P12 with aneous administration of 1 mg/kg of an E-PTP ellular
domain antibody on days P12, 14 and 16.
Fig. 7. Shows the mean area (mmz) of retinal neovascularization in C57BL/6 mice on
day P17 after containment in a 75% oxygen atmosphere from P5 to P12 followed by return to
room air on P12 with subcutaneous administration of 2 mg/kg of an anti-VE-PTP extracellular
domain antibody on days P12, 14 and 16.
DETAILED DESCRIPTION
General Definitions
In this specification and in the claims that , reference will be made to a number
of terms, which shall be d to have the following meanings:
The term -ECD binding agent" and “specific binding agent” are used
interchangeably herein and refer to a molecule that specifically binds to the extracellular n
of HPTPB, and variants and derivatives thereof, as defined herein, that inhibits the Tie2
dephosphorylase activity of HPTPB.
“Agent” as used herein refers to a “HPTPB binding agent” unless otherwise noted.
fically binds HPTPB-ECD” refers to the ability of a specific g agent of
the present invention to recognize and bind to an epitope of the extracellular domain of HPTPB
with higher affinity than to other related and/or ted molecules. Specific binding agents
preferentially bind to HPTPB in a complex e of proteins and/or macromolecules. The
specific binding agent is preferably selective for HPTPB. “Selective” means that the agent has
significantly greater activity toward HPTPB compared with other related and/or unrelated
molecules, not that it is completely inactive with regard to other molecules. For example, a
selective agent may show 10-fold, lOO-fold, or lOOO-fold selectivity toward HPTPB than to other
related or unrelated molecules.
The term “anti-HPTPB-ECD antibodies” refers to antibodies or antibody fragments
that bind to the extracellular domain of HPTPB. Anti-HPTPB-ECD antibodies are a type of
HPTPB-ECD binding agent as defined herein.
The term “VE-PTP” refers to the mouse og of HPTPB.
All percentages, ratios and proportions herein are by weight, unless otherwise
ied. All temperatures are in degrees s (0C) unless otherwise specified.
Ranges may be expressed herein as from one particular value to another particular
value, the endpoints are included in the range. For example for the range from “lmg to 50mg”
includes the specific values lmg and 50mg. The antecedent “about” indicates that the values are
2012/060263
approximate. For example for the range from “about lmg to about 50mg” indicates that the
values are approximate values. Additionally, when such a range is expressed, the range includes
the range “from lmg to 50mg.” It will be further understood that the endpoints of each of the
ranges are significant both in relation to the other nt, and independently of the other
endpoint. For example the range “from lmg to 50mg”, includes the range “from 30mg to 40mg.”
tive ” means an amount of an active agent or combination of agents
effective to ameliorate or prevent the symptoms, or prolong the al of the patient being
treated. An effective amount may vary according to s known in the art, such as the disease
state, age, sex and weight of the human or animal being treated. Although particular dosage
regimes may be described in examples herein, a person skilled in the art would appreciate that
the dosage regime may be altered to provide optimum therapeutic response. For example,
several d doses may be administered daily or the dose may be proportionally d as
indicated by the exigencies of the therapeutic situation. In addition, the compositions of this
disclosure can be administered as frequently as necessary to e a therapeutic amount.
Determination of a therapeutically effective amount is well within the capabilities of those skilled
in the art, especially in light of the detailed disclosure provided herein.
As used herein the term “inhibit” or “inhibiting” refers to a statistically significant
and measurable reduction in activity, preferably a reduction of at least about 10% versus control,
more preferably a ion of about 50% or more, still more ably a reduction of about
80% or more.
As used herein the term “increase” or “increasing” refers to a statistically significant
and measurable increase in activity, preferably an increase of at least about 10% versus control,
more preferably an increase of about 50% or more, still more preferably an increase of about
80% or more.
“HPTP beta” or “HPTPB” are used interchangeably herein and are abbreviations for
human protein tyrosine phosphatase beta.
As used herein, “subject” means an individual. Thus, the “subject” can include
domesticated animals (e. g., cats, dogs, etc.), livestock (e. g., , horses, pigs, sheep, goats,
etc.), laboratory animals (e. g., mouse, rabbit, rat, guinea pig, etc.) and birds. “Subject” can also
e a mammal, such as a primate or a human. “Subject” and “patient” are used
interchangeably herein. Preferably the subject is a human.
By “reduce” or other forms of the word, such as “reducing” or “reduction,” is meant
lowering of an event or characteristic (e.g., ar leakage). It is understood that this is
typically in relation to some standard or expected value, in other words it is relative, but that it is
not always necessary for the standard or relative value to be referred to.
The terms “treatment”, “treating”, “treat” and the like, refer to obtaining a desired
pharmacologic and/or physiologic effect such as mitigating a e or a er in a host
and/or ng, inhibiting, or eliminating a ular characteristic or event associated with a
disorder (e. g., ocular edema). Thus, the term "treatment" es, preventing a disorder from
occurring in a host, particularly when the host is predisposed to acquiring the disease, but has not
yet been diagnosed with the disease; inhibiting the disorder; and/or alleviating or reversing the
disorder. Insofar as the methods of the present invention are directed to preventing disorders, it
is understood that the term "prevent" does not require that the e state be completely
thwarted. Rather, as used herein, the term preventing refers to the ability of the skilled artisan to
identify a tion that is susceptible to disorders, such that administration of the compounds
of the present invention may occur prior to onset of a disease. The term does not imply that the
disease state is completely avoided.
Unless otherwise ied, diabetic retinopathy includes all stages of
non-proliferative retinopathy and proliferative retinopathy.
Throughout the description and claims of this specification the word “comprise” and
other forms of the word, such as “comprising” and ises,” means including but not limited
to, and is not intended to exclude, for example, other additives, components, integers, or steps.
As used in the description and the appended claims, the singular forms CC 77 CC
a an” and
“the” include plural referents unless the context clearly dictates ise. Thus, for example,
reference to “a composition” includes one ition or mixtures of two or more such
compositions.
Optional” or “optionally” means that the subsequently described event or
circumstance can or cannot occur, and that the description includes instances where the event or
circumstance occurs and instances where it does not.
“Specifically binds HPTPB” refers to the ability of an agent of the present invention to
recognize and bind to an epitope of the extracellular domain of HPTPB with higher affinity than
to the other related and/or ted molecules. The agent is preferably selective for HPTPB.
“Specific” means that the agent has icantly r activity toward HPTPB compared with
other related and/or unrelated molecules, not that it is completely inactive with regard to other
molecules. For example, a selective agent may show 10-fold, lOO-fold, or lOOO-fold selectivity
toward HPTPB than to other related or unrelated molecule.
The term “epitope” refers to any portion of any molecule capable of being recognized
by and bound by a agent at one or more of the agent’s antigen binding regions. Epitopes usually
consist of distinct, recognizable surface groupings such as amino acids, sugars, lipids,
phosphoryl, or sulfonyl, and, in certain ments, may have ic three dimensional
structural characteristics, and/or specific charge characteristics. Epitopes as used herein may be
conformational or linear.
“Peptibody” is a molecule comprising an dy Fc domain attached to at least one
peptide. The production of peptibodies is generally bed in /24782.
“Fragment” refers to a portion of an agent. A fragment may retain the desired
biological activity of the agent and may be considered to be an agent itself. For example a
truncated protein in which the amino terminus and/or carboxy terminus and/or an internal amino
acid residue is d is a fragment of the protein and an Fab of an immunoglobulin molecule is
a fragment of the immunoglobulin. Such fragments may also be connected to a another molecule
by way of a direct connection (e. g. a peptide or disulfide bond) or by way of a linker.
“Protein” is used herein interchangeably with peptide and polypeptide.
es of the present ion include, but are not limited to amino acid sequences
having from about 3 to about 75 amino acids, or from about 5 to about 50 amino acids, or from
about 10 to about 25 amino acids. Peptides may be naturally occurring or cial amino acid
sequences .
A protein of the invention may be obtained by methods well known in the art, for
example, using rd direct peptide synthesizing techniques such as via solid-phase synthesis.
If the gene sequence is known or can be d then the protein may be produced by standard
recombinant methods. The proteins may be isolated or purified in a variety of ways known to
one skilled in the art. Standard cation methods include itation with salts,
electrophoretic, chromatographic techniques and the like.
Agents may be covalently or non-covalently conjugated to a vehicle. The term
“vehicle” refers to a molecule that prevents degradation and/or increase half-life, reduces
toxicity, reduces genicity, or ses biological activity of the agent. Exemplary
vehicles include, but are not limited, Fc domains of immunoglobulins and rs, for example:
polyethylene glycol (PEG), sine, dextran, a lipid, a cholesterol group (such as a steroid); a
carbohydrate or oligosaccharide; or any natural or synthetic protein, or e that binds to a
salvage receptor.
“Derivatives” e those binding agents that have been chemically modified in
some manner distinct from insertion, deletion, or substitution variants. For example, wherein the
binding agent is a protein, the carboxyl terminus may be capped with an amino group, such as
NHZ.
In some embodiments one or more molecules are linked together to form the agent.
For example antibody fragments may be connected by a linker. In general, the chemical
structure of the linker is not critical as it serves primarily as a space. In one embodiment, the
linker is made of amino acids linked together by way of peptide bonds. In another ment,
the linker is a non-peptide linker such as a non-sterically hindering C1-C6 alkyl group. In another
embodiment, the linker is a PEG linker. It will further be appreciated that the linker can be
inserted in a number of locations on the molecule.
Variants of an agent are included within the scope of the present invention. “Variant”
or “Variants” as used herein means an agent having a protein or nucleotide sequence which is
substantially similar to the protein or nucleotide sequence of the non-variant agent and which
shares a similar activity of the riant agent. A n or nucleotide sequence may be
d in various ways to yield a variant assed by the present invention, including
substitutions, deletions, truncations, insertions and other modifications. Methods for such
manipulations are well known in the art. See, for e, Current Protocols in Molecular
Biology (and updates) Ausubel et al., Eds (1996), John Wiley and Sons, New York: Methods in
Molecular Biology, Vol. 182, In vitro Mutagenesis Protocols, 2ndl Edition, Barman Ed. (2002),
Humana Press, and the references cited therein. For example, variants include peptides and
ptides wherein amino acid residues are inserted into, deleted from and/or substituted into
the known amino acid sequence for the binding agent. In one embodiment, the substitution of the
amino acid is conservative in that it minimally alters the biochemical properties of the variant. In
other embodiments, the variant may be an active fragment of a full-length protein, a chemically
modified protein, a protein modified by addition of affinity or epitope tags, or fluorescent or
other labeling moieties, whether accomplished by in vivo or in vitro enzymatic treatment of the
protein, by chemical cation, or by the synthesis of the n using modified amino acids.
Fusions proteins are also plated herein. Using known methods, one of skill in
the art would be able to make fusion proteins of the proteins of the invention; that, while ent
from native form, may be useful. For example, the fusion partner may be a signal (or leader)
polypeptide sequence that co-translationally or post-translationally directs er of the protein
from its site of synthesis to another site (e. g., the yeast alpha-factor leader). Alternatively, it may
be added to facilitate purification or identification of the protein of the invention (e. g., poly-His,
Flag peptide, or fluorescent proteins).
Standard ques may be used for recombinant DNA, oligonucleotide synthesis,
and tissue culture and transformation (e. g., electroporation, lipofection). Enzymatic reactions
and purification techniques may be performed according to manufacturer's specifications or as
commonly accomplished in the art or as described herein. The techniques and procedures are
generally performed according to conventional methods known in the art and as described in
various general and more ic nces that are cited and discussed hout the present
specification. Unless specific tions are ed, the nomenclature utilized in connection
with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic
chemistry, and medicinal and pharmaceutical chemistry described herein are those known and
commonly used in the art. Standard techniques may be used for chemical syntheses, chemical
analyses, pharmaceutical preparation, formulation, delivery and treatment of patients.
Sequence Listing
Table l.
SEQ ID NO:1 Full length Human HPTPB tide sequence (X54131)
SEQ ID NO:2 Full length Human HPTPB amino acid ce (P23467)
SEQ ID NO:3 Extracellular Portion of Human HPTPB with (His)6Gly Tag
SEQ ID NO:4 Extracellular Portion of Human HPTPB
SEQ ID NO:5 Full length mouse VE-PTP nucleotide sequence
SEQ ID NO:6 Full length mouse VE-PTP amino acid ce
SEQ ID NO:7 Extracellular portion of mouse VE-PTP amino acid sequence
HPTPB-ECD binding agents
Agents useful in the present ion include, but are not d to, antibodies,
proteins, darpins, peptides, aptamers, adnectins, odies, or nucleic acids that bind
specifically to the extracellular portion of HPTPB and inhibit at least one phosphatase activity of
HPTPB. As used herein, “phosphatase activity” includes enzymatic activity and ic activity
where biological activity is measured by assessing Tie2 phosphorylation.
Agents useful in the present invention further include: antibodies, or antigen binding
fragments thereof which bind to the extracellular portion of HPTPB wherein the antibody or
n-binding fragment inhibits at least one phosphatase activity of HPTPB. These agents
e monoclonal and polyclonal antibodies. An agent may be a fragment of an antibody,
wherein the fragment comprises the heavy and light chain variable regions, or the fragment is an
F(ab’)2, or the fragment is a dimer or trimer of an Fab, Fv, scFv, or a dia-, tria-, or tetrabody
d from the antibody.
For example, the agent may be, without limitation, an antibody or dy fragment
that binds the extracellular portion of HPTPB; or in particular an antibody that binds an FN3
repeat of HPTPB, or more specifically an antibody that binds the first FN3 repeat of HPTPB.
Agents r include: the monoclonal dy R15E6 which is described in US.
patent number 7,973,142, which is hereby incorporated in its entirety. (The mouse hybridoma,
Balbc spleen cells (B cells) which may be used to produce the antibody are deposited with
American Type Culture Collection (ATCC), PO. Box 1549, Manassas, Va. 20108 USA on 4
May 2006, assigned ATCC No. PTA-7580) (Referred to herein as R15E6)), antibodies having
the same or substantially the same biological characteristics of R15E6; antibody fragments of
R15E6, wherein the fragment comprises the heavy and light chain variable regions; an 2 of
R15E6; dimers or trimers of an Fab, Fv, scFv; and dia-, tria-, or tetrabodies derived from R15E6.
In particular, an agent suitable for use in the present invention is an antibody,
antibody fragment, variant or tives thereof, either alone or in combination with other
amino acid sequences, ed by known techniques. Such ques include, but are not
limited to enzymatic cleavage, chemical cleavage, peptide sis or recombinant
techniques. The invention r es derivative agents, e.g. odies.
Thus, one embodiment of an ΗΡΤΡβ-ECD binding agent is an antibody,
another embodiment is a protein, yet another embodiment is a peptide, and r
embodiment is a darpin, another embodiment is an aptamer, r embodiment is a
peptibody, still another embodiment is an adnectin, another embodiment is a nucleic acid. In
some embodiments the ΗΡΤΡβ-ECD binding agent is an monoclonal dy, or is a
polyclonal antibody. In particular embodiments, the ΗΡΤΡβ-ECD binding agent is an
antibody nt that is capable of binding to ΗΡΤΡβ-ECD. Preferably the ΗΡΤΡβ-ECD
binding agent is an antibody, or an antibody fragment, including but not d to, an F(ab')2,
an Fab, a dimer of an Fab, an Fv, a dimer of an Fv, a scFv, a dimer of a scFv, a dimer an Fab,
an Fv, a dimer of an Fv, a scFv, a dimer of a scFv, a trimer of an Fab, a trimer of an Fv, a
trimer of a scFv, minibodies, a diabody , a triabody, a tetrabody, a linear antibody, a protein,
a peptide, an aptamer, a peptibody, an adnectin, or a nucleic acid, that binds to the
extracellular portion of ΗΡΤΡβ. In certain ments the ΗΡΤΡβ-ECD binding agent is an
F(ab')2 of a monoclonal antibody. In some embodiments the ΗΡΤΡβ-ECD binding agent
ses a plurality of ΗΡΤΡβ-ECD binding sites, for example where the ΗΡΤΡβ-ECD
binding agent is an intact antibody or an F(ab')2, or a dimer of an Fab, or a trimer of an Fab.
For example, in some embodiments an ΗΡΤΡβ-ECD binding agent is able to bind to two
ΗΡΤΡβ molecules simultaneously at the same or different epitope, thereby bringing the two
ΗΡΤΡβ molecules into close proximity with one and other. In other embodiments the ΗΡΤΡβ-
ECD binding agent is able to bind to three ΗΡΤΡβ molecules simultaneously at the same or
different epitope, thereby bringing the three ΗΡΤΡβ molecules into close proximity with one
and other. In another embodiment, the ΗΡΤΡβ-ECD binding agent is the monoclonal
antibody produced by hybridoma cell line ATCC No. PTA-7580. In yet another embodiment,
the ΗΡΤΡβ-ECD binding agent is an antigen binding fragment of the monoclonal antibody
produced by hybridoma cell line ATCC No. PTA-7580. In still r embodiment, the
ΗΡΤΡβ-ECD binding agent is an antibody having the same or ntially the same
biological characteristics the monoclonal antibody produced by oma cell line ATCC
No. PTA-7580 or an antigen binding fragment thereof.
2012/060263
Any of the embodiments of HPTPB-ECD binding agents disclosed in the present
application, may be covalently or valently conjugated to a vehicle. The term “vehicle”
refers to a molecule that affects a biological property of an agent. For example, a vehicle may
prevent degradation, and/or se half-life, absorption, reduce toxicity, reduce
immunogenicity, or increase biological activity of the agent. Exemplary vehicles include, but are
not limited to, EC domains of immunoglobulins; polymers, for example: polyethylene glycol
(PEG), polylysine, dextran; ; cholesterol groups (such as a steroid); carbohydrates,
dendrimers, oligosaccharides, or peptides that binds to a salvage receptor. In some embodiments
the vehicle is polyethylene glycol (PEG), in other embodiments the vehicle is polylysine, in yet
other ments the vehicle is dextran, in still other embodiments the vehicle is a lipid
Water e polymer attachments, such as polyethylene glycol, polyoxyethylene
glycol, or polypropylene , as described U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144;
4,670,417; 4,791,192; and 4,179,337, which are incorporated herein in their ty. Still other
useful polymers known in the art e monomethoxy-polyethylene glycol, dextran, cellulose,
or other ydrate based polymers, poly-(N-vinyl pyrrolidone)-polyethylene glycol,
propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer,
polyoxyethylated polyols (e. g., glycerol) and polyvinyl alcohol, as well as es of these
polymers. Particularly preferred are peptibodies covalently modified with polyethylene glycol
(PEG) subunits. Water soluble polymers may be bonded at specific positions, for example at the
amino terminus of the peptibodies, or randomly attached to one or more side chains of the
polypeptide. The use of PEG for improving the therapeutic capacity for , e.g. peptibodies,
and for humanized antibodies in particular, is bed in U.S. Pat. No. 6,133,426. The
invention also contemplates derivatizing the peptide and/or vehicle n of the agents. Such
derivatives may improve the solubility, absorption, ical half-life, and the like of the agents.
The moieties may atively eliminate or attenuate any undesirable side-effect of the agents
and the like.
The term "antibody" (Ab) as used herein includes monoclonal antibodies, polyclonal
antibodies, multi-specific antibodies (e. g. bispecific antibodies), single chain antibodies, e. g.,
antibodies from llama and camel, antibody fragments, e. g., variable regions and/or constant
region fragments, so long as they exhibit a desired biological activity, e. g., antigen-binding
activity. The term "immunoglobulin" (Ig) is used interchangeably with "antibody" herein.
An “antigen binding fragment” as used herein is a fragment of an agent that binds to a
portion of HPTPB and inhibits the activity of HPTPB.
An "isolated antibody" is an antibody which has been identified, and/or separated,
and/or recovered from its natural environment.
The basic four-chain antibody unit is a heterotetrameric glycoprotein composed of
two identical light (L) chains and two identical heavy (H) chains (an IgM antibody consists of 5
of the basic heterotetramer units along with an additional polypeptide called J chain, and
therefore contain 10 antigen g sites, while secreted IgA antibodies may polymerize to form
polyvalent assemblages comprising 2-5 of the basic 4-chain units along with J chain). In the case
of IgGs, the four-chain unit is generally about 150 kilo s (kDa). Each L chain is linked to
an H chain by one covalent ide bond, while the two H chains are linked to each other by
one or more disulfide bonds depending on the H chain isotype. Each H and L chain also has
regularly spaced intrachain disulfide bridges. Each H chain has at the N-terminus, a variable
domain (VH) followed by three constant domains (CH) for each of the alpha and gamma chains
and four CH domains for mu and n isotypes. Each L chain has at the inus, a variable
domain (VL) followed by a constant domain (CL) at its other end. The VL is aligned with the VH
and the CL is d with the first constant domain of the heavy chain (CH1). Particular amino
acid residues are believed to form an interface between the light chain and heavy chain variable
domains. The pairing of a VH and VL together forms a single antigen-binding site. For the
ure and properties of the different classes of antibodies, see, e. g., Basic and Clinical
Immunology, 8th edition, Daniel P. Stites, Abba I. Terr and Tristram G. Parslow (eds.), on
& Lange, 1994, page 71 and Chapter 6.
The L chain from any rate species may be ed to one of two clearly
distinct types, called kappa and lambda, based on the amino acid sequences of their constant
domains. Depending on the amino acid sequence of the constant domain of their heavy chains
(CH), immunoglobulins may be assigned to ent classes or isotypes. There are five classes of
immunoglobulins: IgA, IgD, IgE, IgG, and IgM, having heavy chains designated alpha, delta,
epsilon, gamma and mu, respectively. The gamma and alpha classes are further divided into
subclasses on the basis of relatively minor differences in CH sequence and function, e.g., humans
express the ing subclasses: IgGl, IgG2, IgG3, IgG4, IgAl and IgA2.
Members of the Camelidae family, e. g., llama, camel and aries, contain a
unique type of antibody, that are devoid of light chains, and further lack the CH1 domain
(Muyldermans, 8., Rev. Mol. Biotechnol., Vol. 74, pp. 277-302 (2001)). The variable region of
these heavy chain antibodies are termed VHH or VHH, and constitute the st available intact
antigen-binding fragment (15 kDa) derived from a functional immunoglobulin.
The term "variable" refers to the fact that certain segments of the variable domains
differ extensively in sequence among antibodies. The V domain mediates antigen binding and
defines specificity of a particular antibody for its n. However, the variability is not evenly
distributed across the llO-amino acid span of the variable domains. Instead, the V regions consist
of vely invariant stretches called framework regions (FR) of 15-30 amino acids separated by
shorter regions of extreme variability called variable regions" that are each 9-12 amino
acids long. The variable domains of native heavy and light chains each comprise four FRs,
largely adopting a t configuration, connected by three hypervariable regions, which form
loops connecting, and in some cases forming part of, the B-sheet structure. The hypervariable
regions in each chain are held together in close proximity by the FRs and, with the hypervariable
regions from the other chain, contribute to the ion of the antigen-binding site of antibodies.
The constant domains are not involved directly in binding an antibody to an antigen, but exhibit
various effector functions, such as ipation of the antibody in antibody dependent cellular
cytotoxicity (ADCC).
The term "hypervariable region" when used herein refers to the amino acid residues of
an antibody which are sible for antigen-binding. The hypervariable region generally
comprises amino acid residues from a ementarity determining region" or "CDR" (e. g.,
around about residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the VL, and around about 1-35
(H1), 50-65 (H2) and 95-102 (H3) in the VH; Kabat et al., ces of Proteins of
Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda,
Md. (1991)) and/or those residues from a "hypervariable loop".
The term "monoclonal antibody" as used herein refers to an antibody obtained from a
population of substantially homogeneous antibodies, i.e., the individual antibodies comprising
the tion are cal except for possible naturally occurring mutations that may be present
in minor amounts. In st to polyclonal antibody preparations which include different
antibodies directed against different epitopes, each monoclonal antibody is ed against a
single epitope, i.e., a single antigenic determinant. In addition to their specificity, the monoclonal
antibodies are advantageous in that they may be synthesized uncontaminated by other antibodies.
The modifier "monoclonal" is not to be construed as requiring production of the antibody by any
particular method. For example, the onal antibodies useful in the present invention may
be prepared by the hybridoma methodology or may be made using recombinant DNA methods in
bacterial, eukaryotic animal or plant cells (see, e.g., U.S. Pat. No. 4,816,567). The "monoclonal
antibodies" may also be isolated from phage antibody libraries, using the available techniques,
e.g., on et al., Nature, Vol. 352, pp. 624-628 (1991).
The monoclonal antibodies herein include "chimeric" antibodies in which a portion of
the heavy and/or light chain is identical with or homologous to corresponding sequences in
antibodies derived from a particular species or ing to a particular antibody class or
subclass, while the remainder of the chain(s) is identical with or homologous to corresponding
sequences in antibodies derived from another species or belonging to another antibody class or
ss, as well as nts of such antibodies, so long as they exhibit the d biological
activity (see U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, Vol. 81,
pp. 6851-6855 (1984)).
An ody fragment" comprises a portion of a multimeric antibody, ably the
antigen binding or variable region of the intact antibody. Examples of antibody fragments include
Fab, Fab‘, F(ab')2, dimers and trimers of Fab conjugates, Fv, scFv, minibodies; dia-, tria- and
odies; linear antibodies (See Hudson et al., Nature Med. Vol. 9, pp. 4 (2003)).
"Fv" is the minimum antibody fragment which contains a complete n binding
site. This fragment consists of a dimer of one heavy- and one light-chain variable region domain
in tight, non-covalent association. From the folding of these two domains e six
hypervariable loops (3 loops each from the H and L chain) that contribute the amino acid
residues for antigen binding and confer antigen binding icity to the antibody. However,
even a single variable domain (or half of an Fv comprising only three CDRs specific for an
antigen) has the ability to recognize and bind antigen, and are therefore included in the definition
of Fv.
A single-chain le fragment (scFv) is a fusion protein of the variable regions of
the heavy (VH) and light chains (VL) of immunoglobulins, connected with a short linker e
of ten to about 25 amino acids. The linker is usually rich in glycine for flexibility, as well as
serine or threonine for solubility, and can either t the N-terminus of the VH with the C-
terminus of the VL, or vice versa. This protein retains the specificity of the original
immunoglobulin, despite removal of the constant regions and the introduction of the linker.
Divalent (or bivalent) single-chain variable fragments (di-scFvs, bi-scFvs) can be
engineered by linking two scFvs. This can be done by producing a single e chain with two
VH and two VL s, yielding tandem scFvs. Another ility is the on of scFvs with
linker peptides that are too short for the two variable regions to fold together (about five amino
acids), forcing scFvs to dimerize. This type is known as diabodies. Diabodies have been shown to
have dissociation constants up to 40-fold lower than corresponding scFvs, g that they
have a much higher affinity to their target. Consequently, diabody drugs could be dosed much
lower than other eutic antibodies and are capable of highly specific ing of tumors in
vivo. Still shorter linkers (one or two amino acids) lead to the formation of trimers, so-called
triabodies or tribodies. Tetrabodies are known and have been shown to exhibit an even higher
affinity to their targets than ies.
The term "humanized antibody" or "human antibody" refers to antibodies which
comprise heavy and light chain variable region ces from a non-human species (e.g., a
mouse) but in which at least a portion of the VH and/or VL sequence has been altered to be more
"human-like", i.e., more similar to human germline variable sequences. One type of
humanized antibody is a CDR-grafted antibody, in which human CDR sequences are introduced
into non-human VH and VL sequences to e the corresponding nonhuman CDR sequences.
Means for making ic, afted and humanized antibodies are known to those of
ordinary skill in the art (see, e. g., U.S. Pat. Nos. 4,816,567 and 5,225,539). One method for
making human antibodies employs the use of transgenic animals, such as a transgenic mouse.
These transgenic animals contain a substantial portion of the human dy producing genome
inserted into their own genome and the animal's own endogenous antibody production is
rendered deficient in the production of antibodies. Methods for making such transgenic animals
are known in the art. Such transgenic animals may be made using XenoMouse.RTM. technology
or by using a "minilocus" approach. Methods for making XenoMice.RTM. are described in U.S.
Pat. Nos. 6,162,963, 6,150,584, 6,114,598 and 6,075,181. Methods for making transgenic
animals using the "minilocus" approach are described in U.S. Pat. Nos. 5,545,807, 5,545,806,
825, and WO 93/12227.
Humanization of a non-human antibody has become routine in recent years, and is
now within the knowledge of one skilled in the art. l companies provide services to make a
humanized antibody, e.g., Xoma, Aries, Medarex, PDL and Cambridge Antibody Technologies.
zation protocols are extensively described in technical literature, e. g., Kipriyanov and Le
Gall, lar hnol, Vol. 26, pp 39-60 , Humana Press, Totowa, N.J.; Lo, Methods
Mol. Biol., Vol. 248, pp 135-159 (2004), Humana Press, Totowa, N.J.; Wu et al., J. Mol. Biol.
Vol. 294, pp. 151-162 (1999).
In certain embodiments, antibodies useful in the present invention may be expressed
in cell lines other than hybridoma cell lines. Sequences encoding particular antibodies may be
used for transformation of a suitable mammalian host cell by known methods for ucing
polynucleotides into a host cell, including, for example packaging the polynucleotide in a virus
(or into a viral vector) and ucing a host cell with the virus (or vector), or by transfection
procedures known in the art, as exemplified by U.S. Pat. Nos. 4,399,216, 4,912,040, 4,740,461
and 4,959,455. The transformation procedure used may depend upon the host to be transformed.
Methods for introduction of heterologous polynucleotides into mammalian cells are known in the
art and e, but are not d to, dextran-mediated transfection, calcium phosphate
precipitation, polybrene mediated transfection, protoplast fusion, electroporation, encapsulation
of the polynucleotide(s) in liposomes, mixing nucleic acid with positively-charged lipids, and
direct microinjection of the DNA into .
A nucleic acid molecule encoding the amino acid sequence of a heavy chain constant
region, a heavy chain le region, a light chain constant region, or a light chain variable
region of an antibody, or a fragment thereof in a suitable combination if desired, is/are inserted
into an appropriate expression vector using standard ligation techniques. The antibody heavy
chain or light chain constant region may be appended to the C-terminus of the appropriate
variable region and is ligated into an expression vector. The vector is typically selected to be
functional in the particular host cell employed (i.e., the vector is compatible with the host cell
ery such that amplification of the gene and/or expression of the gene may occur). For a
review of sion vectors, see Methods Enzymol., Vol. 185, (Goeddel, ed.), 1990, Academic
Press.
Identification of specific binding agents
Suitable HPTPB-ECD binding agents may be identified using a variety of techniques
known in the art. For example, candidate agents can be screened for binding to HPTPB, and
screened for activity. Generally, the ate agents will first be screened for g and those
that show selective binding will then be screened to determine ability to inhibit the HPTPB-
mediated dephosphorylation of Tie2. In some cases however the candidate agents may be first
screened in vitro for activity.
Determination of binding activity
The selection of a le assay for use in identification of a specific binding agent
depends on the nature of the candidate agent to be screened. One of skill in the art would be able
to choose the appropriate assays for the ular candidate agent.
For e, where the candidates are antibodies or peptibodies, which comprises an
Fc moeity, FACS analysis as described in Example 3B allows the candidate agent to be selected
based on its ability to bind to cells, which express HPTPB. The cell may endogenously s
HPTPB or may be genetically engineered to express HPTPB.
For other candidate agents such as aptamers, other techniques are known in the art.
For example, aptamers which specifically bind to HPTPB can be ed using a technique
known as SELEX (systematic evolution of ligands by exponential enrichment) which selects
specific aptamers through repeated rounds of in vitro selection.
Determination of inhibitor activity by Western blot
As exemplified in Example 4, in one suitable assay HUVECs are ed in serum
free media in the presence or absence of various concentrations of candidate agent and lysates of
the cells are prepared, immunoprecipitated with a Tie2 antibody, resolved by polyacrylamide gel
electrophoresis and transferred to a PVDF membrane. Membrane-bound precipitated
proteins are then serially western blotted with an antiphosphotyrosine antibody to quantify Tie2
phosphorylation followed by a Tie2 antibody to quantify total Tie2. Tie2 phosphorylation is
expressed as the ratio of the anti-phosphotyrosine signal over the total Tie2 . Greater levels
of the anti-phosphotyrosine signal indicate greater HPTPB inhibition by the candidate agent.
Candidate agents that can be ed include, but are not limited to, libraries of
known agents, including l products, such as plant or animal extracts, biologically active
molecules ing proteins, peptides including but not limited to members of random peptide
libraries and combinatorial chemistry derived molecular y made of D- or L-configuration
amino acids, antibodies including, but not limited to, polyclonal, monoclonal, chimeric, human,
single chain antibodies, Fab, F(ab)2 and Fab expression library fragments and eptiope-binding
fragments thereof.
As used herein “antibody fragments” include, but are not limited, to a F(ab’)2, a dimer
or trimer of an Fab, Fv, scFv, or a dia-, tria-, or tetrabody derived from an dy.
METHODS
Disclosed are methods for the treatment of diseases or conditions of the eye,
especially retinopathies, ocular edema and ocular neovascularization. miting examples of
these diseases or conditions include diabetic macular edema, age-related macular degeneration
(wet form), dal neovascularization, ic retinopathy, ocular ischemia, uveitis, retinal
vein occlusion (central or branch), ocular trauma, surgery induced edema, surgery induced
neovascularization, cystoid macular edema, ocular ischemia, uveitis, and the like. These diseases
or conditions are characterized by changes in the ocular ature r progressive or non-
progressive, r a result of an acute disease or condition, or a chronic disease or condition.
One aspect of the disclosed methods relates to diseases that are a direct or indirect
result of es, inter alia, diabetic macular edema and ic retinopathy. The ocular
vasculature of the diabetic becomes unstable over time leading to conditions such as non-
proliferative retinopathy, macular edema, and proliferative retinopathy. As fluid leaks into the
center of the macula, the part of the eye where sharp, straight-ahead vision , the buildup of
fluid and the associated protein begin to deposit on or under the macula. This results in swelling
that disturbs the subject’s l vision. This condition is referred to as “macular edema.”
Another condition that may occur is non-proliferative retinopathy in which vascular changes,
such as microaneurysms, may occur outside the macular region of the eye.
These conditions may or may not progress to diabetic erative retinopathy which
is characterized by neovascularization. These new blood vessels are fragile and are susceptible
to bleeding. The result is scaring of the retina, as well as occlusion or total blockage of the light
pathway through the eye due to the over formation of new blood vessels. Typically, subjects
having diabetic macular edema are suffering from the non-proliferative stage of diabetic
retinopathy; however, it is not uncommon for subjects to only begin manifesting macular edema
at the onset of the proliferative stage.
Diabetic retinopathy, if left untreated, can lead ultimately to blindness. ,
diabetic retinopathy is the leading cause of blindness in working-age populations.
Therefore, the disclosed methods relate to preventing, treating, lling, abating,
and/or otherwise minimizing ocular neovascularization in a subject having diabetes or a subject
diagnosed with diabetes. In addition, subjects having or subjects diagnosed with diabetes can be
alerted to or can be made aware of the risks of developing diabetes-related blindness, therefore
the present s can be used to prevent or delay the onset of non-proliferative retinopathy in
subjects known to be at risk. Likewise, the present methods can be used for ng subjects
having or being diagnosed with non-proliferative ic retinopathy to prevent progression of
the condition.
The disclosed methods relate to preventing or lling ocular neovascularization or
treating a disease or condition that is related to the onset of ocular neovascularization by
administering to a subject an effective amount of an HPTPB-ECD binding agent or a
pharmaceutically acceptable salt f.
One aspect of this method relates to ng or preventing ocular neovascularization
by administering to a subject an effective amount of an ECD binding agent or
pharmaceutically acceptable salt thereof. One ment of this aspect relates to a method for
treating ocular neovascularization comprising administering to a subject a composition
comprising an effective amount of an HPTPB-ECD binding agent or a pharmaceutically
acceptable salt thereof, and one or more carrier or compatible excipient.
Thus, one embodiment of the t disclosure is a method of treating or ting
ocular neovascularization in a subject, comprising administering an effective amount of an
ECD binding agent or a pharmaceutically able salt thereof. Another embodiment
of the t disclosure is a method of treating or preventing ocular neovascularization in a
subject, comprising administering an effective amount of a composition comprising an HPTPB-
ECD binding agent or a pharmaceutically acceptable salt thereof, and one or more carrier or
compatible excipient. Yet another embodiment of the present disclosure is the use of an HPTPB-
ECD binding agent in the treatment of ocular neovascularization.
The disclosed methods also relate to preventing or controlling ocular edema or
treating a disease or condition that is related to the onset of ocular edema by administering to a
subject an HPTPB-ECD binding agent.
One aspect of this method relates to treating or preventing ocular edema by
administering to a subject an effective amount of an HPTPB-ECD binding agent or a
pharmaceutically acceptable salt thereof. One embodiment of this aspect s to a method for
treating ocular edema comprising administering to a subject a composition comprising:
a. an effective amount of an HPTPB-ECD binding agent or a pharmaceutically acceptable
salt thereof; and
b. one or more carriers or compatible excipients.
Thus, one embodiment of the present disclosure is a method of ng or preventing
ocular edema in a subject, sing administering an effective amount of an ECD
binding agent or a pharmaceutically acceptable salt thereof. Another embodiment of the present
disclosure is a method of treating or preventing ocular edema in a subject, comprising
administering an effective amount of a composition comprising HPTPB-ECD g agent or a
pharmaceutically acceptable salt thereof, and one or more carriers or compatible excipients. An
embodiment of the present disclosure is the use of an HPTPB-ECD binding agent in the treatment
of ocular edema.
Another disclosed method relates to preventing or controlling retinal edema or retinal
neovascularization, or ng a e or condition that is related to the onset of retinal edema
or retinal cularization, by administering to a subject an HPTPB-ECD g agent. One
aspect of this method relates to treating or preventing l edema or retinal neovascularization
by administering to a subject an effective amount of an ECD g agent or
pharmaceutically acceptable salt thereof. One embodiment of this aspect relates to a method for
treating retinal edema or retinal cularization comprising administering to a subject a
composition comprising an effective amount of an HPTPB-ECD binding agent or
ceutically acceptable salt thereof, and one or more carriers or compatible excipients.
Thus, one embodiment of the present disclosure is a method of ng or preventing
retinal edema in a subject, sing administering an effective amount of an HPTPB-ECD
binding agent or a pharmaceutically acceptable salt thereof. r embodiment is a method of
treating or preventing retinal neovascularization comprising administering an effective amount of
an HPTPB-ECD binding agent or a pharmaceutically acceptable salt thereof. One embodiment of
the present disclosure is a method of treating or preventing retinal edema in a subject, by
administering a composition comprising an effective amount of an HPTPB-ECD g agent or
a pharmaceutically acceptable salt thereof, and one or more carriers or ible excipients.
r embodiment is a method of treating or preventing retinal neovascularization by
administering an effective amount of an HPTPB-ECD binding agent or a pharmaceutically
acceptable salt thereof, and one or more carriers or compatible ents. Another embodiment
is the use of an HPTPB-ECD binding agent in the treatment of retinal edema. A further
embodiment is the use of an ECD binding agent in the treatment of retinal
neovascularization.
A further sed method relates to ng, preventing or controlling diabetic
retinopathy, or treating a disease or condition that is related to the onset of diabetic retinopathy
by administering to a subject an HPTPB-ECD binding agent.
One aspect of this method relates to treating or preventing diabetic retinopathy by
administering to a subject an effective amount of an HPTPB-ECD binding agent or
pharmaceutically acceptable salt thereof. One embodiment of this aspect s to a method for
ng diabetic retinopathy comprising administering to a subject a composition comprising an
effective amount of an HPTPB-ECD binding agent or a pharmaceutically acceptable salt thereof,
and one or more carrier or compatible excipient.
Thus, one embodiment of the present disclosure is a method of treating or preventing
diabetic retinopathy in a subject, comprising stering an effective amount of an HPTPB-
ECD binding agent or a pharmaceutically acceptable salt thereof. Another embodiment of the
t disclosure is a method of treating or preventing diabetic pathy in a subject, by
administering a composition sing an effective amount of an HPTPB-ECD g agent or
a pharmaceutically acceptable salt thereof, and one or more carriers or compatible excipients.
Yet another embodiment of the present disclosure is the use of an HPTPB-ECD binding agent in
the treatment of diabetic retinopathy.
] A further disclosed method relates to a method for treating or preventing non-
proliferative retinopathy comprising administering to a subject an effective amount of an HPTPB-
ECD binding agent or pharmaceutically acceptable salt thereof.
Another embodiment of this aspect relates to a method for treating or preventing non-
proliferative retinopathy comprising administering to a subject a composition comprising an
effective amount of an HPTPB-ECD binding agent or pharmaceutically acceptable salt thereof;
and one or more carrier or ible excipient.
Thus, one embodiment of the present disclosure is a method of treating or preventing
oliferative retinopathy in a subject, comprising administering an effective amount of an
HPTPB-ECD binding agent or a pharmaceutically acceptable salt f. Another ment
of the present disclosure is a method of treating or preventing non-proliferative retinopathy in a
subject, by administering a composition comprising an effective amount of an HPTPB-ECD
binding agent or a pharmaceutically acceptable salt thereof, and one or more carriers or
ible excipients. Yet r embodiment of the present disclosure is the use of an
HPTPB-ECD binding agent in the treatment of non-proliferative retinopathy.
Yet a further disclosed method relates to preventing or controlling diabetic macular
edema, or treating a disease or condition that is d to the onset of diabetic macular edema by
administering to a subject an HPTPB-ECD binding agent.
] One aspect of this method s to ng or preventing diabetic macular edema by
administering to a subject an effective amount of an HPTPB-ECD binding agent or
pharmaceutically acceptable salt thereof. One embodiment of this aspect s to a method for
treating diabetic macular edema comprising administering to a subject a composition comprising:
a) an effective amount of one or more of an HPTPB-ECD g agent or a pharmaceutically
acceptable salt thereof; and b)one or more carriers or compatible excipients.
Thus, one embodiment of the present disclosure is a method of ng or preventing
diabetic macular edema in a t, comprising administering an effective amount of an HPTPB-
ECD binding agent or a pharmaceutically acceptable salt thereof. Another embodiment of the
present disclosure is a method of treating or preventing diabetic macular edema in a subject, by
administering a composition comprising an effective amount of an HPTPB-ECD binding agent or
a ceutically acceptable salt thereof, and one or more rs or compatible excipients.
Yet r embodiment of the present disclosure is the use of an HPTPB-ECD binding agent in
the treatment of diabetic macular edema.
Another ment of the present disclosure is a method for treating, or preventing
age-related wet form macular ration edema in a subject, comprising stering an
effective amount of an HPTPB-ECD binding agent or a pharmaceutically acceptable salt thereof.
Another ment of the present disclosure is a method of treating or preventing age-related
wet form macular degeneration edema in a subject, by administering a composition comprising
an ive amount of an HPTPB-ECD binding agent or a pharmaceutically acceptable salt
thereof, and one or more carriers or compatible excipients. Yet another embodiment of the
present disclosure is the use of an HPTPB-ECD g agent in the treatment of age-related wet
form macular degeneration edema.
A further embodiment is a method for treating, preventing or controlling dal
neovascularization, central retinal vein occlusion, branch retinal vein ion, ocular trauma,
surgery induced edema, surgery induced neovascularization, cystoid macular edema, ocular
ischemia, or uveitis, by administering to a subject an effective amount of an HPTPB-ECD
binding agent or a pharmaceutically acceptable salt thereof. Another embodiment is a method
for treating, preventing or controlling choroidal neovascularization, central retinal vein occlusion,
branch retinal vein occlusion, ocular , surgery induced edema, surgery induced
neovascularization, cystoid macular edema, ocular ischemia, retinal angiomatous proliferation,
r telangiectasia, or uveitis, by administering to a subject a composition comprising an
effective amount of an HPTPB-ECD binding agent or a pharmaceutically acceptable salt thereof,
and one or more carriers or compatible excipients. Yet another embodiment of the present
disclosure is the use of an HPTPB-ECD binding agent in the treatment of choroidal
neovascularization, central retinal vein occlusion, branch retinal vein ion, ocular trauma,
surgery induced edema, surgery induced neovascularization, cystoid macular edema, ocular
ia, retinal angiomatous proliferation, macular telangiectasia or uveitis.
Another embodiment is a ition for treating or preventing an ocular disorder,
comprising an HPTPB-ECD binding agent or pharmaceutically able salt thereof, and one or
more pharmaceutically acceptable carrier. Yet another ment is a composition for treating
or preventing an ocular disorder, comprising an HPTPB-ECD g agent or pharmaceutically
acceptable salt thereof, and one or more pharmaceutically acceptable carrier ition wherein
the ocular disorder is ocular neovascularization, ocular edema, retinal neovascularization,
diabetic retinopathy, diabetic macular edema, age-related macular degeneration, choroidal
neovascularization, central retinal vein occlusion, branch retinal vein occlusion, ocular trauma,
surgery induced edema, surgery induced neovascularization, cystoid macular edema, ocular
ischemia, oliferative retinopathy, retinal angiomatous proliferation, macular telangiectasia,
or uveitis.
In some embodiments, the HPTPB-ECD binding agent or pharmaceutically acceptable
salt thereof is used for treating an ocular disorder. In some embodiments, the HPTPB-ECD
binding agent or pharmaceutically able salt f is used for treating an ocular disorder,
wherein the ocular disorder is ocular neovascularization, ocular edema, retinal
neovascularization, ic retinopathy, diabetic macular edema, age-related macular
degeneration, choroidal neovascularization, central retinal vein occlusion, branch retinal vein
occlusion, ocular trauma, surgery d edema, surgery induced neovascularization, cystoid
macular edema, ocular ia, non-proliferative retinopathy, retinal atous proliferation,
macular telangiectasia or s.
In still other embodiments, the HPTPB-ECD binding agent or pharmaceutically
acceptable salt thereof is used for the manufacture of a medicament for treating an ocular
disorder. In some embodiments the ocular disorder is ocular neovascularization, ocular edema,
retinal neovascularization, diabetic pathy, diabetic macular edema, age-related macular
degeneration, choroidal neovascularization, central retinal vein occlusion, branch retinal vein
occlusion, ocular trauma, surgery induced edema, surgery induced neovascularization, cystoid
macular edema, ocular ischemia, non-proliferative retinopathy, l angiomatous proliferation,
macular iectasia or s.
Dosing
Effective dosages and schedules for administering the ECD g agent
may be determined empirically, and making such determinations is within the skill in the art.
Those skilled in the art will understand that the dosage of the agent that must be administered
will vary depending on, for example, the subject which will e the agent, the route of
administration, the particular type of agent used and other drugs being stered to the
subject. For example, guidance in selecting appropriate doses for dies is found in the
literature on therapeutic uses of antibodies, e. g., Handbook of Monoclonal Antibodies, Ferrone et
al., eds., Noges ations, Park Ridge, N.J., (1985) ch. 22 and pp. 303-357; Smith et al.,
Antibodies in Human Diagnosis and Therapy, Haber et al., eds., Raven Press, New York (1977)
pp. 365-389. A typical dose of the agent used alone might range from about 0.01 mg/kg to up to
500 mg/kg of body weight or more per day, or from about 0.01 mg/kg to about 50 mg/kg, or from
0.1 mg/kg to about 50 mg/kg, or from about 0.1 mg/kg to up to about 10 mg/kg, or from about
0.2 mg/kg to about 1 mg/kg, depending on the s mentioned above.
One embodiment relates to a method for treating ocular edema and/or
neovascularization comprising administering to a subject from about 0.01 mg/kg to about 50
mg/kg of an HPTPB-ECD g agent or pharmaceutically acceptable salt thereof. Another
iteration of this embodiment relates to administering to a subject from about 0.1 mg/kg to about
mg/kg by weight of the t being treated, an HPTPB-ECD binding agent or
pharmaceutically acceptable salt thereof. A further ion of this embodiment relates to a
method for treating or preventing diseases or conditions related to ocular edema and/or
neovascularization comprising administering to a subject from about 1 mg/kg to about 10 mg/kg
by weight of the subject an ECD binding agent or ceutically able salt
thereof. Yet another iteration of this embodiment relates to a method for treating or preventing
diseases or conditions related to ocular edema and/or neovascularization comprising
administering to a subject from about 5 mg/kg to about 10 mg/kg by weight of the t an
HPTPB-ECD binding agent or ceutically acceptable salt thereof. In a further iteration of
this embodiment relates to a method for treating or preventing diseases or conditions related to
ocular edema and/or neovascularization comprising administering to a subject from about 1
mg/kg to about 5 mg/kg by weight of the subject an HPTPB-ECD binding agent or
pharmaceutically acceptable salt thereof. In a yet further iteration of this embodiment relates to a
method for treating or ting diseases or conditions related to ocular edema and/or
neovascularization comprising administering to a subject from about 3 mg/kg to about 7 mg/kg
by weight of the subject an HPTPB-ECD binding agent or pharmaceutically acceptable salt
thereof.
The dosing schedules for administration of an HPTPB-ECD binding agent e, but are
not limited to, once daily, three-times weekly, twice weekly, once weekly, three times, twice monthly,
once monthly and once every other month.
r disclosed are methods of treating or preventing one or more of the diseases or
ions described herein above d to ocular edema and/or neovascularization that are the
result of administration of another pharmaceutically active agent. As such, this aspect s to a
method comprising stering to a subject a composition comprising: a) an effective amount
of an HPTPB-ECD binding agent or pharmaceutically acceptable salt thereof; b) one or more
additional pharmaceutically active agents; and c) one or more carriers or compatible excipients.
The methods of the present invention may be ed with the standard of care,
including, but not limited to, laser treatment.
Non-limiting examples of pharmaceutically active agents suitable for combination
with an HPTPB-ECD binding agent include anti-infectives, i.e., aminoglycosides, antiviral
agents, antimicrobials, anticholinergics/antispasmotics, antidiabetic agents, antihypertensive
agents, oplastics, cardiovascular agents, central nervous system agents, coagulation
modifiers, hormones, logic agents, immunosuppressive agents, lmic preparations
and the like.
The disclosed method also relates to the administration of the disclosed agents and
compositions. Administration can be systemic via subcutaneous or iv. administration; or the
HPTP-B inhibitor will be administered directly to the eye, e. g., local. Local methods of
stration include, for example, by eye drops, subconjunctival injections or implants,
intravitreal injections or implants, sub-Tenon's injections or implants, incorporation in surgical
ting solutions, etc.
The disclosed methods relate to administering an HPTPB-ECD binding agent as part
of a pharmaceutical composition. Compositions suitable for local administration are known to
the art (see, for e, U.S. Pat. Publ. 2005/0059639). In various embodiments, compositions
of the invention can comprise a liquid comprising an active agent in solution, in sion, or
both. As used herein, liquid compositions include gels. In one embodiment, the liquid
composition is aqueous. Alternatively, the composition can take form of an ointment. In another
embodiment, the composition is an in situ gellable aqueous composition. Such a composition can
comprise a gelling agent in a concentration effective to promote gelling upon contact with the
eye or lacrimal fluid in the or of the eye. Aqueous compositions of the invention have
ophthalmically compatible pH and osmolality. The ition can comprise an ophthalmic
depot ation comprising an active agent for subconjunctival administration. The
2012/060263
microparticles comprising active agent can be embedded in a biocompatible pharmaceutically
acceptable r or a lipid encapsulating agent. The depot formulations may be adapted to
release all or substantially all the active material over an extended period of time. The polymer
or lipid matrix, if present, may be adapted to degrade sufficiently to be transported from the site
of administration after release of all or ntially all the active agent. The depot formulation
can be a liquid formulation, comprising a pharmaceutical acceptable polymer and a dissolved or
dispersed active agent. Upon injection, the polymer forms a depot at the injections site, e. g., by
gelifying or precipitating. The composition can comprise a solid article that can be inserted in a
suitable location in the eye, such as between the eye and eyelid or in the conjuctival sac, where
the article es the active agent. Solid articles suitable for implantation in the eye in such
fashion generally comprise polymers and can be bioerodible or non-bioerodible.
In one embodiment of the disclosed methods, a human subject with at least one
visually ed eye is treated with 2-4000 ug of an HPTPB-ECD binding agent via intravitreal
ion. Improvement of clinical symptoms are monitored by one or more methods known to
the art, for example, indirect ophthalmoscopy, fundus photography, fluorescein angiopathy,
electroretinography, external eye examination, slit lamp biomicroscopy, applanation try,
pachymetry, l nce tomography and autorefaction. Subsequent doses can be
administered weekly or monthly, e.g., with a frequency of 2-8 weeks or 1-12 months apart.
The disclosed methods include administration of the disclosed agents in combination
with a pharmaceutically acceptable carrier. “Pharmaceutically acceptable” means a material that
is not biologically or otherwise undesirable, i.e., the material may be administered to a subject
without causing any undesirable biological effects or interacting in a rious manner with any
of the other components of the pharmaceutical formulation in which it is contained. The carrier
would naturally be selected to minimize any degradation of the active ingredient and to minimize
any e side effects in the subject, as would be well known to one of skill in the art. In
another aspect, many of the disclosed agents can be used prophylactically, i.e., as a tive
agent, either neat or with a ceutically able carrier. The ionic liquid compositions
disclosed herein can be conveniently formulated into pharmaceutical compositions composed of
neat ionic liquid or in association with a pharmaceutically acceptable carrier. See Remington's
ceutical Sciences, 18th ed., Gennaro, AR. Ed., Mack Publishing, Easton Pa. (1990),
which discloses typical carriers and tional methods of preparing pharmaceutical
compositions that can be used in conjunction with the preparation of formulations of the agents
described herein and which is incorporated by nce herein. Such pharmaceutical carriers,
most typically, would be standard carriers for administration of compositions to humans and non-
, including solutions such as sterile water, saline and buffered solutions at logical
pH. Other agents can be stered according to standard procedures used by those skilled in
the art. For example, pharmaceutical compositions can also e one or more additional
active ingredients such as antimicrobial agents, anti-inflammatory agents, anesthetics and the
like.
Examples of pharmaceutically-acceptable carriers include, but are not limited to,
saline, Ringer’s solution and dextrose solution. The pH of the solution is preferably from about 5
to about 8, and more preferably from about 7 to about 7.5. Further carriers include sustained
release preparations such as semipermeable matrices of solid hydrophobic polymers containing
the disclosed agents, which matrices are in the form of shaped articles, e. g., films, liposomes,
microparticles, or microcapsules. It will be apparent to those persons skilled in the art that
certain carriers can be more preferable depending upon, for instance, the route of administration
and concentration of composition being administered. Other agents can be administered
according to standard procedures used by those skilled in the art.
Pharmaceutical ations can include additional carriers, as well as ners,
diluents, buffers, preservatives, e active agents and the like in addition to the agents
disclosed herein. Pharmaceutical formulations can also include one or more additional active
ingredients such as antimicrobial , anti-inflammatory agents, anesthetics and the like.
For the purposes of the present disclosure the term “excipient” and er” are used
interchangeably throughout the description of the present disclosure and said terms are defined
herein as, “ingredients which are used in the ce of formulating a safe and effective
pharmaceutical composition.”
The formulator will tand that excipients are used ily to serve in
delivering a safe, stable and functional pharmaceutical, serving not only as part of the overall
vehicle for delivery but also as a means for achieving effective absorption by the recipient of the
active ingredient. An excipient may fill a role as simple and direct as being an inert filler, or an
excipient as used herein may be part of a pH stabilizing system. The formulator can also take
advantage of the fact the agents of the present invention have improved cellular potency,
pharmacokinetic properties.
The disclosed agents can also be present in liquids, emulsions, or suspensions for
delivery of active therapeutic agents. Liquid pharmaceutically administrable compositions can,
for example, be prepared by dissolving, dispersing, etc., an active agent as described herein and
optional pharmaceutical adjuvants in an excipient, such as, for example, water, saline aqueous
dextrose, glycerol, ethanol and the like, to thereby form a on or suspension. If desired, the
pharmaceutical composition to be administered can also contain minor amounts of ic
auxiliary nces such as g or fying agents, pH ing agents and the like, for
e, sodium acetate, sorbitan monolaurate, anolamine sodium acetate, triethanolamine
oleate, etc. Actual methods of preparing such dosage forms are known, or will be apparent, to
those skilled in this art, for example see Remington’s Pharmaceutical Sciences, referenced above.
KITS
Also disclosed are kits comprising the agents and compositions to be delivered into a
human, mammal, or cell. The kits can comprise one or more packaged unit doses of a
composition comprising one or more agents to be delivered into a human, mammal, or cell. The
unit dosage ampoules or multi-dose containers, in which the agents to be red are packaged
prior to use, can se a hermetically sealed container enclosing unit dose of the composition,
or multiples unit doses. The agents can be packaged as a sterile formulation, and the
ically sealed container is ed to preserve sterility of the formulation until use.
EXAMPLES
EXAMPLE 1
Production of HPTPB Extracellular Domain Protein
Full length HPTPB cDNA (SEQ ID NO: 1) is cloned from a human placental library
according to the manufacturer's (Origene) instructions. A cDNA encoding the entire soluble
extracellular domain (ECD) of HPTPB is cloned by PCR from the full length cDNA coding for
amino acids 1-1621 with an added c-terminal His-His-His-His-His-His-Gly (6His-Gly) (SEQ ID
NO:3). The resulting cDNA is cloned into mammalian expression vectors for transient (pShuttle-
CMV) or stable (pcDNA3.l(-)) expression in HEK293 cells. To obtain purified HPTPB ECD
(BED), HEK293 cells transfected with a BECD expression vector are incubated in OptiMEM-
serum free (Gibco) for 24 hours under normal growth ions. The conditioned media is then
recovered, centrifuged to remove debris, and 1 mL of washed Ni-NTA agarose (Qiagen) (SOOuL
2012/060263
packed al) is added to each 10uL of cleared media and allowed to rock overnight at 40 C.
On the following day, the mixture is loaded into a column and washed with 20 bed volumes of
50 mM NaHzPO4, 300 mM NaCl, 20 mM ole, pH 8. The purified HPTPB extracellular
domain n (SEQ ID NO:4) is then eluted with 200 uL/elution in 50 mM 4, 300 mM
NaCl, 250 mM Imidazole, pH 8. Fractions are analyzed for protein content using reducing-
denaturing SDS-polyacrylimide gel electrophoresis and detected by silver stain (Invitrogen) and
confirmed by mass spectrometry.
Example 2
Generation of monoclonal antibodies to HPTPB extracellular domain
Purified HPTPB extracellular domain protein is produced, for example by the
ure described in Example 1. For production of the HPTPB extracellular domain
immunogen, the purified HPTPB extracellular domainHis protein is conjugated to porcine
thyroglobulin (Sigma) using EDC coupling chemistry (Hockfield, S. et al., (1993) Cold Spring
Habor Laboratory Press. Vol. 1 pp. 111-201, Immunocytochemistry). The ing HPTPB
extracellular domain-thyroglobulin conjugate is dialyzed against PBS, pH 7.4. Adult Balb/c
mice are then immunized subcutaneously with the conjugate (100-200 Mg) and complete Freund's
adjuvant in a 1:1 mixture. After 2-3 weeks, the mice are injected intraperitoneally or
subcutaneously with incomplete Freund's adjuvant and the conjugate in a 1:1 mixture. The
injection is repeated at 4-6 weeks. Sera are collected from mice 7 days post-third-injection and
assayed for immunoreactivity to HPTPB extracellular domain antigen by ELISA and n
blotting. Mice that display a good response to the antigen are boosted by a single intra-spleen
injection with 50 ul of purified HPTPB extracellular domain protein mixed 1:1 with Alum
hydroxide using a 31 gauge extra long needle (Goding, J. W., (1996) Monoclonal Antibodies:
Principles and Practices. Third Edition, Academic Press Limited. p. 145). Briefly, mice are
anesthetized with 2.5% avertin, and a 1 centimeter incision is d on the skin and left oblique
body wall. The antigen mixture is administered by inserting the needle from the posterior portion
to the anterior portion of the spleen in a longitudinal injection. The body wall is d and the
skin is sealed with two small metal clips. Mice are monitored for safe recovery. Four days after
surgery the mouse spleen is removed and single cell suspensions are made for fusion with mouse
myeloma cells for the creation of hybridoma cell lines (Spitz, M., (1986) s In
logy, Vol. 121. Eds. John J, Lagone and Helen Van s. pp. 33-41 (Academic
Press, New York, NY)). Resulting hybridomas are cultured in Dulbeccos modified media
(Gibco) supplemented with 15 % fetal calf serum (Hyclone) and hypoxathine, aminopterin
and thymidine.
Screening for positive hybridomas begins 8 days after the fusion and continues
for 15 days. Hybridomas producing anti-ΗΡΤΡβ extracellular domain antibodies are
identified by ELISA on two sets of 96- well plates: one coated with the histidine tagged-
ΗΡΤΡβ ellular domain and another one coated with a histidine-tagged bacterial MurA
protein as a negative control. The secondary antibody is a donkey anti-mouse IgG labeled
with horseradish peroxidase (HRP) on Immunoresearch). Immunoreactivity is
monitored in wells using color development initiated by ABTS tablets dissolved in TBS
buffer, pH 7.5. The individual HRP reaction mixtures are terminated by adding 100
iters of 1% SDS and reading absorbance at 405 nm with a spectrophotometer.
Hybridomas producing antibodies that interact with ΗΡΤΡβ extracellular -6His, and
not with the murA-6His protein are used for further is. Limiting dilutions (0.8 cells per
well) are performed twice on positive clones in 96 well plates, with clonality defined as
having greater than 99% of the wells with positive vity. Isotypes of antibodies are
determined using the rip technology (Roche). To obtain purified antibody for further
evaluation, tissue culture supernatants are affinity purified using a protein A or a n G
column.
] Six monoclonal antibodies immunoreactive to ΗΡΤΡβ-ECD protein were
isolated and given the following nomenclature, R15E6, R12A7, R3A2, R11C3, R15G2 and
R5A8. Based on its reaction with ΗΡΤΡβ-ΕCD protein in ELISA and in western blots, R15E6
was selected for further study.
The monoclonal antibody R15E6
The monoclonal antibody R15E6 was identified and characterized as
described in Example 2 of the t application and in United States Pat. No., 7,973,142;
the procedure and results are summarized below.
A. R15E6 binds endogenous ΗΡΤΡβ as trated by immunoprecipitation.
Materials: Human umbilical vein endothelial cells (HUVECs), EGM media, and
trypsin neutralizing solution from Cambrex; M I (Gibco), bovine serum albumin (BSA;
Santa Cruz), phosphate ed saline (PBS; Gibco), Growth Factors including Angiopoietin l
(Angl), ar endothelial growth factor (VEGF) and fibroblast growth factor (FGF) (R&D
s), Tie2 monoclonal antibody (Duke University/P&GP), VEGF receptor 2 (VEGFR2)
polyclonal antibody (Whitaker et. al), protein A/G agarose (Santa Cruz), Tris-Glycine pre-cast
gel electrophoresis/transfer system (6-8%) (Invitrogen), PVDF membranes (Invitrogen), lysis
buffer (20 mm Tris-HCl, 137 mm NaCl, 10% glycerol, 1% triton-X-100, 2 mM EDTA, 1 mM
NaOH, 1 mM NaF, 1 mM PMSF, 1 ug/ml leupeptin, 1 ug/ml pepstatin).
Method: HUVECs were pre-treated for 30 min with antibody (in OPTIMEM) or
OPTIMEM I alone. After l of pre-treatment, cells were treated with Angl (100 ng/ml) for
6 minutes in PBS+0.2% BSA and lysed in lysis buffer. Lysates were run directly on a Tris-
Glycine gel or immunoprecipitated with 2-5 ug/ml Tie-2 antibody or 10 ug/ml R15E6 antibody
and protein A/G agarose. Immunoprecipitated samples were rinsed once with lysis buffer and
boiled for 5 min in l X times sample buffer. s were ed on a Tris-Glycine gel,
transferred to a PVDF membrane, and detected by western blot using the indicated antibodies
(pTYR Ab (PY99, Santa Cruz), Tie-2, VEGFR2 and/or .
Results: By IP/western blotting, R15E6 recognizes a major, high molecular weight
band tent with the size of HPTPB (Fig. 1, Panel A, Lane 2). The less intense, lower
molecular weight bands likely ent less glycosylated precursor forms of HPTPB. An
immunoprecipitation (IP) with control, non-immune IgG shows no bands in the lar weight
range of HPTPB (Fig. 1, Panel A, Lane 1), and a combined Tie2/VEGFR2 IP shows bands of the
expected molecular weight (Fig. 1, Panel A, Lane 3). This result trates that R15E6
recognizes and is specific for HPTPB.
B. R15E6 binds endogenous HPTPB as demonstrated by FACS analysis
Materials: HUVECs, EGM media, and trypsin neutralizing solution from Cambrex;
ary Alexfluor 488-tagged antibody from Molecular Probes; Hanks balanced salt solution
(Gibco); FACSCAN flow cytometer and CellQuest software from Becton Dickenson.
Method: HUVECs are trypsinized, treated with trypsin neutralizing solution and
rinsed with HBSS. R15E6 antibody (0.6 ug) is added to 250,000 cells in 50ul of HBSS and
incubated on ice for 20 minutes. Cells were rinsed with 1 ml HBSS followed by adding 2 ug of
fluorescent-conjugated secondary antibody for 20 minutes on ice. Cells were rinsed and
ended in 1 ml HBSS then analyzed on the N flow cytometer with CellQuest
software. Control cells were treated with fluorescent-conjugated secondary antibody only.
Results: By FACS analysis, intact HUVECs, R15E6 causes a robust shift (>90% of
cells) in the fluorescence signal compared to the secondary antibody alone (Fig. 1, Panel B). This
result indicates that R15E6 binds to endogenous HPTPB presented on the surface of intact
endothelial cells.
EXAMPLE 4
R15E6 Enhances Tie2 Activation
R15E6 es Tie2 phosphorylation in the absence and presence of the
angiopoietin 1 (Angl), the Tie2 ligand.
Methods: HUVECs are cultured in serum free media as described above in the
presence or absence of various concentrations of R15E6 and with or without added Angl.
Lysates are prepared, immunoprecipitated with a Tie2 dy, resolved by polyacrylamide gel
electrophoresis and transferred to a PVDF membrane. Membrane-bound immunoprecipitated
proteins are then ly western blotted with an antiphosphotyrosine antibody to quantify Tie2
phosphorylation followed by a Tie2 antibody to quantify total Tie2. Tie2 phosphorylation is
expressed as the ratio of the antiphosphotyrosine signal over the total Tie2 .
Results: R15E6 enhances Tie2 phosphorylation both in the absence and presence of
Angl (Fig. 2). This result indicates that binding of R15E6 to HPTPB on the surface of endothelial
cells modulates its ical function ing in enhanced activation of Tie2 in the absence or
ce of ligand.
EXAMPLE 5
tion of anti-VE-PTP extracellular domain antibodies
A. Production of mouse VE-PTP extracellular domain protein (VE-PTP-ECD)
VE-PTP —ECD may be produced by any suitable method. Such methods are well
known in the art. For example, VE-PTP —ECD can be ed using a method similar to
Example 1 of the present disclosure where VE-PTP-ECD cDNA is used in place of cDNA
encoding ΗΡΤΡβ-ECD. SEQ ID NO:5 provides a nucleotide sequence that encodes VE-PTP-
ECD. SEQ ID NO:7 provides the amino acid ce of VE-PTP-ECD.
B. tion of antibodies to VE-PTP ECD
Anti-VE-PTP antibodies are readily generated by methods that are well known
in the art. For example, anti VE-PTP antibodies can be generated using the method of
Example 2 of the t disclosure by substituting VE-PTP-ECD for the ΗΡΤΡβ
extracellular domain and immunizing rats with the resulting protein. The rat anti-mouse VEPTP
antibody used in the present studies was kindly provided by Dr. D. Vestweber (mAb
109). The antibody was generated as described in Baumer S. et al., Blood, 2006; 107: 4754-
4762. Briefly, the antibody was generated by immunizing rats with a -Fc fusion
protein. Immunization, hybridoma-fusion, and screening were conducted as described in
Gotsch U., et al., J Cell Sci. 1997, Vol. 110, pp. 8 and Bosse R. and Vestweber D., Eur
J Immunol. 1994, Vol. 24, pp. 3019-3024.
The fusion protein was constructed such that the first 8 fibronectin type III-
like repeats ending with the amino acid proline at on 732 of VE-PTP were fused in
frame with the Fc part of human IgGl (starting with amino acid e at position 239). This
construct cloned into pcDNA3 (Invitrogen) was stably transfected into CHO cells, and the
fusion protein was purified by protein A Sepharose affinity purification.
Intravitreal injections of an anti- VE-PTP ECD antibody
Laser-induced Choroidal cularization Model: The choroidal
neovascularization model is considered to represent a model of neovascular age-related
macular degeneration. Choroidal NV was generated as previously described. See Tobe T, et
al., Am. J. Pathol. 1998, Vol. 153, pp. 1641-1646. Adult 6 mice had laser-induced
rupture of Bruch' s membrane in three locations in each eye and were then given 1 μL·
intravitreal injections of 1 or 2 μg of a VE-PTP-ECD antibody (IgG2a), in one eye and
vehicle (5% dextrose) in the fellow eye. These treatments were repeated on day 7. Fourteen
days after laser, the mice were perfused with fluorescein-labeled n (2xl06 average MW,
Sigma, St. Louis, MO) and the extent of neovascularization was assessed in choroidal flat
mounts by fluorescence copy. The area of CNV at each Bruch's membrane rupture site
was measured by image analysis by an
observer masked with respect to treatment group. The area of CNV is the average of the three
rupture sites in one eye. As shown in Fig. 3, treatment with the VE-PTP-ECD dy
significantly reduced choroidal neovascularization at both 1 and 2 ug doses versus treatment with
vehicle control.
Example 7
Oxygen-Induced Ischemic Retinopathy
] The oxygen-induced ischemic retinopathy model is considered to represent a model of
proliferative diabetic retinopathy. Ischemic retinopathy was produced in C57BL/6 mice by a
method described by Smith, L.E.H., et al. Oxygen-induced retinopathy in the mouse. Invest.
Ophthalmol. Vis. Sci. 35, 101-111 (1994).
C57BL/6 mice at postnatal day 7 (P7) and their mothers were placed in an airtight
chamber and exposed to xia (75 i 3% oxygen) for five days. Oxygen was continuously
red with a PROOX model 110 oxygen controller (Reming Bioinstruments Co., ld,
NY). On P12, mice were returned to room air and under a dissecting microscope, a Harvard
Pump Microinjection System and pulled glass es were used to deliver a 1 ul itreal
injection of 1 or 2 ug of a VE-PTP-ECD antibody was made in one eye and vehicle was injected
in the fellow eye. At P17, the area of NV on the surface of the retina was measured at P17 as
previously described. See Shen J, et al., Invest. Ophthalmol. Vis. Sci. 2007, Vol. 48, pp. 4335-
4341. Briefly, mice were given an intraocular injection of 1 ul containing 0.5 ug rat anti-mouse
PECAM antibody (Pharmingen, San Jose, CA). Twelve hours later, the mice were euthanized,
the eyes fixed in 10% formalin. The retinas were dissected, incubated for 40 minutes in 1:500
goat at IgG conjugated with Alexa488 (Invitrogen, Carlsbad, CA), washed, and whole
mounted. An observer masked with respect to treatment group examined the slides with a Nikon
Fluorescence microscope and measured the area of NV per retina by computerized image
analysis using ImagePro Plus software (Media Cybernetics, Silver Spring, MD). Fig. 4 shows
that treatment with the VE-PTP-ECD antibody significantly d retinal neovascularization at
both 1 and 2 ug doses versus ent with vehicle control. Fig. 5 shows representative retinal
whole mounts from a mouse treated with e versus a mouse treated with 2 ug of the VE-
PTP-ECD antibody.
Example 8
aneous injection of a VE-PTP-ECD antibody
The oxygen-induced ischemic pathy model was conducted as described in
Example 7 (containment in a 75% oxygen atmosphere from P5 to P12) for intravitreal dosing
except that the VE-PTP-ECD antibody (1 mg/kg) was dosed subcutaneously at P12 when the
mice were returned to room air and again on days P14 and P16 (three total doses).
Neovascularization was assessed as described above on day (P17). Fig. 6 shows that
subcutaneous dosing of the VE-PTP-ECD antibody reduces the area of retinal
neovascularization.
Example 9
] The experiment described in Example 8 was repeated at a subcutaneous dose of
2 mg/kg. (Fig. 7)
While a number of embodiments of this disclosure are described, it is apparent that
the basic examples may be altered to provide other ments that utilize or encompass the
HPTPB-ECD binding agent, s and processes of this invention. The embodiments and
examples are for illustrative es and are not to be interpreted as limiting the disclosure, but
rather, the appended claims define the scope of this invention.
Claims (21)
1. Use of an ΗΡΤΡβ-ECD binding agent or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the alleviation of an ocular edema.
2. The use according to claim 1, wherein the ment further comprises a pharmaceutically acceptable carrier.
3. The use according to claim 1 or claim 2, wherein the ocular edema is surgery induced edema.
4. The use according to claim 1 or claim 2, wherein the ocular edema is retinal edema.
5. The use according to claim 1 or claim 2, n the ocular edema is diabetic macular edema.
6. The use according to claim 1 or claim 2, wherein the ocular edema is cystoid macular edema.
7. The use ing to any one of claims 1 to 6, wherein the ΗΡΤΡβ-ECD binding agent is an dy, a protein, a peptide, an aptamer, a peptibody, an adnectin, or a nucleic acid, that binds to the extracellular portion of ΗΡΤΡβ.
8. The use according to any one of claims 1 to 6, wherein the ΗΡΤΡβ-ECD binding agent is a monoclonal dy or an antigen binding fragment thereof, or a polyclonal antibody or an antigen binding fragment thereof.
9. The use ing claim 8, wherein the ΗΡΤΡβ-ECD binding agent is a monoclonal antibody.
10. The use according to any one of claims 1 to 6, wherein the ΗΡΤΡβ-ECD g agent is: the monoclonal antibody produced by hybridoma cell line ATCC No. PTA-7580.
11. The use according to claim 8, wherein the ΗΡΤΡβ-ECD binding agent is an antigen binding fragment of an antibody, wherein the antigen binding fragment is a F(ab')2, Fab, dimer of a Fab, Fv, dimer of a Fv, or dimer of a scFv.
12. The use according to claim 8, wherein the ΗΡΤΡβ-ECD binding agent is an antigen binding nt of an antibody, wherein the antigen binding fragment is a F(ab')2, a dimer of a Fab, a dimer of a Fv, or a dimer of a scFv.
13. The use according to claim 7, wherein the ΗΡΤΡβ-ECD g agent is a protein, a e, an aptamer, a peptibody, a nucleic acid, or an adnectin.
14. The use according to any one of claims 1 to 13, wherein the medicament ses a dose of the ΗΡΤΡβ-ECD binding agent or a pharmaceutically acceptable salt thereof from about 0.01 mg/kg to about 500 mg/kg by weight of a subject.
15. The use according to any one of claims 1 to 13, wherein the medicament ses a dose of the ΗΡΤΡβ-ECD binding agent or a pharmaceutically acceptable salt thereof from about 0.1 mg/kg to about 10 mg/kg by weight of a subject.
16. The use according to any one of claims 1 to 15, wherein the ΗΡΤΡβ-ECD binding agent is conjugated to a vehicle.
17. The use according to claim 16, wherein the vehicle is PEG.
18. The use according to any one of claims 1 to 17, wherein the medicament is formulated for administration by cular ion.
19. The use ing to any one of claims 1 to 17 wherein the medicament is formulated for administration by subcutaneous injection.
20. The use according to any one of claims 1 to 17 wherein the medicament is formulated for administration by intravenous injection.
21. The use according to any one of claims 1 to 9 and 14 to 20, wherein the HPTPβ-ECD g agent is a humanized antibody.
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161546708P | 2011-10-13 | 2011-10-13 | |
US61/546,708 | 2011-10-13 | ||
PCT/US2012/060263 WO2013056233A1 (en) | 2011-10-13 | 2012-10-15 | Treatment of ocular disease |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ623275A NZ623275A (en) | 2016-05-27 |
NZ623275B2 true NZ623275B2 (en) | 2016-08-30 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US10329357B2 (en) | Treatment of ocular disease | |
US11814425B2 (en) | Antibodies that bind human protein tyrosine phosphatase beta (HPTPbeta) and uses thereof | |
JP7550251B2 (en) | Compositions and methods for the treatment of diabetic macular edema - Patent Application 2007022333 | |
JP2017512790A5 (en) | ||
US20210000969A1 (en) | Monoclonal antibody specifically binding to human plasmalemma vesicle-associated protein pv-1, preparation and use thereof | |
NZ623275B2 (en) | Treatment of ocular disease |