WO2024110770A1 - A new promoter for retinal pigment epithelium (rpe) targeted gene therapy - Google Patents
A new promoter for retinal pigment epithelium (rpe) targeted gene therapy Download PDFInfo
- Publication number
- WO2024110770A1 WO2024110770A1 PCT/IB2022/000679 IB2022000679W WO2024110770A1 WO 2024110770 A1 WO2024110770 A1 WO 2024110770A1 IB 2022000679 W IB2022000679 W IB 2022000679W WO 2024110770 A1 WO2024110770 A1 WO 2024110770A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- rpe
- polynucleotide
- vector
- gene
- expression cassette
- Prior art date
Links
- 210000003583 retinal pigment epithelium Anatomy 0.000 title claims abstract description 73
- 238000001415 gene therapy Methods 0.000 title abstract description 11
- 230000014509 gene expression Effects 0.000 claims abstract description 58
- 108091033319 polynucleotide Proteins 0.000 claims abstract description 49
- 102000040430 polynucleotide Human genes 0.000 claims abstract description 49
- 239000002157 polynucleotide Substances 0.000 claims abstract description 41
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 claims abstract description 26
- 238000000034 method Methods 0.000 claims abstract description 24
- 201000010099 disease Diseases 0.000 claims abstract description 21
- 239000000790 retinal pigment Substances 0.000 claims abstract description 8
- 108090000623 proteins and genes Proteins 0.000 claims description 73
- 239000013598 vector Substances 0.000 claims description 62
- 150000007523 nucleic acids Chemical group 0.000 claims description 37
- 108090000765 processed proteins & peptides Proteins 0.000 claims description 25
- 229920001184 polypeptide Polymers 0.000 claims description 24
- 102000004196 processed proteins & peptides Human genes 0.000 claims description 24
- 230000001225 therapeutic effect Effects 0.000 claims description 21
- 108091028043 Nucleic acid sequence Proteins 0.000 claims description 20
- 102000039446 nucleic acids Human genes 0.000 claims description 19
- 108020004707 nucleic acids Proteins 0.000 claims description 19
- 108700019146 Transgenes Proteins 0.000 claims description 18
- 230000000694 effects Effects 0.000 claims description 18
- 239000008194 pharmaceutical composition Substances 0.000 claims description 13
- 108010042407 Endonucleases Proteins 0.000 claims description 11
- 239000013607 AAV vector Substances 0.000 claims description 10
- 239000013603 viral vector Substances 0.000 claims description 8
- 208000007014 Retinitis pigmentosa Diseases 0.000 claims description 6
- 206010064930 age-related macular degeneration Diseases 0.000 claims description 5
- 238000003780 insertion Methods 0.000 claims description 5
- 230000037431 insertion Effects 0.000 claims description 5
- 208000002780 macular degeneration Diseases 0.000 claims description 5
- 238000006467 substitution reaction Methods 0.000 claims description 5
- 238000002560 therapeutic procedure Methods 0.000 claims description 5
- 208000027073 Stargardt disease Diseases 0.000 claims description 4
- 238000012217 deletion Methods 0.000 claims description 4
- 230000037430 deletion Effects 0.000 claims description 4
- 208000036443 AIPL1-related retinopathy Diseases 0.000 claims description 2
- 108020005544 Antisense RNA Proteins 0.000 claims description 2
- 108090000994 Catalytic RNA Proteins 0.000 claims description 2
- 102000053642 Catalytic RNA Human genes 0.000 claims description 2
- 208000033810 Choroidal dystrophy Diseases 0.000 claims description 2
- 108091027757 Deoxyribozyme Proteins 0.000 claims description 2
- 206010012689 Diabetic retinopathy Diseases 0.000 claims description 2
- 208000032087 Hereditary Leber Optic Atrophy Diseases 0.000 claims description 2
- 201000003533 Leber congenital amaurosis Diseases 0.000 claims description 2
- 201000000639 Leber hereditary optic neuropathy Diseases 0.000 claims description 2
- 206010025412 Macular dystrophy congenital Diseases 0.000 claims description 2
- 108091030071 RNAI Proteins 0.000 claims description 2
- 206010038848 Retinal detachment Diseases 0.000 claims description 2
- 108091027967 Small hairpin RNA Proteins 0.000 claims description 2
- 108020004459 Small interfering RNA Proteins 0.000 claims description 2
- 208000003571 choroideremia Diseases 0.000 claims description 2
- 230000000295 complement effect Effects 0.000 claims description 2
- 239000003184 complementary RNA Substances 0.000 claims description 2
- 201000006754 cone-rod dystrophy Diseases 0.000 claims description 2
- 238000003197 gene knockdown Methods 0.000 claims description 2
- 230000009368 gene silencing by RNA Effects 0.000 claims description 2
- 108091070501 miRNA Proteins 0.000 claims description 2
- 239000002679 microRNA Substances 0.000 claims description 2
- 230000004264 retinal detachment Effects 0.000 claims description 2
- 108091092562 ribozyme Proteins 0.000 claims description 2
- 239000004055 small Interfering RNA Substances 0.000 claims description 2
- 201000007790 vitelliform macular dystrophy Diseases 0.000 claims description 2
- 102000004533 Endonucleases Human genes 0.000 claims 1
- 238000011282 treatment Methods 0.000 abstract description 18
- 102100025275 Monocarboxylate transporter 3 Human genes 0.000 abstract description 11
- 108091006607 SLC16A8 Proteins 0.000 abstract description 11
- 241000700605 Viruses Species 0.000 abstract description 10
- 210000001519 tissue Anatomy 0.000 abstract description 9
- 101150012136 SLC16A8 gene Proteins 0.000 abstract description 8
- 108700039691 Genetic Promoter Regions Proteins 0.000 abstract description 6
- 239000012634 fragment Substances 0.000 abstract description 6
- 239000013612 plasmid Substances 0.000 abstract description 6
- 208000017442 Retinal disease Diseases 0.000 abstract description 5
- 238000001727 in vivo Methods 0.000 abstract description 4
- 108700008625 Reporter Genes Proteins 0.000 abstract description 3
- 241000701161 unidentified adenovirus Species 0.000 abstract description 3
- 206010038923 Retinopathy Diseases 0.000 abstract description 2
- 230000004075 alteration Effects 0.000 abstract description 2
- 238000013459 approach Methods 0.000 abstract description 2
- 238000002474 experimental method Methods 0.000 abstract description 2
- 238000012360 testing method Methods 0.000 abstract description 2
- 101000577126 Homo sapiens Monocarboxylate transporter 4 Proteins 0.000 abstract 1
- 210000004027 cell Anatomy 0.000 description 88
- 239000002245 particle Substances 0.000 description 33
- 102000004169 proteins and genes Human genes 0.000 description 23
- 230000003612 virological effect Effects 0.000 description 22
- 108020004414 DNA Proteins 0.000 description 14
- 230000002950 deficient Effects 0.000 description 12
- 239000002773 nucleotide Substances 0.000 description 12
- 125000003729 nucleotide group Chemical group 0.000 description 12
- 230000006870 function Effects 0.000 description 11
- 239000005090 green fluorescent protein Substances 0.000 description 11
- 108700028369 Alleles Proteins 0.000 description 10
- 102100031780 Endonuclease Human genes 0.000 description 10
- 239000003814 drug Substances 0.000 description 10
- 230000001105 regulatory effect Effects 0.000 description 10
- 241000699670 Mus sp. Species 0.000 description 9
- 238000004519 manufacturing process Methods 0.000 description 9
- 239000000546 pharmaceutical excipient Substances 0.000 description 9
- 238000013518 transcription Methods 0.000 description 9
- 230000035897 transcription Effects 0.000 description 9
- 125000003275 alpha amino acid group Chemical group 0.000 description 8
- 210000000234 capsid Anatomy 0.000 description 8
- 210000003161 choroid Anatomy 0.000 description 8
- 210000001525 retina Anatomy 0.000 description 8
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 7
- 108091026890 Coding region Proteins 0.000 description 7
- 108700009124 Transcription Initiation Site Proteins 0.000 description 7
- 238000012423 maintenance Methods 0.000 description 7
- 108020004999 messenger RNA Proteins 0.000 description 7
- 239000000126 substance Substances 0.000 description 7
- ZHNUHDYFZUAESO-UHFFFAOYSA-N Formamide Chemical compound NC=O ZHNUHDYFZUAESO-UHFFFAOYSA-N 0.000 description 6
- 239000000203 mixture Substances 0.000 description 6
- 238000011285 therapeutic regimen Methods 0.000 description 6
- 108090000565 Capsid Proteins Proteins 0.000 description 5
- 102100023321 Ceruloplasmin Human genes 0.000 description 5
- 230000009286 beneficial effect Effects 0.000 description 5
- 239000002299 complementary DNA Substances 0.000 description 5
- 208000035475 disorder Diseases 0.000 description 5
- 229940079593 drug Drugs 0.000 description 5
- 230000006698 induction Effects 0.000 description 5
- 229920000642 polymer Polymers 0.000 description 5
- 230000002207 retinal effect Effects 0.000 description 5
- 102100022794 Bestrophin-1 Human genes 0.000 description 4
- 108091033409 CRISPR Proteins 0.000 description 4
- 101000903449 Homo sapiens Bestrophin-1 Proteins 0.000 description 4
- 101000597428 Homo sapiens Nucleoredoxin-like protein 1 Proteins 0.000 description 4
- 102000017298 Monocarboxylate transporters Human genes 0.000 description 4
- 108050005244 Monocarboxylate transporters Proteins 0.000 description 4
- 102100035399 Nucleoredoxin-like protein 1 Human genes 0.000 description 4
- 208000022873 Ocular disease Diseases 0.000 description 4
- 150000001413 amino acids Chemical class 0.000 description 4
- 108010048367 enhanced green fluorescent protein Proteins 0.000 description 4
- 230000002255 enzymatic effect Effects 0.000 description 4
- 239000010410 layer Substances 0.000 description 4
- 150000003839 salts Chemical class 0.000 description 4
- 229940124597 therapeutic agent Drugs 0.000 description 4
- 238000010361 transduction Methods 0.000 description 4
- 230000026683 transduction Effects 0.000 description 4
- 241000702423 Adeno-associated virus - 2 Species 0.000 description 3
- 241000972773 Aulopiformes Species 0.000 description 3
- 238000010354 CRISPR gene editing Methods 0.000 description 3
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 3
- 101710163270 Nuclease Proteins 0.000 description 3
- 238000002105 Southern blotting Methods 0.000 description 3
- 208000037065 Subacute sclerosing leukoencephalitis Diseases 0.000 description 3
- 206010042297 Subacute sclerosing panencephalitis Diseases 0.000 description 3
- 210000001775 bruch membrane Anatomy 0.000 description 3
- 239000012876 carrier material Substances 0.000 description 3
- 238000009396 hybridization Methods 0.000 description 3
- 239000007924 injection Substances 0.000 description 3
- 238000002347 injection Methods 0.000 description 3
- 239000007925 intracardiac injection Substances 0.000 description 3
- 125000002467 phosphate group Chemical group [H]OP(=O)(O[H])O[*] 0.000 description 3
- 235000019515 salmon Nutrition 0.000 description 3
- 239000000523 sample Substances 0.000 description 3
- 230000008685 targeting Effects 0.000 description 3
- 238000013519 translation Methods 0.000 description 3
- 230000000007 visual effect Effects 0.000 description 3
- 238000001262 western blot Methods 0.000 description 3
- NHBKXEKEPDILRR-UHFFFAOYSA-N 2,3-bis(butanoylsulfanyl)propyl butanoate Chemical compound CCCC(=O)OCC(SC(=O)CCC)CSC(=O)CCC NHBKXEKEPDILRR-UHFFFAOYSA-N 0.000 description 2
- KDCGOANMDULRCW-UHFFFAOYSA-N 7H-purine Chemical compound N1=CNC2=NC=NC2=C1 KDCGOANMDULRCW-UHFFFAOYSA-N 0.000 description 2
- 241001164825 Adeno-associated virus - 8 Species 0.000 description 2
- 241000713704 Bovine immunodeficiency virus Species 0.000 description 2
- 108091033380 Coding strand Proteins 0.000 description 2
- 241000702421 Dependoparvovirus Species 0.000 description 2
- 241000713730 Equine infectious anemia virus Species 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 241000713800 Feline immunodeficiency virus Species 0.000 description 2
- 101000834253 Gallus gallus Actin, cytoplasmic 1 Proteins 0.000 description 2
- 108700028146 Genetic Enhancer Elements Proteins 0.000 description 2
- 108010043121 Green Fluorescent Proteins Proteins 0.000 description 2
- 241000238631 Hexapoda Species 0.000 description 2
- 101000729271 Homo sapiens Retinoid isomerohydrolase Proteins 0.000 description 2
- 241000725303 Human immunodeficiency virus Species 0.000 description 2
- 241000829100 Macaca mulatta polyomavirus 1 Species 0.000 description 2
- 241000713869 Moloney murine leukemia virus Species 0.000 description 2
- 102000000562 Monocarboxylic Acid Transporters Human genes 0.000 description 2
- 108010041817 Monocarboxylic Acid Transporters Proteins 0.000 description 2
- 101000597433 Mus musculus Nucleoredoxin-like protein 1 Proteins 0.000 description 2
- 102100031176 Retinoid isomerohydrolase Human genes 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- 241000713311 Simian immunodeficiency virus Species 0.000 description 2
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 2
- 208000036142 Viral infection Diseases 0.000 description 2
- 230000002159 abnormal effect Effects 0.000 description 2
- 239000003242 anti bacterial agent Substances 0.000 description 2
- 238000010367 cloning Methods 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 239000003246 corticosteroid Substances 0.000 description 2
- 230000007812 deficiency Effects 0.000 description 2
- 239000003085 diluting agent Substances 0.000 description 2
- 229960003444 immunosuppressant agent Drugs 0.000 description 2
- 239000003018 immunosuppressive agent Substances 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 230000002458 infectious effect Effects 0.000 description 2
- 238000002372 labelling Methods 0.000 description 2
- 239000007788 liquid Substances 0.000 description 2
- 210000004962 mammalian cell Anatomy 0.000 description 2
- 230000002503 metabolic effect Effects 0.000 description 2
- 238000004806 packaging method and process Methods 0.000 description 2
- 230000036961 partial effect Effects 0.000 description 2
- 108091008695 photoreceptors Proteins 0.000 description 2
- 238000003752 polymerase chain reaction Methods 0.000 description 2
- 230000008569 process Effects 0.000 description 2
- 239000003642 reactive oxygen metabolite Substances 0.000 description 2
- 230000010076 replication Effects 0.000 description 2
- 239000002356 single layer Substances 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 238000010561 standard procedure Methods 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 208000024891 symptom Diseases 0.000 description 2
- 238000003786 synthesis reaction Methods 0.000 description 2
- 230000009885 systemic effect Effects 0.000 description 2
- 238000001890 transfection Methods 0.000 description 2
- 230000009466 transformation Effects 0.000 description 2
- 230000001228 trophic effect Effects 0.000 description 2
- 241000701447 unidentified baculovirus Species 0.000 description 2
- 241001529453 unidentified herpesvirus Species 0.000 description 2
- 230000009385 viral infection Effects 0.000 description 2
- 108020005345 3' Untranslated Regions Proteins 0.000 description 1
- 241001655883 Adeno-associated virus - 1 Species 0.000 description 1
- 241000202702 Adeno-associated virus - 3 Species 0.000 description 1
- 241000580270 Adeno-associated virus - 4 Species 0.000 description 1
- 241001634120 Adeno-associated virus - 5 Species 0.000 description 1
- 241000972680 Adeno-associated virus - 6 Species 0.000 description 1
- 241001164823 Adeno-associated virus - 7 Species 0.000 description 1
- 241000649045 Adeno-associated virus 10 Species 0.000 description 1
- 241000649046 Adeno-associated virus 11 Species 0.000 description 1
- 241000649047 Adeno-associated virus 12 Species 0.000 description 1
- 102400000068 Angiostatin Human genes 0.000 description 1
- 108010079709 Angiostatins Proteins 0.000 description 1
- 102100021569 Apoptosis regulator Bcl-2 Human genes 0.000 description 1
- 102100033890 Arylsulfatase G Human genes 0.000 description 1
- 108091012583 BCL2 Proteins 0.000 description 1
- 101150077415 BEST1 gene Proteins 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 102100026596 Bcl-2-like protein 1 Human genes 0.000 description 1
- 101150008012 Bcl2l1 gene Proteins 0.000 description 1
- 201000004569 Blindness Diseases 0.000 description 1
- 241000701822 Bovine papillomavirus Species 0.000 description 1
- 102000004219 Brain-derived neurotrophic factor Human genes 0.000 description 1
- 108090000715 Brain-derived neurotrophic factor Proteins 0.000 description 1
- 206010006187 Breast cancer Diseases 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- 101150044789 Cap gene Proteins 0.000 description 1
- 108010005939 Ciliary Neurotrophic Factor Proteins 0.000 description 1
- 102100031614 Ciliary neurotrophic factor Human genes 0.000 description 1
- 102100031162 Collagen alpha-1(XVIII) chain Human genes 0.000 description 1
- 108050001175 Connexin Proteins 0.000 description 1
- 102000010970 Connexin Human genes 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- FBPFZTCFMRRESA-FSIIMWSLSA-N D-Glucitol Natural products OC[C@H](O)[C@H](O)[C@@H](O)[C@H](O)CO FBPFZTCFMRRESA-FSIIMWSLSA-N 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 229920002307 Dextran Polymers 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 108010079505 Endostatins Proteins 0.000 description 1
- 102000004190 Enzymes Human genes 0.000 description 1
- 108090000790 Enzymes Proteins 0.000 description 1
- 102000003974 Fibroblast growth factor 2 Human genes 0.000 description 1
- 108090000379 Fibroblast growth factor 2 Proteins 0.000 description 1
- 102000034615 Glial cell line-derived neurotrophic factor Human genes 0.000 description 1
- 108091010837 Glial cell line-derived neurotrophic factor Proteins 0.000 description 1
- 241000713858 Harvey murine sarcoma virus Species 0.000 description 1
- 241000282412 Homo Species 0.000 description 1
- 101000925538 Homo sapiens Arylsulfatase G Proteins 0.000 description 1
- 101001011412 Homo sapiens IQ calmodulin-binding motif-containing protein 1 Proteins 0.000 description 1
- 101001076418 Homo sapiens Interleukin-1 receptor type 1 Proteins 0.000 description 1
- 101001047038 Homo sapiens Inward rectifier potassium channel 13 Proteins 0.000 description 1
- 101001008411 Homo sapiens Lebercilin Proteins 0.000 description 1
- 101000594760 Homo sapiens Nucleoredoxin-like protein 2 Proteins 0.000 description 1
- 101001092166 Homo sapiens RPE-retinal G protein-coupled receptor Proteins 0.000 description 1
- 101000670189 Homo sapiens Ribulose-phosphate 3-epimerase Proteins 0.000 description 1
- 101000652369 Homo sapiens Spermatogenesis-associated protein 7 Proteins 0.000 description 1
- 101000894525 Homo sapiens Transforming growth factor-beta-induced protein ig-h3 Proteins 0.000 description 1
- 101000666127 Homo sapiens Whirlin Proteins 0.000 description 1
- CPELXLSAUQHCOX-UHFFFAOYSA-N Hydrogen bromide Chemical class Br CPELXLSAUQHCOX-UHFFFAOYSA-N 0.000 description 1
- 102000026659 IL10 Human genes 0.000 description 1
- 102100029842 IQ calmodulin-binding motif-containing protein 1 Human genes 0.000 description 1
- 208000032578 Inherited retinal disease Diseases 0.000 description 1
- 102100026016 Interleukin-1 receptor type 1 Human genes 0.000 description 1
- 108090000174 Interleukin-10 Proteins 0.000 description 1
- 108091092195 Intron Proteins 0.000 description 1
- 102100022843 Inward rectifier potassium channel 13 Human genes 0.000 description 1
- 102100027443 Lebercilin Human genes 0.000 description 1
- 102100033356 Lecithin retinol acyltransferase Human genes 0.000 description 1
- 241001465754 Metazoa Species 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 108010025020 Nerve Growth Factor Proteins 0.000 description 1
- 102000007072 Nerve Growth Factors Human genes 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 108700026244 Open Reading Frames Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 102000004590 Peripherins Human genes 0.000 description 1
- 108010003081 Peripherins Proteins 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- XBDQKXXYIPTUBI-UHFFFAOYSA-N Propionic acid Chemical class CCC(O)=O XBDQKXXYIPTUBI-UHFFFAOYSA-N 0.000 description 1
- 108010076504 Protein Sorting Signals Proteins 0.000 description 1
- 241000125945 Protoparvovirus Species 0.000 description 1
- CZPWVGJYEJSRLH-UHFFFAOYSA-N Pyrimidine Chemical compound C1=CN=CN=C1 CZPWVGJYEJSRLH-UHFFFAOYSA-N 0.000 description 1
- 102100035582 Ral-GDS-related protein Human genes 0.000 description 1
- 101710111169 Retinoschisin Proteins 0.000 description 1
- 102100039507 Retinoschisin Human genes 0.000 description 1
- 108091028664 Ribonucleotide Proteins 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 102100030257 Spermatogenesis-associated protein 7 Human genes 0.000 description 1
- 241000713880 Spleen focus-forming virus Species 0.000 description 1
- 108091081024 Start codon Proteins 0.000 description 1
- 101710172711 Structural protein Proteins 0.000 description 1
- 238000010459 TALEN Methods 0.000 description 1
- 108091036066 Three prime untranslated region Proteins 0.000 description 1
- 108010002321 Tight Junction Proteins Proteins 0.000 description 1
- 102000000591 Tight Junction Proteins Human genes 0.000 description 1
- 108010043645 Transcription Activator-Like Effector Nucleases Proteins 0.000 description 1
- 102100021398 Transforming growth factor-beta-induced protein ig-h3 Human genes 0.000 description 1
- 102100022356 Tyrosine-protein kinase Mer Human genes 0.000 description 1
- 241000700618 Vaccinia virus Species 0.000 description 1
- 108010067390 Viral Proteins Proteins 0.000 description 1
- 102100038102 Whirlin Human genes 0.000 description 1
- 238000009825 accumulation Methods 0.000 description 1
- 150000001242 acetic acid derivatives Chemical class 0.000 description 1
- 239000002253 acid Substances 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 230000000202 analgesic effect Effects 0.000 description 1
- 229940035676 analgesics Drugs 0.000 description 1
- 238000004458 analytical method Methods 0.000 description 1
- 239000002870 angiogenesis inducing agent Substances 0.000 description 1
- 210000004102 animal cell Anatomy 0.000 description 1
- 239000000730 antalgic agent Substances 0.000 description 1
- 230000001772 anti-angiogenic effect Effects 0.000 description 1
- 230000002424 anti-apoptotic effect Effects 0.000 description 1
- 230000003110 anti-inflammatory effect Effects 0.000 description 1
- 229940088710 antibiotic agent Drugs 0.000 description 1
- FZCSTZYAHCUGEM-UHFFFAOYSA-N aspergillomarasmine B Natural products OC(=O)CNC(C(O)=O)CNC(C(O)=O)CC(O)=O FZCSTZYAHCUGEM-UHFFFAOYSA-N 0.000 description 1
- 238000003556 assay Methods 0.000 description 1
- 201000011340 autosomal recessive nonsyndromic deafness 31 Diseases 0.000 description 1
- 208000035257 autosomal recessive nonsyndromic hearing loss 31 Diseases 0.000 description 1
- 101150066033 baiap2l2 gene Proteins 0.000 description 1
- 239000003855 balanced salt solution Substances 0.000 description 1
- 230000004888 barrier function Effects 0.000 description 1
- 210000002469 basement membrane Anatomy 0.000 description 1
- 108700000711 bcl-X Proteins 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- 150000001558 benzoic acid derivatives Chemical class 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 230000031018 biological processes and functions Effects 0.000 description 1
- 230000015572 biosynthetic process Effects 0.000 description 1
- 238000006664 bond formation reaction Methods 0.000 description 1
- 239000000872 buffer Substances 0.000 description 1
- 108010018804 c-Mer Tyrosine Kinase Proteins 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- -1 carrier Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000005779 cell damage Effects 0.000 description 1
- 208000037887 cell injury Diseases 0.000 description 1
- 238000005119 centrifugation Methods 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 238000007398 colorimetric assay Methods 0.000 description 1
- 238000004891 communication Methods 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 238000011340 continuous therapy Methods 0.000 description 1
- 230000001276 controlling effect Effects 0.000 description 1
- 229960001334 corticosteroids Drugs 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 230000007850 degeneration Effects 0.000 description 1
- 230000003412 degenerative effect Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 239000005547 deoxyribonucleotide Substances 0.000 description 1
- 125000002637 deoxyribonucleotide group Chemical group 0.000 description 1
- 210000001047 desmosome Anatomy 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 230000004064 dysfunction Effects 0.000 description 1
- 238000004520 electroporation Methods 0.000 description 1
- 230000008030 elimination Effects 0.000 description 1
- 238000003379 elimination reaction Methods 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000000839 emulsion Substances 0.000 description 1
- 239000008393 encapsulating agent Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- 239000000284 extract Substances 0.000 description 1
- 210000003976 gap junction Anatomy 0.000 description 1
- 238000007429 general method Methods 0.000 description 1
- 230000002068 genetic effect Effects 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 230000006801 homologous recombination Effects 0.000 description 1
- 238000002744 homologous recombination Methods 0.000 description 1
- 210000005260 human cell Anatomy 0.000 description 1
- 150000003840 hydrochlorides Chemical class 0.000 description 1
- 230000005847 immunogenicity Effects 0.000 description 1
- 230000001861 immunosuppressant effect Effects 0.000 description 1
- 230000005764 inhibitory process Effects 0.000 description 1
- 238000011221 initial treatment Methods 0.000 description 1
- 230000000977 initiatory effect Effects 0.000 description 1
- 229910052500 inorganic mineral Inorganic materials 0.000 description 1
- 230000002452 interceptive effect Effects 0.000 description 1
- 108010084957 lecithin-retinol acyltransferase Proteins 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 230000029226 lipidation Effects 0.000 description 1
- 238000001638 lipofection Methods 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 239000006194 liquid suspension Substances 0.000 description 1
- 238000011068 loading method Methods 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 208000018769 loss of vision Diseases 0.000 description 1
- 231100000864 loss of vision Toxicity 0.000 description 1
- 229920002521 macromolecule Polymers 0.000 description 1
- 150000002690 malonic acid derivatives Chemical class 0.000 description 1
- 239000003550 marker Substances 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 239000012092 media component Substances 0.000 description 1
- 230000036564 melanin content Effects 0.000 description 1
- 244000005700 microbiome Species 0.000 description 1
- 238000000520 microinjection Methods 0.000 description 1
- 239000011707 mineral Substances 0.000 description 1
- 230000004048 modification Effects 0.000 description 1
- 238000012986 modification Methods 0.000 description 1
- 238000001823 molecular biology technique Methods 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 210000005036 nerve Anatomy 0.000 description 1
- 210000005157 neural retina Anatomy 0.000 description 1
- 230000000926 neurological effect Effects 0.000 description 1
- 239000003900 neurotrophic factor Substances 0.000 description 1
- 210000000056 organ Anatomy 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 230000004792 oxidative damage Effects 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- 230000000242 pagocytic effect Effects 0.000 description 1
- 230000008506 pathogenesis Effects 0.000 description 1
- 230000001717 pathogenic effect Effects 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 210000005047 peripherin Anatomy 0.000 description 1
- 239000008177 pharmaceutical agent Substances 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 210000000608 photoreceptor cell Anatomy 0.000 description 1
- 230000008488 polyadenylation Effects 0.000 description 1
- 210000001316 polygonal cell Anatomy 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 230000003449 preventive effect Effects 0.000 description 1
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 description 1
- 230000000069 prophylactic effect Effects 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 238000010188 recombinant method Methods 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 1
- 230000008439 repair process Effects 0.000 description 1
- 238000003571 reporter gene assay Methods 0.000 description 1
- 238000011160 research Methods 0.000 description 1
- 230000000452 restraining effect Effects 0.000 description 1
- 230000004243 retinal function Effects 0.000 description 1
- 230000001177 retroviral effect Effects 0.000 description 1
- 239000002336 ribonucleotide Substances 0.000 description 1
- 125000002652 ribonucleotide group Chemical group 0.000 description 1
- 101150018078 rpe gene Proteins 0.000 description 1
- 210000003786 sclera Anatomy 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 238000000527 sonication Methods 0.000 description 1
- 239000000600 sorbitol Substances 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 1
- 230000001629 suppression Effects 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 239000000725 suspension Substances 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 210000001578 tight junction Anatomy 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 238000012546 transfer Methods 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000014621 translational initiation Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 230000010415 tropism Effects 0.000 description 1
- 230000001173 tumoral effect Effects 0.000 description 1
- 238000011144 upstream manufacturing Methods 0.000 description 1
- 108700026220 vif Genes Proteins 0.000 description 1
- 230000004393 visual impairment Effects 0.000 description 1
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 1
- 230000003442 weekly effect Effects 0.000 description 1
- 238000009736 wetting Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
Classifications
-
- 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
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/005—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
- A61K48/0058—Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K48/00—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
- A61K48/0075—Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the delivery route, e.g. oral, subcutaneous
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
- C12N15/79—Vectors or expression systems specially adapted for eukaryotic hosts
- C12N15/85—Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
- C12N15/86—Viral vectors
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2750/00—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
- C12N2750/00011—Details
- C12N2750/14011—Parvoviridae
- C12N2750/14111—Dependovirus, e.g. adenoassociated viruses
- C12N2750/14141—Use of virus, viral particle or viral elements as a vector
- C12N2750/14143—Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2800/00—Nucleic acids vectors
- C12N2800/10—Plasmid DNA
- C12N2800/106—Plasmid DNA for vertebrates
- C12N2800/107—Plasmid DNA for vertebrates for mammalian
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2830/00—Vector systems having a special element relevant for transcription
- C12N2830/008—Vector systems having a special element relevant for transcription cell type or tissue specific enhancer/promoter combination
Definitions
- a NEW PROMOTER FOR RETINAL PIGMENT EPITHELIUM (RPE) TARGETED GENE THERAPY FIELD OF THE PRESENT INVENTION The present invention is in the field of medicine, in particular ophthalmology.
- BACKGROUND OF THE PRESENT INVENTION Retinal pigment epithelium (RPE) is formed from a single layer of regular polygonal cells arranged at the outermost layer of the retina. The outer side of the RPE is connected to Bruch’s membrane and the choroid, while the inner side is connected to the outer segment of photoreceptor cells. The outer side exhibits basal infolding, which increases cell surface area and facilitates substance exchange.
- the basement membrane is closely connected to the basal folds by half desmosomes located in the innermost layer of Bruch’s membrane.
- the inside of RPE cells harbors microvillous structures extending between photoreceptor outer segments (POS), which participate in the phagocytic function of the RPE.
- POS photoreceptor outer segments
- the tight junction formed between the single-layer RPE and the gap junction control the movement of substances and at the same time forms the choroid-blood-retinal barrier with Bruch’s membrane and choroid at the lateral retina.
- the RPE appears dark brown due to its melanin content, which reduces damage to the retina and internal nerves from ultraviolet light.
- the RPE also harbors a complex metabolic system that reduces excessive accumulation of reactive oxygen species (ROS) and consequent oxidative damage.
- ROS reactive oxygen species
- RPE structure and function are essential to normal vision, and alterations in the RPE can impair function and lead to retinopathy.
- retinitis pigmentosa RP
- AMD age-related macular degeneration
- SD Stargardt disease
- SLC16A8 gene encodes for a RPE-specific lactate transporter MCT3 and several risk alleles at the SLC16A8 locus for AMD.
- the promoter of said gene could represent an interesting candidate for restraining gene expression in RPE but its characterization has not been carried out.
- SUMMARY OF THE PRESENT INVENTION The present invention is defined by the claims.
- the present invention relates to a new promoter for Retinal pigment epithelium (RPE) targeted gene therapy.
- RPE Retinal pigment epithelium
- DETAILED DESCRIPTION OF THE PRESENT INVENTION Main definitions:
- polypeptide polypeptide
- peptide protein
- protein are used interchangeably herein to refer to polymers of amino acids of any length.
- the terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, phosphorylation, or conjugation with a labeling component.
- Polypeptides when discussed in the context of gene therapy refer to the respective intact polypeptide, or any fragment or genetically engineered derivative thereof, which retains the desired biochemical function of the intact protein.
- polynucleotide or “nucleic acid” refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides.
- this term includes, but is not limited to, single-, double- or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases, or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases.
- the backbone of the polynucleotide can comprise sugars and phosphate groups (as may typically be found in RNA or DNA), or modified or substituted sugar or phosphate groups.
- the backbone of the polynucleotide can comprise a polymer of synthetic subunits.
- the promoter of the present invention can be prepared by any method known to one skilled in the art, including chemical synthesis, recombination, and mutagenesis.
- the promoter of the present invention is a DNA molecule, typically synthesized by recombinant methods well known to those skilled in the art.
- the expression “derived from” refers to a process whereby a first component (e.g., a first polypeptide or polynucleotide), or information from that first component, is used to isolate, derive or make a different second component (e.g., a second polypeptide or polynucleotide that is different from the first one).
- the term "encoding" refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as, for example, a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
- a gene, cDNA, or RNA encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system.
- nucleic acid sequence encoding an amino acid sequence includes all nucleic acid sequences that are degenerate versions of each other and that encode the same amino acid sequence.
- nucleic acid sequence that encodes a protein or a RNA may also include introns to the extent that the nucleic acid sequence encoding the protein may in some version contain an intron(s).
- transgene refers to a polynucleotide that is introduced into the cells of a tissue or an organ and is capable of being expressed under appropriate conditions, or otherwise conferring a beneficial property to the cells.
- a transgene is selected based upon a desired therapeutic outcome.
- transgene product refers to any molecule that is encoded by a transgene and confers a beneficial property to the cells or a desired therapeutic outcome.
- the transgene product is a polypeptide.
- vector refers to the vehicle by which a polynucleotide can be introduced into a host cell, so as to transform the host and promote expression (e.g., transcription and translation) of the introduced sequence.
- viral vector encompasses vector DNA as well as viral particles generated thereof. Viral vectors can be replication-competent, or can be genetically disabled so as to be replication-defective or replication-impaired.
- replication-competent as used herein encompasses replication-selective and conditionally-replicative viral vectors which are engineered to replicate better or selectively in specific host cells (e.g. tumoral cells).
- non-viral vector notably refers to a vector of plasmid origin, and optionally such a vector combined with one or more substances improving the transfectional efficiency and/or the stability of said vector and/or the protection of said vector.
- promoter refers to a polynucleic acid sequence (such as, for example, a DNA sequence) recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleic acid sequence, thereby allowing the expression of a gene product operably linked to the promoter/regulatory sequence.
- this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
- the promoter sequence may, for example, be one which expresses the gene product in a tissue specific manner.
- promoter activity refers to the ability of a promoter to initiate transcription of a nucleic acid to which it is operably linked. Promoter activity can be measured using procedures known in the art or as described in the Examples. For example, promoter activity can be measured as an amount of mRNA transcribed by using, for example, Northern blotting or polymerase chain reaction (PCR).
- promoter activity can be measured as an amount of translated protein product, for example, by Western blotting, ELISA, colorimetric assays and various activity assays, including reporter gene assays and other procedures known in the art.
- operably linked or “transcriptional control” refers to functional linkage between a regulatory sequence and a heterologous polynucleic acid sequence resulting in expression of the latter.
- a first polynucleic acid sequence is operably linked with a second polynucleic acid sequence when the first polynucleic acid sequence is placed in a functional relationship with the second polynucleic acid sequence.
- a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence.
- Operably linked DNA sequences can be contiguous with each other and, e.g., where necessary to join two protein coding regions, are in the same reading frame.
- the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
- the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch algorithm (Needleman, Saul B. & Wunsch, Christian D. (1970). "A general method applicable to the search for similarities in the amino acid sequence of two proteins". Journal of Molecular Biology.48 (3): 443–53.).
- the percent identity between two nucleotide or amino acid sequences may also be determined using for example algorithms such as EMBOSS Needle (pair wise alignment; available at www.ebi.ac.uk).
- EMBOSS Needle may be used with a BLOSUM62 matrix, a “gap open penalty” of 10, a “gap extend penalty” of 0.5, a false “end gap penalty”, an “end gap open penalty” of 10 and an “end gap extend penalty” of 0.5.
- the “percent identity” is a function of the number of matching positions divided by the number of positions compared and multiplied by 100. For instance, if 6 out of 10 sequence positions are identical between the two compared sequences after alignment, then the identity is 60%. The % identity is typically determined over the whole length of the query sequence on which the analysis is performed. Two molecules having the same primary amino acid sequence or polynucleic acid sequence are identical irrespective of any chemical and/or biological modification.
- a first amino acid sequence having at least 80% of identity with a second amino acid sequence means that the first sequence has 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% of identity with the second amino acid sequence.
- RPE retina pigment epithelium
- RPE provides vital metabolic support to other retinal layers but is not directly involved in encoding visual stimuli into neurological signals, and is not responsive to light.
- RPE cells are darkly pigmented and absorb stray photons that would otherwise contribute to light scatter within the eye.
- gene therapy refers to the introduction of a polynucleotide into a cell's genome that restores, corrects, or modifies the gene and/or expression of the gene.
- RPE gene therapy refers to a gene therapy that is applied to the RPE, in particular for expressing a transgene product in RPE cells.
- the term “therapeutic gene” refers to a gene encoding a therapeutic protein which is useful in the treatment of a pathological condition.
- the therapeutic gene when expressed, confers a beneficial effect on the cell or tissue in which it is present, or on a patient in which the gene is expressed. Examples of beneficial effects include amelioration of a sign or symptom of a condition or disease, prevention or inhibition of a condition or disease, or conferral of a desired characteristic.
- Therapeutic genes include genes that partially or wholly correct a genetic deficiency in the patient.
- the therapeutic gene may be, without limitation, a nucleic acid sequence encoding a protein useful in gene therapy to relieve deficiencies caused by missing, defective or sub-optimal levels of said protein in a cell or tissue of a subject.
- the therapeutic polypeptide may, e.g., supply a polypeptide and/or enzymatic activity that is absent, defective or present at a sub-optimal level in RPE cells, supply a polypeptide and/or enzymatic activity that indirectly counteracts an imbalance in RPE cells.
- the therapeutic polypeptide may also be used to reduce the activity of a polypeptide by acting, e.g., as a dominant-negative polypeptide.
- the therapeutic polypeptide supplies a polypeptide and/or enzymatic activity that is absent, defective or present at a sub-optimal level in RPE cells, more typically a polypeptide and/or enzymatic activity that is absent or defective in RPE cells.
- the term “expression cassette” refers to a nucleic acid construct comprising a coding sequence and one or more control sequences required for expression of said coding sequence.
- one of these control sequence is the promoter of the present invention that has a promoter activity in RPE.
- the expression cassette comprises a coding sequence and regulatory sequences preceding (5′ non-coding sequences) and following (3′ non- coding sequences) the coding sequence that are required for expression of the selected gene product.
- an expression cassette typically comprises a promoter sequence, a coding sequence and a 3′ untranslated region that usually contains a polyadenylation site and/or transcription terminator.
- the expression cassette may also comprise additional regulatory elements such as, for example, enhancer sequences, a polylinker sequence facilitating the insertion of a DNA fragment within a vector and/or splicing signal sequences.
- the expression cassette is usually included within a vector, to facilitate cloning and transformation.
- transformation means the introduction of a "foreign” (i.e., extrinsic or extracellular) gene, DNA or RNA sequence to a host cell, so that the host cell will express the introduced gene or sequence to produce a desired substance, typically a protein or enzyme coded by the introduced gene or sequence.
- a host cell that receives and expresses introduced DNA or RNA bas been "transformed".
- treatment refers to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse.
- the treatment may be administered to a patient having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a patient beyond that expected in the absence of such treatment.
- therapeutic regimen is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy.
- a therapeutic regimen may include an induction regimen and a maintenance regimen.
- the phrase “induction regimen” or “induction period” refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease.
- the general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen.
- An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both.
- maintenance regimen refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years).
- a maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular interval, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or treatment upon achievement of a particular predetermined criteria [e.g., pain, disease manifestation, etc.]).
- the term “therapeutic level” refers to the amount of a transgene product or the level of activity of a transgene product sufficient to confer its therapeutic or beneficial effect(s) in the host receiving the transgene. Expression levels of the transgene or the levels of activity of the transgene product can be measured at the protein or the mRNA level using methods known in the art. As used herein, the term “therapeutically efficient amount” is intended an amount of pharmaceutical composition of the present invention administered to a subject that is sufficient to constitute a treatment as defined above of ocular disease.
- the first object of the present invention relates to a polynucleotide having a promoter activity in retinal pigment epithelium (RPE) having a nucleic acid sequence having at least 80% of identity with the nucleic acid sequence as set forth in SEQ ID NO:1.
- RPE retinal pigment epithelium
- the promoter of the present invention thus exhibits a promoter activity in RPE, i.e. when introduced in RPE, it can initiate transcription of a nucleic acid to which it is operably linked.
- the promoter activity is specific of RPE cells.
- the term “specific of RPE cells” shall be understood to mean a promoter mainly active in RPE cells. It shall be understood that a residual expression, generally lower, in other tissues or cells cannot be entirely excluded.
- the promoter of the present invention is not active in Muller cells or in photoreceptors.
- the promoter of the present invention comprises, or consists of, the sequence of SEQ ID NO: 1.
- the promoter of the present invention comprises, or consists of, a functional variant of SEQ ID NO: 1.
- the term “variant” refers to a nucleic acid sequence differing from the original sequence, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the original polynucleotide. The sequence of the variant may differ by nucleotide substitutions, deletions or insertions of one or more nucleotides in the sequence, which do not impair the promoter activity. The variant may have the same length of the original sequence, or may be shorter or longer.
- the term “functional variant” refers to a variant of SEQ ID NO: 1 that exhibits a promoter activity of SEQ ID NO: 1, i.e. that exhibits a promoter activity in RPE cells, typically a promoter activity specific of RPE cells.
- the promoter of the present invention comprises, or consists of, a functional variant having at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% identity to SEQ ID NO: 1, typically over the entire sequence of SEQ ID NO: 1.
- the promoter of the present invention may differ from the polynucleotide of SEQ ID NO: 1 by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 substitutions, deletions and/or insertions.
- the promoter of the present invention comprises, or consists of, a functional variant having a sequence capable of hybridizing under low, medium or high stringency conditions with the nucleic acid sequence of SEQ ID NO: 1 or its complementary strand, typically under medium stringency conditions, more typically under high stringency conditions.
- the term “low stringency conditions” means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C.
- the carrier material is finally washed three times each for 15 minutes using 2 ⁇ SSC, 0.2% SDS at 50°C.
- medium stringency conditions means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C. in 5 ⁇ SSPE, 0.3% SDS, 200 micrograms/mL sheared and denatured salmon sperm DNA, and 35% formamide, following standard Southern blotting procedures for 12 to 24 hours.
- the carrier material is finally washed three times each for 15 minutes using 2 ⁇ SSC, 0.2% SDS at 55°C.
- high stringency conditions means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C. in 5 ⁇ SSPE, 0.3% SDS, 200 micrograms/mL sheared and denatured salmon sperm DNA, and 50% formamide, following standard Southern blotting procedures for 12 to 24 hours.
- the carrier material is finally washed three times each for 15 minutes using 2 ⁇ SSC, 0.2% SDS at 65°C.
- a further object of the present invention relates to an expression cassette comprising a polynucleotide of the present invention operably linked to a polynucleotide of interest.
- the nucleic acid operably linked to the promoter of the present invention is a transgene that encodes a polypeptide of interest or a polynucleotide of interest.
- the promoter of the present invention is operably linked to a heterologous polynucleotide.
- heterologous means a nucleic acid other than the nucleic acid that the promoter is operably linked to in a naturally occurring genome.
- the promoter of the present invention is not operably linked to SLC16A8 gene, and in particular to the human SLC16A8 gene.
- the polynucleotide operably linked to the promoter of the present invention encodes a polypeptide of interest.
- the polypeptide of interest may be any polypeptide of which expression in RPE cells is desired.
- the polypeptide of interest may be a therapeutic polypeptide, or reporter protein.
- the nucleic acid operably linked to the promoter of the present invention is a therapeutic gene.
- therapeutic genes include, but are not limited to, nucleic acids for replacement of a missing or mutated gene known to cause retinal disease such as ARSG, BEST1, DFNB31, IQCB1, KCNJ13, LCA5, LRAT, MERTK, RGR, RPE65, SPATA7.
- the therapeutic gene may also encode neurotrophic factors such as GDNF (Gene ID: 2668), CNTF (Gene ID: 1270), FGF2 (Gene ID: 2247), BDNF (Gene ID: 627) and EPO (Gene ID: 2056), anti-apoptotic genes such as BCL2 (Gene ID: 596) and BCL2L1 (Gene ID: 598), anti-angiogenic factors such as endostatin, angiostatin and sFlt, anti-inflammatory factors such as IL10 (Gene ID: 3586), IL1R1 (Gene ID: 3554), TGFBI (Gene ID; 7045) and IL4 (Gene ID: 3565), or nucleoredoxin-like 1 the rod-derived cone viability factor (RdCVF) and RdCVFL (Gene ID: 115861).
- neurotrophic factors such as GDNF (Gene ID: 2668), CNTF (Gene ID: 1270), FGF2 (Gene ID:
- the transgene product of interest is an endonuclease that provides for site-specific knock-down of gene function, e.g., where the endonuclease knocks out an allele associated with a RPE disease.
- a dominant allele encodes a defective copy of a gene that, when wild-type, is a retinal structural protein and/or provides for normal retinal function
- a site-specific endonuclease can be targeted to the defective allele and knock out the defective allele.
- a site-specific nuclease can also be used to stimulate homologous recombination with a donor DNA that encodes a functional copy of the protein encoded by the defective allele.
- the method of the invention can be used to deliver both a site-specific endonuclease that knocks out a defective allele, and can be used to deliver a functional copy of the defective allele, resulting in repair of the defective allele, thereby providing for production of a functional retinal protein (e.g., functional retinoschisin, functional RPE65, functional peripherin, etc.). See, e.g., Li et al. (2011) Nature 475:217.
- the DNA targeting endonuclease of the present invention is a TALEN. In some embodiments, the DNA targeting endonuclease of the present invention is a ZFN. In some embodiments, the DNA targeting endonuclease of the present invention is a CRISPR-associated endonuclease. In some embodiments, the CRISPR-associated endonuclease is a Cas9 nuclease. In some embodiments, the CRISPR-associated endonuclease is a Cpf1 nuclease. In some embodiments, the polynucleotide operably linked to the promoter of the present invention encodes a polynucleotide of interest.
- the polynucleotide of interest may be any nucleic acid of which expression in RPE cells is desired.
- the polynucleotide of interest may be a therapeutic nucleic acid.
- the nucleic acid may be, for example, an siRNA, an shRNA an RNAi, a miRNA, an antisense RNA, a ribozyme or a DNAzyme.
- the nucleic acid encodes an RNA that when transcribed from the nucleic acid operably linked to the promoter of the present invention can treat or prevent a retinal pigment epithelial disease by interfering with translation or transcription of an abnormal or excess protein associated with said disorder.
- the polynucleotide of interest may encode for an RNA, which treats the disease by highly specific elimination or reduction of mRNA encoding the abnormal and/or excess proteins.
- the expression cassette of the present invention may comprise one or more nucleic acids operably linked to the promoter of the present invention.
- the promoter may be operably linked to one or more therapeutic genes and a nucleic acid encoding a reporter protein or to a therapeutic gene.
- Vectors of the present invention A further object of the present invention relates to a vector comprising the promoter of the present invention or the expression cassette of the present invention.
- the vector of the present invention is a vector suitable for use in gene therapy, and in particular is suitable to target RPE cells.
- the vector of the present invention is typically a viral genome vector including any element required to establish the expression of the polypeptide of interest in a host cell such as, for example, a promoter, e.g., a polynucleotide of the present invention, an ITR, a ribosome binding element, terminator, enhancer, selection marker, intron, polyA signal, and/or origin of replication.
- a promoter e.g., a polynucleotide of the present invention
- an ITR e.g., an ITR, a ribosome binding element, terminator, enhancer, selection marker, intron, polyA signal, and/or origin of replication.
- the vector is a viral vector, such as vectors derived from Moloney murine leukemia virus vectors (MoMLV), MSCV, SFFV, MPSV or SNV, lentiviral vectors (e.g.
- HIV human immunodeficiency virus
- SIV simian immunodeficiency virus
- FV feline immunodeficiency virus
- BIV bovine immunodeficiency virus
- EIAV equine infectious anemia virus
- adenoviral (Ad) vectors adeno-associated viral (AAV) vectors
- AAV adeno-associated viral vectors
- SV-40 simian virus 40 vectors
- bovine papilloma virus vectors Epstein-Ban virus
- herpes virus vectors vaccinia virus vectors
- Harvey murine sarcoma virus vectors murine mammary tumor virus vectors
- Rous sarcoma virus vectors Rous sarcoma virus vectors.
- the vector is a retroviral vector, typically a lentiviral vector or a non- pathogenic parvovirus.
- suitable sequences should be introduced in the vector of the present invention for obtaining a functional viral vector, such as AAV ITRs for an AAV vector, or LTRs for lentiviral vectors.
- the vector is an AAV vector.
- AAV vector refers to a polynucleotide vector comprising one or more heterologous sequences (i.e., nucleic acid sequence not of AAV origin) that are flanked by at least one AAV inverted terminal repeat sequence (ITR), typically two ITRs.
- AAV vectors can be replicated and packaged into infectious viral particles when present in a host cell that has been infected with a suitable helper virus (or that is expressing suitable helper functions) and that is expressing AAV rep and cap gene products (i.e. AAV Rep and Cap proteins).
- An “inverted terminal repeat” or “ITR” sequence is a term well understood in the art and refers to relatively short sequences found at the termini of viral genomes which are in opposite orientation.
- An “AAV inverted terminal repeat (ITR)” sequence is an approximately 145-nucleic acid sequence that is present at both termini of the native single-stranded AAV genome.
- the outermost 125 nucleotides of the ITR can be present in either of two alternative orientations, leading to heterogeneity between different AAV genomes and between the two ends of a single AAV genome.
- the outermost 125 nucleotides also contain several shorter regions of self-complementarity (designated A, A′, B, B′, C, C and D regions), allowing intra-strand base-pairing to occur within this portion of the ITR.
- AAV ITRs for use in the vectors of the present invention may have a wild-type nucleic acid sequence or may be altered by the insertion, deletion or substitution.
- the serotype of the inverted terminal repeats (ITRs) of the AAV vector may be selected from any known human or nonhuman AAV serotype.
- the promoter or expression cassette of the present invention may be introduced into the vector by any method known by the skilled person.
- the vector of the present invention may be packaged into a virus capsid to generate a “viral particle”.
- the present invention also relates to a viral particle comprising a vector of the present invention.
- the vector is an AAV vector and is packaged into an AAV-derived capsid to generate an “adeno-associated viral particle” or “AAV particle”.
- AAV particle refers to a viral particle composed of at least one AAV capsid protein and an encapsidated AAV vector genome.
- the capsid serotype determines the tropism range of the AAV particle.
- AAV adeno-associated virus
- human serotype 2 was the first AAV developed as a gene transfer vector.
- Other currently used AAV serotypes include, but are not limited to, AAV1, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAVrh74 and AAVdj, etc.
- non-natural engineered variants and chimeric AAV can also be useful.
- the capsid proteins may be variants comprising one or more amino acid substitutions enhancing transduction efficiency.
- Different AAV serotypes are used to optimize transduction of particular target cells or to target specific cell types within a particular target tissue (e.g., RPE cells).
- An AAV particle can comprise viral proteins and viral nucleic acids of the same serotype or any natural or artificial sequence variant of AAV.
- the AAV particle may comprise AAV2 capsid proteins and at least one, typically two, AAV2 ITR. Any combination of AAV serotypes for production of an AAV particle is provided herein as if each combination had been expressly stated herein.
- the AAV particle comprises an AAV-derived capsid selected from the group consisting of AAV2, AAV-5, AAV-7m8 (AAV2-7m8, Dalkara et al. Sci Transl Med (2013), 5, 189ra76), AAV9 or AAV8 capsid.
- AAV viruses may be engineered using conventional molecular biology techniques, making it possible to optimize these particles for cell specific delivery of nucleic acid sequences, for minimizing immunogenicity, for tuning stability and particle lifetime, for efficient degradation, for accurate delivery to the nucleus.
- artificial AAV serotypes may be used in the context of the present invention, including, without limitation, AAV with a non-naturally occurring capsid protein.
- Such an artificial capsid may be generated by any suitable technique, using a selected AAV sequence (e.g., a fragment of a VP1 capsid protein) in combination with heterologous sequences which may be obtained from a different selected AAV serotype, non- contiguous portions of the same AAV serotype, from a non-AAV viral source, or from a non- viral source.
- AAV serotype may be, without limitation, a chimeric AAV capsid or a mutated AAV capsid.
- AAV particles Numerous methods are known in the art for production of viral particles, and in particular AAV particles, including transfection, stable cell line production, and infectious hybrid virus production systems which include adenovirus-AAV hybrids, herpesvirus-AAV hybrids (Conway, J E et al., (1997) Virology 71(11):8780-8789) and baculovirus-AAV hybrids).
- AAV production cultures for the production of AAV virus particles all require; 1) suitable host cells, including, for example, human-derived cell lines such as HeLa, A549, or 293 cells, or insect- derived cell lines such as SF-9, in the case of baculovirus production systems; 2) suitable helper virus function, provided by wild-type or mutant adenovirus (such as temperature sensitive adenovirus), herpes virus, baculovirus, or a plasmid construct providing helper functions; 3) AAV rep and cap genes and gene products; 4) a polynucleotide of interest flanked by at least one AAV ITR sequences, e.g., a vector of the present invention; and 5) suitable media and media components to support AAV production that are well-known in the art.
- suitable host cells including, for example, human-derived cell lines such as HeLa, A549, or 293 cells, or insect- derived cell lines such as SF-9, in the case of baculovirus production systems
- host cells for producing AAV particles include mammalian cells, insect cells, plant cells, microorganisms and yeast.
- Host cells can also be packaging cells in which the AAV rep and cap genes are stably maintained in the host cell or producer cells in which the AAV vector genome is stably maintained.
- Exemplary packaging and producer cells are derived from 293, A549 or HeLa cells.
- AAV particles are then purified and formulated using standard techniques known in the art.
- Host cells of the present invention A further object of the present invention to an isolated host cell transformed or transfected with an expression cassette, vector or viral particle of the present invention.
- the host cell may be any animal cell, plant cell, bacterium cell or yeast. Typically, the host cell is a mammalian cell or an insect cell.
- the host cell is a human cell.
- the host cell is a RPE cell, in particular a human RPE cell.
- the expression cassette or vector of the present invention may be transferred into host cells using any known technique including, but being not limited to, calcium phosphate-DNA precipitation, DEAE-Dextran transfection, electroporation, microinjection, biolistic, lipofection, or viral infection, and may be maintained in the host cell in an ectopic form or may be integrated into the genome.
- the expression cassette or vector of the present invention is transferred into the host cell by viral infection, typically using a viral particle of the present invention, more typically using an AAV particle of the present invention.
- compositions of the present invention also relates to a pharmaceutical composition comprising an expression cassette, vector, viral particle or cell of the present invention.
- Such compositions comprise a therapeutically effective amount of the therapeutic agent (an expression cassette, vector, viral particle or cell of the present invention), and a pharmaceutically acceptable excipient.
- pharmaceutically acceptable means approved by a regulatory agency or recognized pharmacopeia such as European Pharmacopeia, for use in animals and/or humans.
- excipient refers to a diluent, adjuvant, carrier, or vehicle with which the therapeutic agent is administered.
- excipients are relatively inert substances that facilitate administration of a pharmacologically effective substance and can be supplied as liquid solutions or suspensions, as emulsions, or as solid forms suitable for dissolution or suspension in liquid prior to use.
- an excipient can give form or consistency, or act as a diluent.
- Suitable excipients include but are not limited to stabilizing agents, wetting and emulsifying agents, salts for varying osmolality, encapsulating agents, pH buffering substances, and buffers.
- excipients include any pharmaceutical agent suitable for direct delivery to the eye which may be administered without undue toxicity.
- Pharmaceutically acceptable excipients include, but are not limited to, sorbitol, any of the various tween compounds, and liquids such as water, saline, glycerol and ethanol.
- Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
- mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
- organic acids such as acetates, propionates, malonates, benzoates, and the like.
- the composition is formulated to be administered to the eye, in particular by intraocular injection, e.g., by subretinal and/or intravitreal administration.
- the composition can be combined with pharmaceutically acceptable excipient such as saline, Ringer's balanced salt solution (pH 7.4), and the like.
- the pharmaceutical compositions described herein can be packaged in single unit dosages or in multidosage forms.
- the pharmaceutical composition comprises a vector or viral particle of the present invention, more typically an AAV vector or particle.
- the pharmaceutical composition comprises host cells of the present invention, typically human host cell of the present invention, i.e. transformed or transfected with an expression cassette, vector or viral particle of the present invention, typically with an AAV particle.
- the composition comprising host cells may be frozen for storage at any temperature appropriate for storage of the cells.
- the cells may be frozen at about ⁇ 20° C., ⁇ 80°C. or any other appropriate temperature.
- Cryogenically frozen cells may be stored in appropriate containers and prepared for storage to reduce risk of cell damage and maximize the likelihood that the cells will survive thawing.
- the cells may also be maintained at room temperature of refrigerated, e.g. at about 4°C.
- the amount of pharmaceutical composition to be administered may be determined by standard procedure well known by those of ordinary skill in the art. Physiological data of the patient (e.g. age, size, and weight) and type and severity of the disease being treated have to be taken into account to determine the appropriate dosage.
- the pharmaceutical composition of the present invention may be administered as a single dose or in multiple doses.
- the composition comprises viral particles of the present invention and each unit dosage comprises from 108 to 1013 viral particles, typically from 109 to 1012 particles.
- the pharmaceutical composition may further comprise one or several additional active compounds such as corticosteroids, antibiotics, analgesics, immunosuppressants, trophic factors, or any combinations thereof.
- Methods of therapy of the present invention A further object of the present invention relates to a method of therapy in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an expression cassette, a vector, a viral particle, a host cell or a pharmaceutical composition of the present invention.
- the method of the present invention is particularly suitable for the treatment of an ocular disease.
- the method of the present invention is particularly suitable for the treatment of a retinal pigment epithelial disease.
- retinal pigment epithelial diseases include, but are not limited to age-related macular degeneration, Leber's hereditary optic neuropathy, cone-rod dystrophy, Leber congenital amaurosis, Stargardt's disease, diabetic retinopathy, retinal detachment, Best's disease, retinitis pigmentosa, choroideremia and a tapetoretinal degeneration.
- the ocular disease is selected from disease which are not necessarily or specifically associated with RPE, but which can be treated or prevented by expressing specifically nucleic acid encoding a polypeptide or polynucleotide of interest in RPE cell.
- the pharmaceutical composition of the present invention is typically administered intraocularly, more typically by subretinal or intravitreal administration.
- the method of the present invention may also further comprise administering at least one additional therapeutic agent to the subject.
- said therapeutic agent may be selected from the group consisting of a corticosteroid, an antibiotic, an analgesic, an immunosuppressant, or a trophic factor, or any combinations thereof.
- composition of the present invention may be administered before or after the disease becomes symptomatic, e.g., before or after partial or complete RPE degeneration and/or before or after partial or complete loss of vision.
- the invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
- EXAMPLE To study the promoter of the SLC16A8 gene, we first needed to identify the promoter region of the gene and hypothesized that the genomic sequence between the transcription start site (TSS) of SLC16A8 and the end of the BAIAP2L2 gene located immediately on 5' end (Chr22: 38,083,143-38,084,902) might contain the promoter region.
- TSS transcription start site
- the digestion/ligation process was used to remove the CMV/CBA promoter from the recipient plasmid to introduce our candidate promoter region instead.
- rAAV recombinant adeno-associated viruses
- BALB/cJ strain mice were selected as the experimental model. Knowing that the efficiency of transduction in the retina by an adeno- associated vector serotype 9 (AAV9) following systemic injection has been demonstrated previously, we generated an AAV9 vector to assess the ability of the SLC16A81.8 region(AAV2.9-SLC16A81.8-eGFP) to direct GFP expression in vivo in RPE cells.
- SLC16A81.8 a RPE-restricted fluorescent signal after systemic injection of AAV2.9-SLC16A81.8-eGFP (“SLC16A81.8”) would imply that the 1.8 kb fragment contains the entire promoter with its regulatory elements.
- Viruses were administered systemically by intracardiac injection in 3- to 4-day-old wt/wt BALB/cJ mice (D3-J4 ) to study in vivo GFP expression. Intracardiac injection of CMV/CBA- led to very strong GFP expression, confirming the correct performance of the technical procedure.
- SLC16A81.8 did not show a green fluorescent signal at the macroscopic scale.
- mice injected with SLC16A81.8 expressed GFP
- GFP expression is very high in both the RPE/choroid and retinas of CMV/CBV-injected mice.
- mice injected with SLC16A81.8 express low levels of GFP in the RPE/choroid.
- the absence or low expression of GFP in the RPE of mice injected with SLC16A81.8 led us to question whether the TSS reported in the UCSC database corresponded to a minor TSS.
- SLC16A8 expression is restricted to the RPE, a tissue that is not among those analysed by ENCODE.
- Results from the study of the NXNL1 and NXNL2 genes show that there are several TSSs.
- AAV2.9-5'585BEST1-eGFP – “5'BEST1”) for use as a specific control for expression by RPE.
- AAV2.9-5'585BEST1- eGFP shows a GFP signal as green fluorescent dots.
- the specific expression of 5'BEST1 by the RPE suggests that these dots represent expression of the GFP reporter protein under the control of the active promoter in the RPE and not in the retina.
- mice injected with the SLC16A82.1vector show intense fluorescent dots associated with more diffuse labeling confirming the presence of regulatory elements in the SLC16A82.1 vector.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Engineering & Computer Science (AREA)
- Genetics & Genomics (AREA)
- Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Molecular Biology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- Animal Behavior & Ethology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Biomedical Technology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- Virology (AREA)
- Biophysics (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Ophthalmology & Optometry (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Retinal pigment epithelium (RPE) structure and function are essential to normal vision, and alterations in the RPE can impair function and lead to retinopathy. Gene therapy is currently investigated for the treatment of retinal pigment epithelial diseases but there is a need for identifying promoter polynucleotides that will allow specific expression of polynucleotides of interest in said tissue. The SLC16A8 gene encodes for a RPE-specific lactate transporter MCT3. The inventors aimed at defining the sequence sufficient to drive specific expression of SLC16A8 in the RPE. To identify the promoter region of SLC16A8, different candidate fragments were then cloned into plasmids expressing a reporter gene to test the ability of each of these regions to activate the expression of this reporter. They also chose to exploit the restricted expression of SLC16A8 and to perform this experiment by a new in vivo approach using recombinant adenovirus associated virus (rAAV). By combining the methods, the inventors succeed in identifying the promoter region of SLC16A8.
Description
A NEW PROMOTER FOR RETINAL PIGMENT EPITHELIUM (RPE) TARGETED GENE THERAPY FIELD OF THE PRESENT INVENTION: The present invention is in the field of medicine, in particular ophthalmology. BACKGROUND OF THE PRESENT INVENTION: Retinal pigment epithelium (RPE) is formed from a single layer of regular polygonal cells arranged at the outermost layer of the retina. The outer side of the RPE is connected to Bruch’s membrane and the choroid, while the inner side is connected to the outer segment of photoreceptor cells. The outer side exhibits basal infolding, which increases cell surface area and facilitates substance exchange. The basement membrane is closely connected to the basal folds by half desmosomes located in the innermost layer of Bruch’s membrane. The inside of RPE cells harbors microvillous structures extending between photoreceptor outer segments (POS), which participate in the phagocytic function of the RPE. The tight junction formed between the single-layer RPE and the gap junction control the movement of substances and at the same time forms the choroid-blood-retinal barrier with Bruch’s membrane and choroid at the lateral retina. The RPE appears dark brown due to its melanin content, which reduces damage to the retina and internal nerves from ultraviolet light. The RPE also harbors a complex metabolic system that reduces excessive accumulation of reactive oxygen species (ROS) and consequent oxidative damage. Therefore, RPE structure and function are essential to normal vision, and alterations in the RPE can impair function and lead to retinopathy. For example, retinitis pigmentosa (RP), age-related macular degeneration (AMD), and Stargardt disease (SD) are degenerative retinal diseases in which RPE dysfunction has been implicated in their pathogenesis. Gene therapy is currently investigated for the treatment of retinal pigment epithelial diseases but there is a need for identifying promoter polynucleotides that will allow specific expression of polynucleotides of interest in said tissue. The SLC16A8 gene encodes for a RPE-specific lactate transporter MCT3 and several risk alleles at the SLC16A8 locus for AMD. The promoter of said gene could represent an interesting candidate for restraining gene expression in RPE but its characterization has not been carried out. SUMMARY OF THE PRESENT INVENTION:
The present invention is defined by the claims. In particular, the present invention relates to a new promoter for Retinal pigment epithelium (RPE) targeted gene therapy. DETAILED DESCRIPTION OF THE PRESENT INVENTION: Main definitions: As used herein, the terms “polypeptide”, “peptide”, and “protein” are used interchangeably herein to refer to polymers of amino acids of any length. The terms also encompass an amino acid polymer that has been modified; for example, disulfide bond formation, glycosylation, lipidation, phosphorylation, or conjugation with a labeling component. Polypeptides when discussed in the context of gene therapy refer to the respective intact polypeptide, or any fragment or genetically engineered derivative thereof, which retains the desired biochemical function of the intact protein. As used herein, the term “polynucleotide” or “nucleic acid” refers to a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. Thus, this term includes, but is not limited to, single-, double- or multi-stranded DNA or RNA, genomic DNA, cDNA, DNA-RNA hybrids, or a polymer comprising purine and pyrimidine bases, or other natural, chemically or biochemically modified, non-natural, or derivatized nucleotide bases. The backbone of the polynucleotide can comprise sugars and phosphate groups (as may typically be found in RNA or DNA), or modified or substituted sugar or phosphate groups. Alternatively, the backbone of the polynucleotide can comprise a polymer of synthetic subunits. The promoter of the present invention can be prepared by any method known to one skilled in the art, including chemical synthesis, recombination, and mutagenesis. In particular, the promoter of the present invention is a DNA molecule, typically synthesized by recombinant methods well known to those skilled in the art. As used herein, the expression “derived from” refers to a process whereby a first component (e.g., a first polypeptide or polynucleotide), or information from that first component, is used to isolate, derive or make a different second component (e.g., a second polypeptide or polynucleotide that is different from the first one).
As used herein, the term "encoding" refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as, for example, a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (e.g., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene, cDNA, or RNA, encodes a protein if transcription and translation of mRNA corresponding to that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleic acid sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that gene or cDNA. Unless otherwise specified, a "nucleic acid sequence encoding an amino acid sequence" includes all nucleic acid sequences that are degenerate versions of each other and that encode the same amino acid sequence. The phrase “nucleic acid sequence that encodes a protein or a RNA” may also include introns to the extent that the nucleic acid sequence encoding the protein may in some version contain an intron(s). As used herein, the term “transgene” refers to a polynucleotide that is introduced into the cells of a tissue or an organ and is capable of being expressed under appropriate conditions, or otherwise conferring a beneficial property to the cells. A transgene is selected based upon a desired therapeutic outcome. As used herein, the term “transgene product” refers to any molecule that is encoded by a transgene and confers a beneficial property to the cells or a desired therapeutic outcome. Typically, the transgene product is a polypeptide. As used herein, the terms "vector" refers to the vehicle by which a polynucleotide can be introduced into a host cell, so as to transform the host and promote expression (e.g., transcription and translation) of the introduced sequence. As used herein, the term “viral vector” encompasses vector DNA as well as viral particles generated thereof. Viral vectors can be replication-competent, or can be genetically disabled so as to be replication-defective or replication-impaired. The term “replication-competent” as used herein encompasses replication-selective and conditionally-replicative viral vectors which are engineered to replicate better or selectively in specific host cells (e.g. tumoral cells). As used herein, the term “non-viral vector” notably refers to a vector of plasmid origin, and optionally such a vector combined with one or more substances improving the transfectional efficiency and/or the stability of said vector and/or the protection of said vector.
As used herein, the term “promoter” refers to a polynucleic acid sequence (such as, for example, a DNA sequence) recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a polynucleic acid sequence, thereby allowing the expression of a gene product operably linked to the promoter/regulatory sequence. In particular, this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product. The promoter sequence may, for example, be one which expresses the gene product in a tissue specific manner. As used herein, the term “promoter activity” refers to the ability of a promoter to initiate transcription of a nucleic acid to which it is operably linked. Promoter activity can be measured using procedures known in the art or as described in the Examples. For example, promoter activity can be measured as an amount of mRNA transcribed by using, for example, Northern blotting or polymerase chain reaction (PCR). Alternatively, promoter activity can be measured as an amount of translated protein product, for example, by Western blotting, ELISA, colorimetric assays and various activity assays, including reporter gene assays and other procedures known in the art. As used herein, the term "operably linked" or "transcriptional control" refers to functional linkage between a regulatory sequence and a heterologous polynucleic acid sequence resulting in expression of the latter. For example, a first polynucleic acid sequence is operably linked with a second polynucleic acid sequence when the first polynucleic acid sequence is placed in a functional relationship with the second polynucleic acid sequence. For instance, a promoter is operably linked to a coding sequence if the promoter affects the transcription or expression of the coding sequence. Operably linked DNA sequences can be contiguous with each other and, e.g., where necessary to join two protein coding regions, are in the same reading frame. As used herein, the “percent identity” between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = number of identical positions/total number of positions x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below. The percent identity between two amino acid sequences can be determined using the Needleman and Wunsch
algorithm (Needleman, Saul B. & Wunsch, Christian D. (1970). "A general method applicable to the search for similarities in the amino acid sequence of two proteins". Journal of Molecular Biology.48 (3): 443–53.). The percent identity between two nucleotide or amino acid sequences may also be determined using for example algorithms such as EMBOSS Needle (pair wise alignment; available at www.ebi.ac.uk). For example, EMBOSS Needle may be used with a BLOSUM62 matrix, a “gap open penalty” of 10, a “gap extend penalty” of 0.5, a false “end gap penalty”, an “end gap open penalty” of 10 and an “end gap extend penalty” of 0.5. In general, the “percent identity” is a function of the number of matching positions divided by the number of positions compared and multiplied by 100. For instance, if 6 out of 10 sequence positions are identical between the two compared sequences after alignment, then the identity is 60%. The % identity is typically determined over the whole length of the query sequence on which the analysis is performed. Two molecules having the same primary amino acid sequence or polynucleic acid sequence are identical irrespective of any chemical and/or biological modification. According to the invention a first amino acid sequence having at least 80% of identity with a second amino acid sequence means that the first sequence has 80; 81; 82; 83; 84; 85; 86; 87; 88; 89; 90; 91; 92; 93; 94; 95; 96; 97; 98; 99 or 100% of identity with the second amino acid sequence. As used herein, the term “retinal pigment epithelium” or “RPE” has its general meaning in the art and refers to the pigmented cell layer just outside the neurosensory retina that nourishes retinal visual cells, and that is firmly attached to the underlying choroid and overlying retinal visual cells. RPE provides vital metabolic support to other retinal layers but is not directly involved in encoding visual stimuli into neurological signals, and is not responsive to light. RPE cells are darkly pigmented and absorb stray photons that would otherwise contribute to light scatter within the eye. As used herein, the term "gene therapy" refers to the introduction of a polynucleotide into a cell's genome that restores, corrects, or modifies the gene and/or expression of the gene. Thus the term “RPE gene therapy” refers to a gene therapy that is applied to the RPE, in particular for expressing a transgene product in RPE cells. As used herein, the term “therapeutic gene” refers to a gene encoding a therapeutic protein which is useful in the treatment of a pathological condition. The therapeutic gene, when expressed, confers a beneficial effect on the cell or tissue in which it is present, or on a patient
in which the gene is expressed. Examples of beneficial effects include amelioration of a sign or symptom of a condition or disease, prevention or inhibition of a condition or disease, or conferral of a desired characteristic. Therapeutic genes include genes that partially or wholly correct a genetic deficiency in the patient. In particular, the therapeutic gene may be, without limitation, a nucleic acid sequence encoding a protein useful in gene therapy to relieve deficiencies caused by missing, defective or sub-optimal levels of said protein in a cell or tissue of a subject. The therapeutic polypeptide may, e.g., supply a polypeptide and/or enzymatic activity that is absent, defective or present at a sub-optimal level in RPE cells, supply a polypeptide and/or enzymatic activity that indirectly counteracts an imbalance in RPE cells. The therapeutic polypeptide may also be used to reduce the activity of a polypeptide by acting, e.g., as a dominant-negative polypeptide. Typically, the therapeutic polypeptide supplies a polypeptide and/or enzymatic activity that is absent, defective or present at a sub-optimal level in RPE cells, more typically a polypeptide and/or enzymatic activity that is absent or defective in RPE cells. As used herein, the term “expression cassette” refers to a nucleic acid construct comprising a coding sequence and one or more control sequences required for expression of said coding sequence. In particular, one of these control sequence is the promoter of the present invention that has a promoter activity in RPE. Generally, the expression cassette comprises a coding sequence and regulatory sequences preceding (5′ non-coding sequences) and following (3′ non- coding sequences) the coding sequence that are required for expression of the selected gene product. Thus, an expression cassette typically comprises a promoter sequence, a coding sequence and a 3′ untranslated region that usually contains a polyadenylation site and/or transcription terminator. The expression cassette may also comprise additional regulatory elements such as, for example, enhancer sequences, a polylinker sequence facilitating the insertion of a DNA fragment within a vector and/or splicing signal sequences. The expression cassette is usually included within a vector, to facilitate cloning and transformation. As used herein, the term "transformation" means the introduction of a "foreign" (i.e., extrinsic or extracellular) gene, DNA or RNA sequence to a host cell, so that the host cell will express the introduced gene or sequence to produce a desired substance, typically a protein or enzyme coded by the introduced gene or sequence. A host cell that receives and expresses introduced DNA or RNA bas been "transformed".
As used herein, the term "transduced cell" relates to a genetically modified cell i.e. a cell wherein the transgene has been introduced deliberately. The herein provided transduced cell comprises the transgene of the present invention. As used herein, the term "patient" or "patient in need thereof", is intended for a human or non-human mammal. Typically, the patient is affected or likely to be affected with a retinal pigment epithelial disease. As used herein, the term “retinal pigment epithelial disease” refers to an inherited retinal disease or an acquired retinal disease that affects the function of the RPE. As used herein, the term "treatment" or "treat" refer to both prophylactic or preventive treatment as well as curative or disease modifying treatment, including treatment of patient at risk of contracting the disease or suspected to have contracted the disease as well as patients who are ill or have been diagnosed as suffering from a disease or medical condition, and includes suppression of clinical relapse. The treatment may be administered to a patient having a medical disorder or who ultimately may acquire the disorder, in order to prevent, cure, delay the onset of, reduce the severity of, or ameliorate one or more symptoms of a disorder or recurring disorder, or in order to prolong the survival of a patient beyond that expected in the absence of such treatment. By "therapeutic regimen" is meant the pattern of treatment of an illness, e.g., the pattern of dosing used during therapy. A therapeutic regimen may include an induction regimen and a maintenance regimen. The phrase "induction regimen" or "induction period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the initial treatment of a disease. The general goal of an induction regimen is to provide a high level of drug to a patient during the initial period of a treatment regimen. An induction regimen may employ (in part or in whole) a "loading regimen", which may include administering a greater dose of the drug than a physician would employ during a maintenance regimen, administering a drug more frequently than a physician would administer the drug during a maintenance regimen, or both. The phrase "maintenance regimen" or "maintenance period" refers to a therapeutic regimen (or the portion of a therapeutic regimen) that is used for the maintenance of a patient during treatment of an illness, e.g., to keep the patient in remission for long periods of time (months or years). A maintenance regimen may employ continuous therapy (e.g., administering a drug at a regular interval, e.g., weekly, monthly, yearly, etc.) or intermittent therapy (e.g., interrupted treatment, intermittent treatment, treatment at relapse, or
treatment upon achievement of a particular predetermined criteria [e.g., pain, disease manifestation, etc.]). As used herein, the term “therapeutic level” refers to the amount of a transgene product or the level of activity of a transgene product sufficient to confer its therapeutic or beneficial effect(s) in the host receiving the transgene. Expression levels of the transgene or the levels of activity of the transgene product can be measured at the protein or the mRNA level using methods known in the art. As used herein, the term “therapeutically efficient amount” is intended an amount of pharmaceutical composition of the present invention administered to a subject that is sufficient to constitute a treatment as defined above of ocular disease. Promoter of the present invention: The first object of the present invention relates to a polynucleotide having a promoter activity in retinal pigment epithelium (RPE) having a nucleic acid sequence having at least 80% of identity with the nucleic acid sequence as set forth in SEQ ID NO:1. > SEQ ID NO:1 CGCTGCCTCTGTTGGGAGGGGGCGGGGACAAGAGGGAGGGGCTGGGCCAGCAGCCTAGAG AAGGAGGGGGATGGAGACGAAGCATGGGGAAACTGAGGCACAGGGCTACCCACGTGAGGG TGGCTCAGGTCTGGGAACCAAAAGGGGAAACGGAGGTAGGACTCTCACCCCAAGTCTCCT TCTCCTGCAAAGGGCGCTGAAGGACGAGGGGAGGACGACGGGTCCCACCTGACTCTGCAC CTCTGCAGGCTCCCTCTGAAGGACAACTGCTGGCCCCTCAGGGCCTCAGGTGGAAGGCGT CGGGAAGTGGAGCAGGTATGTGCCTGGAGGTGGGCGTCCAGCACCAAAGTCGCCCCGTAC GCGTGAGGGTCTCGGGACAACAGAACAAACAGCAGAGGGGCAGATGCCGGCTGCAAAGGG CCGCTGTGCGTGATTTTCCTCTTTAGCCTTCCGCCATGTTCCTAATGTTTCTCAGTGATC ACAAATGACTATTGTCATTAGAACATTAAGGGGACAAAAGCCACTTTCAACAGATATCAG CCAGGGGAGCTGAGTTCCCATGCATCTCAGCTCAGCCATGGATTCCCTCCCTGACTTGGG CCGGTCGCCTCCCCTCTCCCAACCTCCCTTTCCTCCGATGTAAGATGGGTGATTGATCAC CCCGAGGCCAGCTGGGAATCTTGCGGGAGAGGAGTCAGGTCTCTGGCTTCTGCCAGACAG GTAGGAACCTTAGCAGCACTGAGAGAGTGGCCTTCTGCCCTCCAAACCTCAAACCCTCTA TATGAAAAGAATGGCTCCAAAGCCTGTCTCATTCCTGGGGCTCCACCCGGGGCCCTCACA GCAGTTACAAGCGCCCCCATAACAGATGAGGAGGGTGTACTCAGAGACAGGTGCACCAGG AGCCGGGGGCTGGGGATAGCCTCCTGCCCTCCTGCCTGGAGTGTCCCTCCTGGCACTGGG CAGTGCCACCACCCCCAGGGTATCCGGGACTCTGCCCCTCCTCCACTAGGACCAGGTTGT GGCTTTTCCTGCCTGTTGTCCTCTTGTCACAGGGGCAGCCACTTGAAGGTGGAGACAGGA GGGTGATCCCAGGCAGCCCTCACATCACACTCACCTGAGCCTGCCTCGTGGGTTTGGGGG TCCAGCCTCACATCTCCTGAGGCCTGCAGGGAGGGGCCGCCGCCGCAGCTCCCGGTGAAG CTTACCCGGATGGGGGCTGCCAGACACCCCAGCTTCCCACCAGCCTGGGCTGAGCCTCTG GCCGACTCCCGCCTGACGCTCCCTGCCCCAGGCCTGGAGCTTAGAGTGTGTGCCCCCACC ACACCTCACCCCTGCTAAGAGGCTTTTGTCCAGGAGGGGAGAAGAGACTGGGAGCCCCAG GCCAGGCTGGGCCTCGGCGGTGGGCTCCCTGTCCCAAGCCAGTCCAGGAGGGAGGAAGAG AATCCCTGTGCCTCCTGGAGCCTGGGGTGGTAGACACAGAGACCTTGCCCAGGGGGTCCT AGTCTGGTGCTGGGGGCACAGCCCTGCCCTGAAGAGACCCCAACGGTTCTAATCACCTCA
GTGCCTCATCTGCCCCCGCCCCAACCCCCCGCCCCAGGCCACATTCCAGGAGGGGCTGGC AGCTTTCTCTAGAATGAAGCTAATCCACGCCTGTGCCTGGGAATGTGGGGGGGGGAGGGG CCAAAACAAAGGGTGCTGTGTCCAGGCCGCCTGAGGGCAGCTTGCTGGTGGTCCCAGCCC TTGGCCACTGCCACCTCTCCCCTGACCCGAGCAGCCGCTACTTCCTAGGAAACCCACAGG TCGAAATGAGAGCAGGTGGAGATGGGGGCTGCAGCGCGAGGGCTTTGGGGCAGCCCTAAA GAGTTCTCCAAACAAACAAACAAGGGGCTGGGAGTGAGATGGCTTCCCGGGGAACAGCCA GGACCTCAGTACCCCCTAGACCCCATTCTCCAATGGGGGAGTCTCTATCCCTCACCCCCT GCCCTGTCCCCCAAATAAGACACCACCAGTCCTG The nucleic acid sequence of SEQ ID NO: 1 was derived from the regulatory region of the SLC16A8 gene encodes for a retinal pigment epithelium (RPE)-specific lactate transporter MCT3. The promoter of the present invention thus exhibits a promoter activity in RPE, i.e. when introduced in RPE, it can initiate transcription of a nucleic acid to which it is operably linked. Typically, the promoter activity is specific of RPE cells. The term “specific of RPE cells” shall be understood to mean a promoter mainly active in RPE cells. It shall be understood that a residual expression, generally lower, in other tissues or cells cannot be entirely excluded. In some embodiments, the promoter of the present invention is not active in Muller cells or in photoreceptors. In some embodiments, the promoter of the present invention comprises, or consists of, the sequence of SEQ ID NO: 1. In some embodiments, the promoter of the present invention comprises, or consists of, a functional variant of SEQ ID NO: 1. As used herein, the term “variant” refers to a nucleic acid sequence differing from the original sequence, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the original polynucleotide. The sequence of the variant may differ by nucleotide substitutions, deletions or insertions of one or more nucleotides in the sequence, which do not impair the promoter activity. The variant may have the same length of the original sequence, or may be shorter or longer. The term “functional variant” refers to a variant of SEQ ID NO: 1 that exhibits a promoter activity of SEQ ID NO: 1, i.e. that exhibits a promoter activity in RPE cells, typically a promoter activity specific of RPE cells. In some embodiments, the promoter of the present invention comprises, or consists of, a functional variant having at least 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% identity to SEQ ID NO: 1, typically over the entire sequence of SEQ ID NO:
1. The promoter of the present invention may differ from the polynucleotide of SEQ ID NO: 1 by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 substitutions, deletions and/or insertions. In some embodiments, the promoter of the present invention comprises, or consists of, a functional variant having a sequence capable of hybridizing under low, medium or high stringency conditions with the nucleic acid sequence of SEQ ID NO: 1 or its complementary strand, typically under medium stringency conditions, more typically under high stringency conditions. As used herein, the term “low stringency conditions” means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C. in 5×SSPE, 0.3% SDS, 200 micrograms/mL sheared and denatured salmon sperm DNA, and 25% formamide, following standard Southern blotting procedures for 12 to 24 hours. The carrier material is finally washed three times each for 15 minutes using 2×SSC, 0.2% SDS at 50°C. The term “medium stringency conditions” means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C. in 5×SSPE, 0.3% SDS, 200 micrograms/mL sheared and denatured salmon sperm DNA, and 35% formamide, following standard Southern blotting procedures for 12 to 24 hours. The carrier material is finally washed three times each for 15 minutes using 2×SSC, 0.2% SDS at 55°C. The term “high stringency conditions” means for probes of at least 100 nucleotides in length, prehybridization and hybridization at 42°C. in 5×SSPE, 0.3% SDS, 200 micrograms/mL sheared and denatured salmon sperm DNA, and 50% formamide, following standard Southern blotting procedures for 12 to 24 hours. The carrier material is finally washed three times each for 15 minutes using 2×SSC, 0.2% SDS at 65°C. Expression cassettes of the present invention: A further object of the present invention relates to an expression cassette comprising a polynucleotide of the present invention operably linked to a polynucleotide of interest. Typically, the nucleic acid operably linked to the promoter of the present invention is a transgene that encodes a polypeptide of interest or a polynucleotide of interest. Typically, the promoter of the present invention is operably linked to a heterologous polynucleotide. As used herein, the term “heterologous” means a nucleic acid other than the
nucleic acid that the promoter is operably linked to in a naturally occurring genome. In some embodiments, the promoter of the present invention is not operably linked to SLC16A8 gene, and in particular to the human SLC16A8 gene. In some embodiments, the polynucleotide operably linked to the promoter of the present invention encodes a polypeptide of interest. The polypeptide of interest may be any polypeptide of which expression in RPE cells is desired. In particular, the polypeptide of interest may be a therapeutic polypeptide, or reporter protein. In some embodiments, the nucleic acid operably linked to the promoter of the present invention is a therapeutic gene. Examples of therapeutic genes include, but are not limited to, nucleic acids for replacement of a missing or mutated gene known to cause retinal disease such as ARSG, BEST1, DFNB31, IQCB1, KCNJ13, LCA5, LRAT, MERTK, RGR, RPE65, SPATA7. In some embodiments, the therapeutic gene may also encode neurotrophic factors such as GDNF (Gene ID: 2668), CNTF (Gene ID: 1270), FGF2 (Gene ID: 2247), BDNF (Gene ID: 627) and EPO (Gene ID: 2056), anti-apoptotic genes such as BCL2 (Gene ID: 596) and BCL2L1 (Gene ID: 598), anti-angiogenic factors such as endostatin, angiostatin and sFlt, anti-inflammatory factors such as IL10 (Gene ID: 3586), IL1R1 (Gene ID: 3554), TGFBI (Gene ID; 7045) and IL4 (Gene ID: 3565), or nucleoredoxin-like 1 the rod-derived cone viability factor (RdCVF) and RdCVFL (Gene ID: 115861). In some embodiments, the transgene product of interest is an endonuclease that provides for site-specific knock-down of gene function, e.g., where the endonuclease knocks out an allele associated with a RPE disease. For example, where a dominant allele encodes a defective copy of a gene that, when wild-type, is a retinal structural protein and/or provides for normal retinal function, a site-specific endonuclease can be targeted to the defective allele and knock out the defective allele. In addition to knocking out a defective allele, a site-specific nuclease can also be used to stimulate homologous recombination with a donor DNA that encodes a functional copy of the protein encoded by the defective allele. Thus, e.g., the method of the invention can be used to deliver both a site-specific endonuclease that knocks out a defective allele, and can be used to deliver a functional copy of the defective allele, resulting in repair of the defective allele, thereby providing for production of a functional retinal protein (e.g., functional retinoschisin, functional RPE65, functional peripherin, etc.). See, e.g., Li et al. (2011) Nature 475:217. In some embodiments, the DNA targeting endonuclease of the present invention is a
TALEN. In some embodiments, the DNA targeting endonuclease of the present invention is a ZFN. In some embodiments, the DNA targeting endonuclease of the present invention is a CRISPR-associated endonuclease. In some embodiments, the CRISPR-associated endonuclease is a Cas9 nuclease. In some embodiments, the CRISPR-associated endonuclease is a Cpf1 nuclease. In some embodiments, the polynucleotide operably linked to the promoter of the present invention encodes a polynucleotide of interest. The polynucleotide of interest may be any nucleic acid of which expression in RPE cells is desired. In particular, the polynucleotide of interest may be a therapeutic nucleic acid. The nucleic acid may be, for example, an siRNA, an shRNA an RNAi, a miRNA, an antisense RNA, a ribozyme or a DNAzyme. In some embodiments, the nucleic acid encodes an RNA that when transcribed from the nucleic acid operably linked to the promoter of the present invention can treat or prevent a retinal pigment epithelial disease by interfering with translation or transcription of an abnormal or excess protein associated with said disorder. For example, the polynucleotide of interest may encode for an RNA, which treats the disease by highly specific elimination or reduction of mRNA encoding the abnormal and/or excess proteins. The expression cassette of the present invention may comprise one or more nucleic acids operably linked to the promoter of the present invention. For example, the promoter may be operably linked to one or more therapeutic genes and a nucleic acid encoding a reporter protein or to a therapeutic gene. Vectors of the present invention: A further object of the present invention relates to a vector comprising the promoter of the present invention or the expression cassette of the present invention. Typically, the vector of the present invention is a vector suitable for use in gene therapy, and in particular is suitable to target RPE cells. The vector of the present invention is typically a viral genome vector including any element required to establish the expression of the polypeptide of interest in a host cell such as, for example, a promoter, e.g., a polynucleotide of the present invention, an ITR, a ribosome binding
element, terminator, enhancer, selection marker, intron, polyA signal, and/or origin of replication. In some embodiments, the vector is a viral vector, such as vectors derived from Moloney murine leukemia virus vectors (MoMLV), MSCV, SFFV, MPSV or SNV, lentiviral vectors (e.g. derived from human immunodeficiency virus (HIV), simian immunodeficiency virus (SIV), feline immunodeficiency virus (FIV), bovine immunodeficiency virus (BIV) or equine infectious anemia virus (EIAV)), adenoviral (Ad) vectors, adeno-associated viral (AAV) vectors, simian virus 40 (SV-40) vectors, bovine papilloma virus vectors, Epstein-Ban virus, herpes virus vectors, vaccinia virus vectors, Harvey murine sarcoma virus vectors, murine mammary tumor virus vectors, Rous sarcoma virus vectors. In some embodiments, the vector is a retroviral vector, typically a lentiviral vector or a non- pathogenic parvovirus. As is known in the art, depending on the specific viral vector considered for use, suitable sequences should be introduced in the vector of the present invention for obtaining a functional viral vector, such as AAV ITRs for an AAV vector, or LTRs for lentiviral vectors. In some embodiments, the vector is an AAV vector. As used herein, the term “AAV vector” refers to a polynucleotide vector comprising one or more heterologous sequences (i.e., nucleic acid sequence not of AAV origin) that are flanked by at least one AAV inverted terminal repeat sequence (ITR), typically two ITRs. Such AAV vectors can be replicated and packaged into infectious viral particles when present in a host cell that has been infected with a suitable helper virus (or that is expressing suitable helper functions) and that is expressing AAV rep and cap gene products (i.e. AAV Rep and Cap proteins). An “inverted terminal repeat” or “ITR” sequence is a term well understood in the art and refers to relatively short sequences found at the termini of viral genomes which are in opposite orientation. An “AAV inverted terminal repeat (ITR)” sequence is an approximately 145-nucleic acid sequence that is present at both termini of the native single-stranded AAV genome. The outermost 125 nucleotides of the ITR can be present in either of two alternative orientations, leading to heterogeneity between different AAV genomes and between the two ends of a single AAV genome. The outermost 125 nucleotides also contain several shorter regions of self-complementarity (designated A, A′, B, B′, C, C and D regions), allowing intra-strand base-pairing to occur within this portion of
the ITR. AAV ITRs for use in the vectors of the present invention may have a wild-type nucleic acid sequence or may be altered by the insertion, deletion or substitution. The serotype of the inverted terminal repeats (ITRs) of the AAV vector may be selected from any known human or nonhuman AAV serotype. The promoter or expression cassette of the present invention may be introduced into the vector by any method known by the skilled person. The vector of the present invention may be packaged into a virus capsid to generate a “viral particle”. Thus, in a further aspect, the present invention also relates to a viral particle comprising a vector of the present invention. In some embodiments, the vector is an AAV vector and is packaged into an AAV-derived capsid to generate an “adeno-associated viral particle” or “AAV particle”. Thus, used herein, the term “AAV particle” refers to a viral particle composed of at least one AAV capsid protein and an encapsidated AAV vector genome. The capsid serotype determines the tropism range of the AAV particle. Multiple serotypes of adeno-associated virus (AAV), including 12 human serotypes and more than 100 serotypes from nonhuman primates have now been identified (Howarth al., 2010, Cell Biol Toxicol 26: 1-10). Among these serotypes, human serotype 2 was the first AAV developed as a gene transfer vector. Other currently used AAV serotypes include, but are not limited to, AAV1, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAVrh8, AAV9, AAV10, AAVrh10, AAV11, AAV12, AAVrh74 and AAVdj, etc. In addition, non-natural engineered variants and chimeric AAV can also be useful. In particular, the capsid proteins may be variants comprising one or more amino acid substitutions enhancing transduction efficiency. Different AAV serotypes are used to optimize transduction of particular target cells or to target specific cell types within a particular target tissue (e.g., RPE cells). An AAV particle can comprise viral proteins and viral nucleic acids of the same serotype or any natural or artificial sequence variant of AAV. For example, the AAV particle may comprise AAV2 capsid proteins and at least one, typically two, AAV2 ITR. Any combination of AAV serotypes for production of an AAV particle is provided herein as if each combination had been expressly stated herein. In preferred embodiment, the AAV particle comprises an AAV-derived capsid selected from the group consisting of AAV2, AAV-5, AAV-7m8 (AAV2-7m8, Dalkara et al. Sci Transl Med (2013), 5, 189ra76), AAV9 or AAV8 capsid.
AAV viruses may be engineered using conventional molecular biology techniques, making it possible to optimize these particles for cell specific delivery of nucleic acid sequences, for minimizing immunogenicity, for tuning stability and particle lifetime, for efficient degradation, for accurate delivery to the nucleus. Alternatively, to using AAV natural serotypes, artificial AAV serotypes may be used in the context of the present invention, including, without limitation, AAV with a non-naturally occurring capsid protein. Such an artificial capsid may be generated by any suitable technique, using a selected AAV sequence (e.g., a fragment of a VP1 capsid protein) in combination with heterologous sequences which may be obtained from a different selected AAV serotype, non- contiguous portions of the same AAV serotype, from a non-AAV viral source, or from a non- viral source. An artificial AAV serotype may be, without limitation, a chimeric AAV capsid or a mutated AAV capsid. Numerous methods are known in the art for production of viral particles, and in particular AAV particles, including transfection, stable cell line production, and infectious hybrid virus production systems which include adenovirus-AAV hybrids, herpesvirus-AAV hybrids (Conway, J E et al., (1997) Virology 71(11):8780-8789) and baculovirus-AAV hybrids). AAV production cultures for the production of AAV virus particles all require; 1) suitable host cells, including, for example, human-derived cell lines such as HeLa, A549, or 293 cells, or insect- derived cell lines such as SF-9, in the case of baculovirus production systems; 2) suitable helper virus function, provided by wild-type or mutant adenovirus (such as temperature sensitive adenovirus), herpes virus, baculovirus, or a plasmid construct providing helper functions; 3) AAV rep and cap genes and gene products; 4) a polynucleotide of interest flanked by at least one AAV ITR sequences, e.g., a vector of the present invention; and 5) suitable media and media components to support AAV production that are well-known in the art. In practicing the invention, host cells for producing AAV particles include mammalian cells, insect cells, plant cells, microorganisms and yeast. Host cells can also be packaging cells in which the AAV rep and cap genes are stably maintained in the host cell or producer cells in which the AAV vector genome is stably maintained. Exemplary packaging and producer cells are derived from 293, A549 or HeLa cells. AAV particles are then purified and formulated using standard techniques known in the art. Host cells of the present invention:
A further object of the present invention to an isolated host cell transformed or transfected with an expression cassette, vector or viral particle of the present invention. The host cell may be any animal cell, plant cell, bacterium cell or yeast. Typically, the host cell is a mammalian cell or an insect cell. More typically, the host cell is a human cell. In some embodiments, the host cell is a RPE cell, in particular a human RPE cell. The expression cassette or vector of the present invention may be transferred into host cells using any known technique including, but being not limited to, calcium phosphate-DNA precipitation, DEAE-Dextran transfection, electroporation, microinjection, biolistic, lipofection, or viral infection, and may be maintained in the host cell in an ectopic form or may be integrated into the genome. In some embodiments, the expression cassette or vector of the present invention is transferred into the host cell by viral infection, typically using a viral particle of the present invention, more typically using an AAV particle of the present invention. Pharmaceutical compositions of the present invention: The present invention also relates to a pharmaceutical composition comprising an expression cassette, vector, viral particle or cell of the present invention. Such compositions comprise a therapeutically effective amount of the therapeutic agent (an expression cassette, vector, viral particle or cell of the present invention), and a pharmaceutically acceptable excipient. As used herein, the term “pharmaceutically acceptable” means approved by a regulatory agency or recognized pharmacopeia such as European Pharmacopeia, for use in animals and/or humans. The term “excipient” refers to a diluent, adjuvant, carrier, or vehicle with which the therapeutic agent is administered. As is well known in the art, pharmaceutically acceptable excipients are relatively inert substances that facilitate administration of a pharmacologically effective substance and can be supplied as liquid solutions or suspensions, as emulsions, or as solid forms suitable for dissolution or suspension in liquid prior to use. For example, an excipient can give form or consistency, or act as a diluent. Suitable excipients include but are not limited to stabilizing agents, wetting and
emulsifying agents, salts for varying osmolality, encapsulating agents, pH buffering substances, and buffers. Such excipients include any pharmaceutical agent suitable for direct delivery to the eye which may be administered without undue toxicity. Pharmaceutically acceptable excipients include, but are not limited to, sorbitol, any of the various tween compounds, and liquids such as water, saline, glycerol and ethanol. Pharmaceutically acceptable salts can be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable excipients is available in Remington's Pharmaceutical Sciences, 15th Edition. Typically, the composition is formulated to be administered to the eye, in particular by intraocular injection, e.g., by subretinal and/or intravitreal administration. Accordingly, the composition can be combined with pharmaceutically acceptable excipient such as saline, Ringer's balanced salt solution (pH 7.4), and the like. The pharmaceutical compositions described herein can be packaged in single unit dosages or in multidosage forms. In some embodiments, the pharmaceutical composition comprises a vector or viral particle of the present invention, more typically an AAV vector or particle. In some embodiments, the pharmaceutical composition comprises host cells of the present invention, typically human host cell of the present invention, i.e. transformed or transfected with an expression cassette, vector or viral particle of the present invention, typically with an AAV particle. Optionally, the composition comprising host cells may be frozen for storage at any temperature appropriate for storage of the cells. For example, the cells may be frozen at about −20° C., −80°C. or any other appropriate temperature. Cryogenically frozen cells may be stored in appropriate containers and prepared for storage to reduce risk of cell damage and maximize the likelihood that the cells will survive thawing. Alternatively, the cells may also be maintained at room temperature of refrigerated, e.g. at about 4°C. The amount of pharmaceutical composition to be administered may be determined by standard procedure well known by those of ordinary skill in the art. Physiological data of the patient (e.g. age, size, and weight) and type and severity of the disease being treated have to be taken into account to determine the appropriate dosage.
The pharmaceutical composition of the present invention may be administered as a single dose or in multiple doses. In some embodiments, the composition comprises viral particles of the present invention and each unit dosage comprises from 108 to 1013 viral particles, typically from 109 to 1012 particles. The pharmaceutical composition may further comprise one or several additional active compounds such as corticosteroids, antibiotics, analgesics, immunosuppressants, trophic factors, or any combinations thereof. Methods of therapy of the present invention: A further object of the present invention relates to a method of therapy in a patient in need thereof comprising administering to the patient a therapeutically effective amount of an expression cassette, a vector, a viral particle, a host cell or a pharmaceutical composition of the present invention. The method of the present invention is particularly suitable for the treatment of an ocular disease. The method of the present invention is particularly suitable for the treatment of a retinal pigment epithelial disease. Examples of retinal pigment epithelial diseases include, but are not limited to age-related macular degeneration, Leber's hereditary optic neuropathy, cone-rod dystrophy, Leber congenital amaurosis, Stargardt's disease, diabetic retinopathy, retinal detachment, Best's disease, retinitis pigmentosa, choroideremia and a tapetoretinal degeneration. Alternatively, the ocular disease is selected from disease which are not necessarily or specifically associated with RPE, but which can be treated or prevented by expressing specifically nucleic acid encoding a polypeptide or polynucleotide of interest in RPE cell. In the method for treating ocular disease of the present invention, the pharmaceutical composition of the present invention is typically administered intraocularly, more typically by subretinal or intravitreal administration.
The method of the present invention may also further comprise administering at least one additional therapeutic agent to the subject. In particular, said therapeutic agent may be selected from the group consisting of a corticosteroid, an antibiotic, an analgesic, an immunosuppressant, or a trophic factor, or any combinations thereof. The composition of the present invention may be administered before or after the disease becomes symptomatic, e.g., before or after partial or complete RPE degeneration and/or before or after partial or complete loss of vision. The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention. EXAMPLE: To study the promoter of the SLC16A8 gene, we first needed to identify the promoter region of the gene and hypothesized that the genomic sequence between the transcription start site (TSS) of SLC16A8 and the end of the BAIAP2L2 gene located immediately on 5' end (Chr22: 38,083,143-38,084,902) might contain the promoter region. To test the ability of this promoter region, we combined it with a reporter gene encoding enhanced green fluorescent protein (eGFP) in a plasmid carrying the inverted terminal repeat (ITR) sequences of AAV serotype 2. We therefore performed the cloning of the region measuring 1.7 kb (-1759 to -1 bp) in the plasmid pAAV2-CMV/CBA-eGFP which contains the transgenic cassette of the eGFP reporter gene, under the control of the chimeric promoter associating that of cytomegalovirus and chicken beta-actin or chicken β-actin (CMV/CBA). The digestion/ligation process was used to remove the CMV/CBA promoter from the recipient plasmid to introduce our candidate promoter region instead. We chose to take advantage of the restricted expression of SLC16A8 in the RPE and perform this experiment using a novel in vivo approach through the use of recombinant adeno-associated viruses (rAAV). BALB/cJ strain mice were selected as the experimental model. Knowing that the efficiency of transduction in the retina by an adeno- associated vector serotype 9 (AAV9) following systemic injection has been demonstrated previously, we generated an AAV9 vector to assess the ability of the SLC16A81.8 region(AAV2.9-SLC16A81.8-eGFP) to direct GFP expression in vivo in RPE cells. Therefore, a RPE-restricted fluorescent signal after systemic injection of AAV2.9-SLC16A81.8-eGFP (“SLC16A81.8”) would imply that the 1.8 kb fragment contains the entire promoter with its
regulatory elements. Viruses were administered systemically by intracardiac injection in 3- to 4-day-old wt/wt BALB/cJ mice (D3-J4 ) to study in vivo GFP expression. Intracardiac injection of CMV/CBA- led to very strong GFP expression, confirming the correct performance of the technical procedure. In contrast, SLC16A81.8 did not show a green fluorescent signal at the macroscopic scale. To see on a microscopic scale whether the RPE of mice injected with SLC16A81.8 expressed GFP, we performed a western blot of RPE/choroid protein extracts obtained by sonication and centrifugation of the sclera/choroid/RPE cup and neural retina of injected mice following their sacrifice. As observed in fundus images, GFP expression is very high in both the RPE/choroid and retinas of CMV/CBV-injected mice. However, mice injected with SLC16A81.8, express low levels of GFP in the RPE/choroid. The absence or low expression of GFP in the RPE of mice injected with SLC16A81.8 led us to question whether the TSS reported in the UCSC database corresponded to a minor TSS. Indeed, SLC16A8 expression is restricted to the RPE, a tissue that is not among those analysed by ENCODE. Results from the study of the NXNL1 and NXNL2 genes show that there are several TSSs. Thus, for SLC16A8, we used the TSS of EST AF132610 (38078137-38083142) that was deposited by Nancy Philp, the researcher who identified the gene from the RPE (Yoon, Heeyong, et al. "Identification of a unique monocarboxylate transporter (MCT3) in retinal pigment epithelium." Biochemical and biophysical research communications 234.1 (1997): 90-94.). To circumvent this problem of the position of the TSS in our construct, we constructed a new reporter whose 3' end corresponds to the transduction initiation site, located in exon 3 of the SLC16A8 gene. This construct, which includes the 3' UTR of the SLC16A8 messenger, necessarily includes the TSS(s) regardless of its position in the genome. Therefore, the sequence upstream of the translation initiation codon (ATG) of the SLC16A8 gene (-1724 to +290 bp) was cloned, designated the “SLC16A82.1 promoter”, and was packaged into an AAV9 vector (AAV2.9-SLC16A82.1-eGFP). In addition, we generated an AAV9 vector with the BEST1 gene promoter (AAV2.9-5'585BEST1-eGFP – “5'BEST1”), for use as a specific control for expression by RPE. We again performed intracardiac injections according to the previously described protocol. AAV2.9-5'585BEST1- eGFP shows a GFP signal as green fluorescent dots. The specific expression of 5'BEST1 by the RPE suggests that these dots represent expression of the GFP reporter protein under the control of the active promoter in the RPE and not in the retina. While we did not observe a fluorescent signal in the eye fundus of mice injected with the SLC16A81.8 vector, as before, mice injected with the SLC16A82.1vector show intense fluorescent dots associated with more diffuse labeling confirming the presence of regulatory elements in the SLC16A82.1 vector. These observations were confirmed by quantitative western blot analysis as quantification of specific GFP
expression in the RPE/choroid and in the retina (revealed significantly higher GFP expression by the SLC16A82.1 vector compared with that expressed by the 5'BEST1 vector. Taken together, these results demonstrate that the SLC16A82.1 fragment comprises the regulatory elements controlling SLC16A8 promoter activity by RPE cells. REFERENCES: Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
Claims
CLAIMS: 1. A polynucleotide having a promoter activity in retinal pigment epithelium (RPE) having a nucleic acid sequence having at least 80% of identity with the nucleic acid sequence as set forth in SEQ ID NO:1. 2. The polynucleotide of claim 1 that comprises, or consists of, the sequence of SEQ ID NO: 1. 3. The polynucleotide of claim 1 that comprises, or consists of, a functional variant having at least 80% identity with the nucleic acid sequence as set forth in SEQ ID NO:1 and may differ from the polynucleotide of SEQ ID NO: 1 by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 substitutions, deletions and/or insertions. 4. The polynucleotide of claim 1 that comprises, or consists of, a functional variant having a sequence capable of hybridizing under low, medium or high stringency conditions with the nucleic acid sequence of SEQ ID NO: 1 or its complementary strand, typically under medium stringency conditions, more typically under high stringency conditions. 5. An expression cassette comprising the polynucleotide according to any one of claims 1 to 4 operably linked to a polynucleotide of interest. 6. The expression cassette of claim 5 wherein the polynucleotide of interest is a transgene that encodes a polypeptide of interest or a polynucleotide of interest. 7. The expression cassette of claim 6 wherein the transgene is a therapeutic gene. 8. The expression cassette of claim 6 wherein the transgene encodes for an endonuclease that provides for site-specific knock-down of gene function. 9. The expression cassette of claim 6 wherein the polynucleotide of interest is a therapeutic nucleic acid that is an siRNA, an shRNA an RNAi, a miRNA, an antisense RNA, a ribozyme or a DNAzyme. 10. A vector comprising the polynucleotide according to any of claims 1 to 4 or the expression cassette according to any one of claims 5 to 9. 11. The vector of claim 10 that is a viral vector, in particular an AAV vector.
12. An isolated host cell transformed or transfected with the expression cassette according to any one of claims 5 to 9 or the vector according to claim 10 or 11. 13. A pharmaceutical composition comprising the expression cassette according to any one of claims 5 to 9 or the vector according to claim 10 or 11 or the cell according to claim 12. 14. A method of therapy in a patient in need thereof comprising administering to the patient a therapeutically effective amount of the expression cassette according to any one of claims 5 to 9 or the vector according to claim 10 or 11, the cell according to claim 12 or the pharmaceutical composition according to claim 13. 15. The method of claim 14 wherein the patient suffers from a retinal pigment epithelial disease such as age-related macular degeneration, Leber's hereditary optic neuropathy, cone-rod dystrophy, Leber congenital amaurosis, Stargardt's disease, diabetic retinopathy, retinal detachment, Best's disease, retinitis pigmentosa, choroideremia or a tapetoretinal degeneration.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/IB2022/000679 WO2024110770A1 (en) | 2022-11-22 | 2022-11-22 | A new promoter for retinal pigment epithelium (rpe) targeted gene therapy |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/IB2022/000679 WO2024110770A1 (en) | 2022-11-22 | 2022-11-22 | A new promoter for retinal pigment epithelium (rpe) targeted gene therapy |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2024110770A1 true WO2024110770A1 (en) | 2024-05-30 |
Family
ID=85018710
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/IB2022/000679 WO2024110770A1 (en) | 2022-11-22 | 2022-11-22 | A new promoter for retinal pigment epithelium (rpe) targeted gene therapy |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2024110770A1 (en) |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007078599A2 (en) * | 2005-12-16 | 2007-07-12 | The Board Of Trustees Of The Leland Stanford Junior University | Functional arrays for high throughput characterization of gene expression regulatory elements |
WO2008073303A2 (en) * | 2006-12-07 | 2008-06-19 | Switchgear Genomics | Transcriptional regulatory elements of biological pathways, tools, and methods |
WO2016141078A1 (en) * | 2015-03-02 | 2016-09-09 | Avalanche Biotechnologies, Inc. | Compositions and methods for intravitreal delivery of polynucleotides to retinal cones |
WO2016170348A2 (en) * | 2015-04-22 | 2016-10-27 | Mina Therapeutics Limited | Sarna compositions and methods of use |
WO2019077159A1 (en) * | 2017-10-20 | 2019-04-25 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the subretinal delivery of a therapeutically effective amount of a recombinant aav9-derived vector |
WO2019106035A1 (en) * | 2017-11-30 | 2019-06-06 | Friedrich Miescher Institute For Biomedical Research | Synpiii, a promoter for the specific expression of genes in retinal pigment epithelium |
WO2020152623A1 (en) * | 2019-01-24 | 2020-07-30 | Friedrich Miescher Institute For Biomedical Research | Synp5 (proa9), a promoter for the specific expression of genes in retinal ganglion cells |
-
2022
- 2022-11-22 WO PCT/IB2022/000679 patent/WO2024110770A1/en unknown
Patent Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2007078599A2 (en) * | 2005-12-16 | 2007-07-12 | The Board Of Trustees Of The Leland Stanford Junior University | Functional arrays for high throughput characterization of gene expression regulatory elements |
WO2008073303A2 (en) * | 2006-12-07 | 2008-06-19 | Switchgear Genomics | Transcriptional regulatory elements of biological pathways, tools, and methods |
WO2016141078A1 (en) * | 2015-03-02 | 2016-09-09 | Avalanche Biotechnologies, Inc. | Compositions and methods for intravitreal delivery of polynucleotides to retinal cones |
WO2016170348A2 (en) * | 2015-04-22 | 2016-10-27 | Mina Therapeutics Limited | Sarna compositions and methods of use |
WO2019077159A1 (en) * | 2017-10-20 | 2019-04-25 | INSERM (Institut National de la Santé et de la Recherche Médicale) | Methods of expressing a polynucleotide of interest in the cone photoreceptors of a subject comprising the subretinal delivery of a therapeutically effective amount of a recombinant aav9-derived vector |
WO2019106035A1 (en) * | 2017-11-30 | 2019-06-06 | Friedrich Miescher Institute For Biomedical Research | Synpiii, a promoter for the specific expression of genes in retinal pigment epithelium |
WO2020152623A1 (en) * | 2019-01-24 | 2020-07-30 | Friedrich Miescher Institute For Biomedical Research | Synp5 (proa9), a promoter for the specific expression of genes in retinal ganglion cells |
Non-Patent Citations (7)
Title |
---|
CONWAY, J E ET AL., VIROLOGY, vol. 71, no. 11, 1997, pages 8780 - 8789 |
DALKARA ET AL., SCI TRANSL MED, vol. 5, 2013, pages 189 - 76 |
HOWARTH, CELL BIOL TOXICOL, vol. 26, 2010, pages 1 - 10 |
JOHARI YUSUF B. ET AL: "Design of synthetic promoters for controlled expression of therapeutic genes in retinal pigment epithelial cells", BIOTECHNOLOGY AND BIOENGINEERING, vol. 118, no. 5, 3 March 2021 (2021-03-03), Hoboken, USA, pages 2001 - 2015, XP093045465, ISSN: 0006-3592, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/bit.27713> DOI: 10.1002/bit.27713 * |
LI ET AL., NATURE, vol. 475, 2011, pages 217 |
NEEDLEMAN, SAUL B.WUNSCH, CHRISTIAN D.: "A general method applicable to the search for similarities in the amino acid sequence of two proteins", JOURNAL OF MOLECULAR BIOLOGY, vol. 48, no. 3, 1970, pages 443 - 53, XP024011703, DOI: 10.1016/0022-2836(70)90057-4 |
YOON, HEEYONG ET AL.: "Identification of a unique monocarboxylate transporter (MCT3) in retinal pigment epithelium", BIOCHEMICAL AND, vol. 234, no. 1, 1997, pages 90 - 94, XP002957130, DOI: 10.1006/bbrc.1997.6588 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP2024073536A (en) | Gene editing of deep intronic mutations | |
US20210154325A1 (en) | Promoters, expression cassettes, vectors, kits, and methods for the treatment of achromatopsia and other diseases | |
US9982275B2 (en) | Promoters, expression cassettes, vectors, kits, and methods for the treatment of achromatopsia and other diseases | |
US20210330816A1 (en) | Gene therapy for ocular disorders | |
MX2013009604A (en) | Aav -vectors for use in gene therapy of choroideremia. | |
US20040208847A1 (en) | Method and vectors for selectively transducing retinal pigment epithelium cells | |
US20230233709A1 (en) | Gene therapy for ocular disorders | |
JP2023116709A (en) | Gene therapy for ocular disorders | |
AU2018262427B2 (en) | Gene therapy for ciliopathies | |
US11793887B2 (en) | Gene therapy for treating peroxisomal disorders | |
CN114144518A (en) | Dual leucine zipper kinase inhibitors for gene therapy | |
WO2024110770A1 (en) | A new promoter for retinal pigment epithelium (rpe) targeted gene therapy | |
CN115715185A (en) | Compositions and methods for treating neurological diseases | |
JP2022523050A (en) | Highly efficient transduction and lateral spread in the retina by a novel AAV virus enhanced by rational design | |
WO2023030541A1 (en) | Treatment of rpe65-associated eye diseases and disorders | |
KR20240052746A (en) | KCNV2 gene therapy | |
WO2024229049A1 (en) | Dual aav vectors for treating stargardt disease | |
NZ753155B2 (en) | Promoters, expression cassettes, vectors, kits, and methods for the treatment of achromatopsia and other diseases |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 22846924 Country of ref document: EP Kind code of ref document: A1 |