WO2023131648A1 - Nanoparticulate compositions for gene therapy - Google Patents
Nanoparticulate compositions for gene therapy Download PDFInfo
- Publication number
- WO2023131648A1 WO2023131648A1 PCT/EP2023/050181 EP2023050181W WO2023131648A1 WO 2023131648 A1 WO2023131648 A1 WO 2023131648A1 EP 2023050181 W EP2023050181 W EP 2023050181W WO 2023131648 A1 WO2023131648 A1 WO 2023131648A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- polymer
- group
- composition according
- branched
- nanoparticulate composition
- Prior art date
Links
- 239000000203 mixture Substances 0.000 title claims abstract description 67
- 238000001415 gene therapy Methods 0.000 title claims abstract description 20
- 229920000642 polymer Polymers 0.000 claims abstract description 169
- 108020004707 nucleic acids Proteins 0.000 claims abstract description 46
- 102000039446 nucleic acids Human genes 0.000 claims abstract description 46
- 150000007523 nucleic acids Chemical class 0.000 claims abstract description 46
- 125000004427 diamine group Chemical group 0.000 claims abstract description 44
- 150000004985 diamines Chemical class 0.000 claims abstract description 18
- 125000003277 amino group Chemical group 0.000 claims abstract description 11
- 150000002430 hydrocarbons Chemical group 0.000 claims abstract description 9
- 125000004432 carbon atom Chemical group C* 0.000 claims abstract description 7
- 239000004215 Carbon black (E152) Substances 0.000 claims abstract description 5
- 229930195733 hydrocarbon Natural products 0.000 claims abstract description 5
- 239000000126 substance Substances 0.000 claims abstract description 5
- 125000000467 secondary amino group Chemical class [H]N([*:1])[*:2] 0.000 claims abstract description 4
- 125000002924 primary amino group Chemical group [H]N([H])* 0.000 claims abstract description 3
- 125000001302 tertiary amino group Chemical group 0.000 claims abstract 2
- 125000002496 methyl group Chemical group [H]C([H])([H])* 0.000 claims description 39
- 108020004999 messenger RNA Proteins 0.000 claims description 37
- 125000000484 butyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 24
- 238000000034 method Methods 0.000 claims description 22
- 108091032973 (ribonucleotides)n+m Proteins 0.000 claims description 21
- 125000001183 hydrocarbyl group Chemical group 0.000 claims description 14
- 229920000587 hyperbranched polymer Polymers 0.000 claims description 14
- -1 amino ester Chemical class 0.000 claims description 12
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 claims description 12
- 125000001495 ethyl group Chemical group [H]C([H])([H])C([H])([H])* 0.000 claims description 12
- 125000004051 hexyl group Chemical group [H]C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])* 0.000 claims description 12
- 125000000959 isobutyl group Chemical group [H]C([H])([H])C([H])(C([H])([H])[H])C([H])([H])* 0.000 claims description 12
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 claims description 12
- 125000001147 pentyl group Chemical group C(CCCC)* 0.000 claims description 12
- 125000001436 propyl group Chemical group [H]C([*])([H])C([H])([H])C([H])([H])[H] 0.000 claims description 12
- 208000026350 Inborn Genetic disease Diseases 0.000 claims description 11
- 208000016361 genetic disease Diseases 0.000 claims description 11
- 229920000962 poly(amidoamine) Polymers 0.000 claims description 9
- KIUKXJAPPMFGSW-DNGZLQJQSA-N (2S,3S,4S,5R,6R)-6-[(2S,3R,4R,5S,6R)-3-Acetamido-2-[(2S,3S,4R,5R,6R)-6-[(2R,3R,4R,5S,6R)-3-acetamido-2,5-dihydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-2-carboxy-4,5-dihydroxyoxan-3-yl]oxy-5-hydroxy-6-(hydroxymethyl)oxan-4-yl]oxy-3,4,5-trihydroxyoxane-2-carboxylic acid Chemical compound CC(=O)N[C@H]1[C@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O[C@H]1[C@H](O)[C@@H](O)[C@H](O[C@H]2[C@@H]([C@@H](O[C@H]3[C@@H]([C@@H](O)[C@H](O)[C@H](O3)C(O)=O)O)[C@H](O)[C@@H](CO)O2)NC(C)=O)[C@@H](C(O)=O)O1 KIUKXJAPPMFGSW-DNGZLQJQSA-N 0.000 claims description 6
- 125000000954 2-hydroxyethyl group Chemical group [H]C([*])([H])C([H])([H])O[H] 0.000 claims description 6
- 229920002674 hyaluronan Polymers 0.000 claims description 6
- 229960003160 hyaluronic acid Drugs 0.000 claims description 6
- CBOIHMRHGLHBPB-UHFFFAOYSA-N hydroxymethyl Chemical compound O[CH2] CBOIHMRHGLHBPB-UHFFFAOYSA-N 0.000 claims description 6
- 239000008194 pharmaceutical composition Substances 0.000 claims description 5
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 5
- 108091046869 Telomeric non-coding RNA Proteins 0.000 claims description 4
- 125000000623 heterocyclic group Chemical group 0.000 claims description 4
- 125000004433 nitrogen atom Chemical group N* 0.000 claims description 4
- 229920002454 poly(glycidyl methacrylate) polymer Polymers 0.000 claims description 4
- 229940124531 pharmaceutical excipient Drugs 0.000 claims description 3
- 239000001257 hydrogen Substances 0.000 claims description 2
- 229910052739 hydrogen Inorganic materials 0.000 claims description 2
- 125000004435 hydrogen atom Chemical class [H]* 0.000 claims description 2
- 125000002632 imidazolidinyl group Chemical group 0.000 claims description 2
- 125000004193 piperazinyl group Chemical group 0.000 claims description 2
- 125000003386 piperidinyl group Chemical group 0.000 claims description 2
- 125000003072 pyrazolidinyl group Chemical group 0.000 claims description 2
- 125000000719 pyrrolidinyl group Chemical group 0.000 claims description 2
- 229930195734 saturated hydrocarbon Natural products 0.000 claims description 2
- 108090000623 proteins and genes Proteins 0.000 description 38
- 210000004027 cell Anatomy 0.000 description 35
- 238000010362 genome editing Methods 0.000 description 32
- 239000000243 solution Substances 0.000 description 31
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 27
- 239000002245 particle Substances 0.000 description 25
- 229920001223 polyethylene glycol Polymers 0.000 description 22
- 230000003019 stabilising effect Effects 0.000 description 20
- 239000002202 Polyethylene glycol Substances 0.000 description 19
- 201000010099 disease Diseases 0.000 description 19
- 230000035772 mutation Effects 0.000 description 18
- 102000004389 Ribonucleoproteins Human genes 0.000 description 17
- 108010081734 Ribonucleoproteins Proteins 0.000 description 17
- 239000000178 monomer Substances 0.000 description 17
- 238000001890 transfection Methods 0.000 description 16
- 238000011282 treatment Methods 0.000 description 16
- 238000003776 cleavage reaction Methods 0.000 description 15
- 238000006460 hydrolysis reaction Methods 0.000 description 15
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 13
- 230000007062 hydrolysis Effects 0.000 description 13
- 230000015572 biosynthetic process Effects 0.000 description 12
- 210000001519 tissue Anatomy 0.000 description 12
- 206010028980 Neoplasm Diseases 0.000 description 11
- 150000001412 amines Chemical class 0.000 description 11
- 238000003786 synthesis reaction Methods 0.000 description 11
- 241001465754 Metazoa Species 0.000 description 10
- 108020004414 DNA Proteins 0.000 description 9
- 206010014989 Epidermolysis bullosa Diseases 0.000 description 9
- 229920005601 base polymer Polymers 0.000 description 9
- 208000035475 disorder Diseases 0.000 description 8
- 238000004108 freeze drying Methods 0.000 description 8
- 102000004190 Enzymes Human genes 0.000 description 7
- 108090000790 Enzymes Proteins 0.000 description 7
- 125000004386 diacrylate group Chemical group 0.000 description 7
- 238000003197 gene knockdown Methods 0.000 description 7
- 201000000744 recessive dystrophic epidermolysis bullosa Diseases 0.000 description 7
- 210000003491 skin Anatomy 0.000 description 7
- 239000013598 vector Substances 0.000 description 7
- 108091033409 CRISPR Proteins 0.000 description 6
- IAZDPXIOMUYVGZ-UHFFFAOYSA-N Dimethylsulphoxide Chemical compound CS(C)=O IAZDPXIOMUYVGZ-UHFFFAOYSA-N 0.000 description 6
- 241000283086 Equidae Species 0.000 description 6
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 6
- 238000006845 Michael addition reaction Methods 0.000 description 6
- 101710163270 Nuclease Proteins 0.000 description 6
- 108010017070 Zinc Finger Nucleases Proteins 0.000 description 6
- 201000011510 cancer Diseases 0.000 description 6
- 150000002632 lipids Chemical class 0.000 description 6
- 102000004169 proteins and genes Human genes 0.000 description 6
- NIXOWILDQLNWCW-UHFFFAOYSA-M Acrylate Chemical compound [O-]C(=O)C=C NIXOWILDQLNWCW-UHFFFAOYSA-M 0.000 description 5
- 201000003883 Cystic fibrosis Diseases 0.000 description 5
- 208000023105 Huntington disease Diseases 0.000 description 5
- 239000002253 acid Substances 0.000 description 5
- 229920006317 cationic polymer Polymers 0.000 description 5
- 238000009472 formulation Methods 0.000 description 5
- 230000014509 gene expression Effects 0.000 description 5
- 125000005647 linker group Chemical group 0.000 description 5
- 230000009826 neoplastic cell growth Effects 0.000 description 5
- 208000017520 skin disease Diseases 0.000 description 5
- 230000008685 targeting Effects 0.000 description 5
- 230000000699 topical effect Effects 0.000 description 5
- 102000004321 Atrophin-1 Human genes 0.000 description 4
- 108090000806 Atrophin-1 Proteins 0.000 description 4
- 241000282472 Canis lupus familiaris Species 0.000 description 4
- 201000008163 Dentatorubral pallidoluysian atrophy Diseases 0.000 description 4
- 108060001084 Luciferase Proteins 0.000 description 4
- 208000015439 Lysosomal storage disease Diseases 0.000 description 4
- 208000033063 Progressive myoclonic epilepsy Diseases 0.000 description 4
- 238000010459 TALEN Methods 0.000 description 4
- 208000006269 X-Linked Bulbo-Spinal Atrophy Diseases 0.000 description 4
- 239000004480 active ingredient Substances 0.000 description 4
- 239000000872 buffer Substances 0.000 description 4
- 230000004663 cell proliferation Effects 0.000 description 4
- 239000003795 chemical substances by application Substances 0.000 description 4
- 208000037765 diseases and disorders Diseases 0.000 description 4
- 230000000694 effects Effects 0.000 description 4
- 239000000839 emulsion Substances 0.000 description 4
- 230000006870 function Effects 0.000 description 4
- 238000001476 gene delivery Methods 0.000 description 4
- 206010020718 hyperplasia Diseases 0.000 description 4
- 230000006872 improvement Effects 0.000 description 4
- 238000005259 measurement Methods 0.000 description 4
- 208000011045 mucopolysaccharidosis type 3 Diseases 0.000 description 4
- 239000002105 nanoparticle Substances 0.000 description 4
- 210000000329 smooth muscle myocyte Anatomy 0.000 description 4
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 4
- HDTRYLNUVZCQOY-UHFFFAOYSA-N α-D-glucopyranosyl-α-D-glucopyranoside Natural products OC1C(O)C(O)C(CO)OC1OC1C(O)C(O)C(O)C(CO)O1 HDTRYLNUVZCQOY-UHFFFAOYSA-N 0.000 description 3
- 102000014461 Ataxins Human genes 0.000 description 3
- 108010078286 Ataxins Proteins 0.000 description 3
- 208000023275 Autoimmune disease Diseases 0.000 description 3
- 206010004265 Benign familial pemphigus Diseases 0.000 description 3
- 241000283690 Bos taurus Species 0.000 description 3
- 238000010356 CRISPR-Cas9 genome editing Methods 0.000 description 3
- 206010008025 Cerebellar ataxia Diseases 0.000 description 3
- 208000002506 Darier Disease Diseases 0.000 description 3
- 208000037574 Familial benign chronic pemphigus Diseases 0.000 description 3
- 241000282326 Felis catus Species 0.000 description 3
- 208000027655 Hailey-Hailey disease Diseases 0.000 description 3
- 239000012981 Hank's balanced salt solution Substances 0.000 description 3
- 102100023129 Keratin, type I cytoskeletal 9 Human genes 0.000 description 3
- 206010023369 Keratosis follicular Diseases 0.000 description 3
- 239000005089 Luciferase Substances 0.000 description 3
- 241000124008 Mammalia Species 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 208000032136 Palmoplantar Epidermolytic Keratoderma Diseases 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical group [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 3
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 3
- 208000009415 Spinocerebellar Ataxias Diseases 0.000 description 3
- HDTRYLNUVZCQOY-WSWWMNSNSA-N Trehalose Natural products O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-WSWWMNSNSA-N 0.000 description 3
- 125000000217 alkyl group Chemical group 0.000 description 3
- HDTRYLNUVZCQOY-LIZSDCNHSA-N alpha,alpha-trehalose Chemical compound O[C@@H]1[C@@H](O)[C@H](O)[C@@H](CO)O[C@@H]1O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 HDTRYLNUVZCQOY-LIZSDCNHSA-N 0.000 description 3
- 206010002026 amyotrophic lateral sclerosis Diseases 0.000 description 3
- 201000004562 autosomal dominant cerebellar ataxia Diseases 0.000 description 3
- 210000004369 blood Anatomy 0.000 description 3
- 239000008280 blood Substances 0.000 description 3
- 230000037396 body weight Effects 0.000 description 3
- 230000003833 cell viability Effects 0.000 description 3
- 230000007812 deficiency Effects 0.000 description 3
- 238000012217 deletion Methods 0.000 description 3
- 230000037430 deletion Effects 0.000 description 3
- 238000002296 dynamic light scattering Methods 0.000 description 3
- 201000006011 epidermolytic palmoplantar keratoderma Diseases 0.000 description 3
- 238000012246 gene addition Methods 0.000 description 3
- 201000007726 hypotrichosis 6 Diseases 0.000 description 3
- 210000003405 ileum Anatomy 0.000 description 3
- 238000000338 in vitro Methods 0.000 description 3
- 238000011534 incubation Methods 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 201000004607 keratosis follicularis Diseases 0.000 description 3
- 239000007788 liquid Substances 0.000 description 3
- 238000004020 luminiscence type Methods 0.000 description 3
- 206010025135 lupus erythematosus Diseases 0.000 description 3
- 238000012986 modification Methods 0.000 description 3
- 230000006780 non-homologous end joining Effects 0.000 description 3
- 210000000056 organ Anatomy 0.000 description 3
- 238000006116 polymerization reaction Methods 0.000 description 3
- 239000000843 powder Substances 0.000 description 3
- 230000008569 process Effects 0.000 description 3
- 230000002062 proliferating effect Effects 0.000 description 3
- 150000003254 radicals Chemical class 0.000 description 3
- 235000017281 sodium acetate Nutrition 0.000 description 3
- 239000001632 sodium acetate Substances 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 230000001225 therapeutic effect Effects 0.000 description 3
- VBICKXHEKHSIBG-UHFFFAOYSA-N 1-monostearoylglycerol Chemical compound CCCCCCCCCCCCCCCCCC(=O)OCC(O)CO VBICKXHEKHSIBG-UHFFFAOYSA-N 0.000 description 2
- 208000024827 Alzheimer disease Diseases 0.000 description 2
- 102100032187 Androgen receptor Human genes 0.000 description 2
- 102100026189 Beta-galactosidase Human genes 0.000 description 2
- 206010068597 Bulbospinal muscular atrophy congenital Diseases 0.000 description 2
- 101150056204 COL7A1 gene Proteins 0.000 description 2
- 241000282832 Camelidae Species 0.000 description 2
- 108700004991 Cas12a Proteins 0.000 description 2
- 241000700198 Cavia Species 0.000 description 2
- 108010035532 Collagen Proteins 0.000 description 2
- 102000008186 Collagen Human genes 0.000 description 2
- 241000283074 Equus asinus Species 0.000 description 2
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 2
- 102000004547 Glucosylceramidase Human genes 0.000 description 2
- 108010017544 Glucosylceramidase Proteins 0.000 description 2
- 241000282412 Homo Species 0.000 description 2
- 101000775732 Homo sapiens Androgen receptor Proteins 0.000 description 2
- 101100496573 Homo sapiens COL7A1 gene Proteins 0.000 description 2
- 208000022559 Inflammatory bowel disease Diseases 0.000 description 2
- 208000027747 Kennedy disease Diseases 0.000 description 2
- 102100033448 Lysosomal alpha-glucosidase Human genes 0.000 description 2
- BAPJBEWLBFYGME-UHFFFAOYSA-N Methyl acrylate Chemical compound COC(=O)C=C BAPJBEWLBFYGME-UHFFFAOYSA-N 0.000 description 2
- 206010028095 Mucopolysaccharidosis IV Diseases 0.000 description 2
- 241000699670 Mus sp. Species 0.000 description 2
- 208000023178 Musculoskeletal disease Diseases 0.000 description 2
- 238000005481 NMR spectroscopy Methods 0.000 description 2
- 208000018737 Parkinson disease Diseases 0.000 description 2
- 201000011252 Phenylketonuria Diseases 0.000 description 2
- 201000004681 Psoriasis Diseases 0.000 description 2
- 241000700159 Rattus Species 0.000 description 2
- 102000002278 Ribosomal Proteins Human genes 0.000 description 2
- 108010000605 Ribosomal Proteins Proteins 0.000 description 2
- 206010039710 Scleroderma Diseases 0.000 description 2
- 108091027544 Subgenomic mRNA Proteins 0.000 description 2
- 108700005077 Viral Genes Proteins 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- ZSLZBFCDCINBPY-ZSJPKINUSA-N acetyl-CoA Chemical compound O[C@@H]1[C@H](OP(O)(O)=O)[C@@H](COP(O)(=O)OP(O)(=O)OCC(C)(C)[C@@H](O)C(=O)NCCC(=O)NCCSC(=O)C)O[C@H]1N1C2=NC=NC(N)=C2N=C1 ZSLZBFCDCINBPY-ZSJPKINUSA-N 0.000 description 2
- 108010070626 acid beta-galactosidase Proteins 0.000 description 2
- 238000005903 acid hydrolysis reaction Methods 0.000 description 2
- 230000009471 action Effects 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 125000003545 alkoxy group Chemical group 0.000 description 2
- 206010002022 amyloidosis Diseases 0.000 description 2
- 239000000074 antisense oligonucleotide Substances 0.000 description 2
- 238000012230 antisense oligonucleotides Methods 0.000 description 2
- 238000013459 approach Methods 0.000 description 2
- 239000007864 aqueous solution Substances 0.000 description 2
- 238000003556 assay Methods 0.000 description 2
- 230000008901 benefit Effects 0.000 description 2
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 2
- 108010005774 beta-Galactosidase Proteins 0.000 description 2
- 239000002775 capsule Substances 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 230000015271 coagulation Effects 0.000 description 2
- 238000005345 coagulation Methods 0.000 description 2
- 229920001436 collagen Polymers 0.000 description 2
- 150000001875 compounds Chemical class 0.000 description 2
- 230000008878 coupling Effects 0.000 description 2
- 238000010168 coupling process Methods 0.000 description 2
- 238000005859 coupling reaction Methods 0.000 description 2
- 239000006071 cream Substances 0.000 description 2
- 230000002950 deficient Effects 0.000 description 2
- 206010012601 diabetes mellitus Diseases 0.000 description 2
- 230000034431 double-strand break repair via homologous recombination Effects 0.000 description 2
- 208000031169 hemorrhagic disease Diseases 0.000 description 2
- 230000002209 hydrophobic effect Effects 0.000 description 2
- 125000002887 hydroxy group Chemical group [H]O* 0.000 description 2
- 208000015181 infectious disease Diseases 0.000 description 2
- 238000002347 injection Methods 0.000 description 2
- 239000007924 injection Substances 0.000 description 2
- 208000014674 injury Diseases 0.000 description 2
- 230000003993 interaction Effects 0.000 description 2
- 238000001361 intraarterial administration Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 210000004185 liver Anatomy 0.000 description 2
- 230000007774 longterm Effects 0.000 description 2
- 208000002780 macular degeneration Diseases 0.000 description 2
- 230000003211 malignant effect Effects 0.000 description 2
- 230000001404 mediated effect Effects 0.000 description 2
- 238000002156 mixing Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 208000010978 mucopolysaccharidosis type 4 Diseases 0.000 description 2
- 201000006938 muscular dystrophy Diseases 0.000 description 2
- 230000001537 neural effect Effects 0.000 description 2
- 230000000926 neurological effect Effects 0.000 description 2
- 210000002569 neuron Anatomy 0.000 description 2
- 239000002674 ointment Substances 0.000 description 2
- 230000035790 physiological processes and functions Effects 0.000 description 2
- 239000006187 pill Substances 0.000 description 2
- 238000011321 prophylaxis Methods 0.000 description 2
- 230000002829 reductive effect Effects 0.000 description 2
- 230000008439 repair process Effects 0.000 description 2
- 239000000523 sample Substances 0.000 description 2
- 230000007017 scission Effects 0.000 description 2
- 208000002491 severe combined immunodeficiency Diseases 0.000 description 2
- 208000007056 sickle cell anemia Diseases 0.000 description 2
- 239000011780 sodium chloride Substances 0.000 description 2
- 239000000725 suspension Substances 0.000 description 2
- 239000003826 tablet Substances 0.000 description 2
- 150000003512 tertiary amines Chemical group 0.000 description 2
- 238000002560 therapeutic procedure Methods 0.000 description 2
- 231100000331 toxic Toxicity 0.000 description 2
- 230000002588 toxic effect Effects 0.000 description 2
- 210000005166 vasculature Anatomy 0.000 description 2
- 230000035899 viability Effects 0.000 description 2
- 239000012224 working solution Substances 0.000 description 2
- OKAHFCCFNVPTED-UHFFFAOYSA-N 3-(2-aminoethyl)-4-methyl-1h-indole-5,6-diol;2-amino-3-methyl-4h-imidazol-5-one;sulfuric acid Chemical compound OS(O)(=O)=O.CN1CC(=O)N=C1N.CC1=C(O)C(O)=CC2=C1C(CCN)=CN2 OKAHFCCFNVPTED-UHFFFAOYSA-N 0.000 description 1
- 208000030507 AIDS Diseases 0.000 description 1
- 102000006772 Acid Ceramidase Human genes 0.000 description 1
- 108020005296 Acid Ceramidase Proteins 0.000 description 1
- 206010001052 Acute respiratory distress syndrome Diseases 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 102100031317 Alpha-N-acetylgalactosaminidase Human genes 0.000 description 1
- 102100034561 Alpha-N-acetylglucosaminidase Human genes 0.000 description 1
- 102100026277 Alpha-galactosidase A Human genes 0.000 description 1
- 208000003808 Amyloid Neuropathies Diseases 0.000 description 1
- 102000002659 Amyloid Precursor Protein Secretases Human genes 0.000 description 1
- 108010043324 Amyloid Precursor Protein Secretases Proteins 0.000 description 1
- 206010002023 Amyloidoses Diseases 0.000 description 1
- 102100022146 Arylsulfatase A Human genes 0.000 description 1
- 102100031491 Arylsulfatase B Human genes 0.000 description 1
- 206010003591 Ataxia Diseases 0.000 description 1
- 206010003805 Autism Diseases 0.000 description 1
- 208000020706 Autistic disease Diseases 0.000 description 1
- 208000000659 Autoimmune lymphoproliferative syndrome Diseases 0.000 description 1
- 208000036075 Autosomal dominant tubulointerstitial kidney disease Diseases 0.000 description 1
- 241000894006 Bacteria Species 0.000 description 1
- 206010004446 Benign prostatic hyperplasia Diseases 0.000 description 1
- 101710124976 Beta-hexosaminidase A Proteins 0.000 description 1
- 101710124978 Beta-hexosaminidase B Proteins 0.000 description 1
- 102100022548 Beta-hexosaminidase subunit alpha Human genes 0.000 description 1
- 102100032487 Beta-mannosidase Human genes 0.000 description 1
- 241000157302 Bison bison athabascae Species 0.000 description 1
- 101150029409 CFTR gene Proteins 0.000 description 1
- 241001264766 Callistemon Species 0.000 description 1
- 241000282421 Canidae Species 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 241000282693 Cercopithecidae Species 0.000 description 1
- 108010036867 Cerebroside-Sulfatase Proteins 0.000 description 1
- 241000282994 Cervidae Species 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 102100023804 Coagulation factor VII Human genes 0.000 description 1
- 206010009900 Colitis ulcerative Diseases 0.000 description 1
- 206010010099 Combined immunodeficiency Diseases 0.000 description 1
- 102000016550 Complement Factor H Human genes 0.000 description 1
- 108010053085 Complement Factor H Proteins 0.000 description 1
- 206010010356 Congenital anomaly Diseases 0.000 description 1
- 206010053138 Congenital aplastic anaemia Diseases 0.000 description 1
- 208000026372 Congenital cystic kidney disease Diseases 0.000 description 1
- 208000006069 Corneal Opacity Diseases 0.000 description 1
- 241000699800 Cricetinae Species 0.000 description 1
- 208000011231 Crohn disease Diseases 0.000 description 1
- 102000012605 Cystic Fibrosis Transmembrane Conductance Regulator Human genes 0.000 description 1
- 108010079245 Cystic Fibrosis Transmembrane Conductance Regulator Proteins 0.000 description 1
- HMFHBZSHGGEWLO-SOOFDHNKSA-N D-ribofuranose Chemical compound OC[C@H]1OC(O)[C@H](O)[C@@H]1O HMFHBZSHGGEWLO-SOOFDHNKSA-N 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 206010059352 Desmoid tumour Diseases 0.000 description 1
- 206010012689 Diabetic retinopathy Diseases 0.000 description 1
- 206010013801 Duchenne Muscular Dystrophy Diseases 0.000 description 1
- 208000011345 Duchenne and Becker muscular dystrophy Diseases 0.000 description 1
- 206010013883 Dwarfism Diseases 0.000 description 1
- 206010058314 Dysplasia Diseases 0.000 description 1
- 206010014561 Emphysema Diseases 0.000 description 1
- 201000009273 Endometriosis Diseases 0.000 description 1
- 241000283073 Equus caballus Species 0.000 description 1
- 241000359025 Equus kiang Species 0.000 description 1
- 241000283070 Equus zebra Species 0.000 description 1
- IAYPIBMASNFSPL-UHFFFAOYSA-N Ethylene oxide Chemical compound C1CO1 IAYPIBMASNFSPL-UHFFFAOYSA-N 0.000 description 1
- 108091029865 Exogenous DNA Proteins 0.000 description 1
- 208000024720 Fabry Disease Diseases 0.000 description 1
- 108010014172 Factor V Proteins 0.000 description 1
- 108010023321 Factor VII Proteins 0.000 description 1
- 108010054218 Factor VIII Proteins 0.000 description 1
- 102000001690 Factor VIII Human genes 0.000 description 1
- 201000003542 Factor VIII deficiency Diseases 0.000 description 1
- 108010014173 Factor X Proteins 0.000 description 1
- 108010074864 Factor XI Proteins 0.000 description 1
- 108010080865 Factor XII Proteins 0.000 description 1
- 102000000429 Factor XII Human genes 0.000 description 1
- 201000004939 Fanconi anemia Diseases 0.000 description 1
- 208000001948 Farber Lipogranulomatosis Diseases 0.000 description 1
- 208000033149 Farber disease Diseases 0.000 description 1
- 206010016654 Fibrosis Diseases 0.000 description 1
- WSFSSNUMVMOOMR-UHFFFAOYSA-N Formaldehyde Chemical compound O=C WSFSSNUMVMOOMR-UHFFFAOYSA-N 0.000 description 1
- 208000001914 Fragile X syndrome Diseases 0.000 description 1
- 102100028496 Galactocerebrosidase Human genes 0.000 description 1
- 108010042681 Galactosylceramidase Proteins 0.000 description 1
- 208000009796 Gangliosidoses Diseases 0.000 description 1
- 208000015872 Gaucher disease Diseases 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 206010064571 Gene mutation Diseases 0.000 description 1
- 229940123611 Genome editing Drugs 0.000 description 1
- 241000282818 Giraffidae Species 0.000 description 1
- 208000010412 Glaucoma Diseases 0.000 description 1
- 208000010055 Globoid Cell Leukodystrophy Diseases 0.000 description 1
- 206010018364 Glomerulonephritis Diseases 0.000 description 1
- 102000053187 Glucuronidase Human genes 0.000 description 1
- 108010060309 Glucuronidase Proteins 0.000 description 1
- 208000032007 Glycogen storage disease due to acid maltase deficiency Diseases 0.000 description 1
- 206010053185 Glycogen storage disease type II Diseases 0.000 description 1
- 208000003807 Graves Disease Diseases 0.000 description 1
- 208000015023 Graves' disease Diseases 0.000 description 1
- 208000035895 Guillain-Barré syndrome Diseases 0.000 description 1
- 206010018852 Haematoma Diseases 0.000 description 1
- 208000030836 Hashimoto thyroiditis Diseases 0.000 description 1
- 208000018565 Hemochromatosis Diseases 0.000 description 1
- 206010019629 Hepatic adenoma Diseases 0.000 description 1
- 206010019663 Hepatic failure Diseases 0.000 description 1
- 102000019267 Hepatic lipases Human genes 0.000 description 1
- 108050006747 Hepatic lipases Proteins 0.000 description 1
- 208000003923 Hereditary Corneal Dystrophies Diseases 0.000 description 1
- 208000032672 Histiocytosis haematophagic Diseases 0.000 description 1
- 241001272567 Hominoidea Species 0.000 description 1
- 206010020649 Hyperkeratosis Diseases 0.000 description 1
- 206010061598 Immunodeficiency Diseases 0.000 description 1
- 208000029462 Immunodeficiency disease Diseases 0.000 description 1
- 108090000862 Ion Channels Proteins 0.000 description 1
- 102000004310 Ion Channels Human genes 0.000 description 1
- 208000001126 Keratosis Diseases 0.000 description 1
- 206010023421 Kidney fibrosis Diseases 0.000 description 1
- 208000028226 Krabbe disease Diseases 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 201000003533 Leber congenital amaurosis Diseases 0.000 description 1
- 206010025219 Lymphangioma Diseases 0.000 description 1
- 208000002569 Machado-Joseph Disease Diseases 0.000 description 1
- 208000035719 Maculopathy Diseases 0.000 description 1
- 208000002720 Malnutrition Diseases 0.000 description 1
- 208000001826 Marfan syndrome Diseases 0.000 description 1
- 201000011442 Metachromatic leukodystrophy Diseases 0.000 description 1
- 206010054949 Metaplasia Diseases 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- CERQOIWHTDAKMF-UHFFFAOYSA-M Methacrylate Chemical compound CC(=C)C([O-])=O CERQOIWHTDAKMF-UHFFFAOYSA-M 0.000 description 1
- 206010049567 Miller Fisher syndrome Diseases 0.000 description 1
- 208000008955 Mucolipidoses Diseases 0.000 description 1
- 206010072927 Mucolipidosis type I Diseases 0.000 description 1
- 206010056893 Mucopolysaccharidosis VII Diseases 0.000 description 1
- 208000025915 Mucopolysaccharidosis type 6 Diseases 0.000 description 1
- 206010068871 Myotonic dystrophy Diseases 0.000 description 1
- 108010027520 N-Acetylgalactosamine-4-Sulfatase Proteins 0.000 description 1
- GXCLVBGFBYZDAG-UHFFFAOYSA-N N-[2-(1H-indol-3-yl)ethyl]-N-methylprop-2-en-1-amine Chemical compound CN(CCC1=CNC2=C1C=CC=C2)CC=C GXCLVBGFBYZDAG-UHFFFAOYSA-N 0.000 description 1
- 102100031688 N-acetylgalactosamine-6-sulfatase Human genes 0.000 description 1
- 101710099863 N-acetylgalactosamine-6-sulfatase Proteins 0.000 description 1
- 108010023320 N-acetylglucosamine-6-sulfatase Proteins 0.000 description 1
- 101710202061 N-acetyltransferase Proteins 0.000 description 1
- 208000012902 Nervous system disease Diseases 0.000 description 1
- 102000005348 Neuraminidase Human genes 0.000 description 1
- 108010006232 Neuraminidase Proteins 0.000 description 1
- 208000009905 Neurofibromatoses Diseases 0.000 description 1
- 208000014060 Niemann-Pick disease Diseases 0.000 description 1
- 102000011931 Nucleoproteins Human genes 0.000 description 1
- 108010061100 Nucleoproteins Proteins 0.000 description 1
- 208000022873 Ocular disease Diseases 0.000 description 1
- 241000283973 Oryctolagus cuniculus Species 0.000 description 1
- 240000007594 Oryza sativa Species 0.000 description 1
- 235000007164 Oryza sativa Nutrition 0.000 description 1
- 206010031243 Osteogenesis imperfecta Diseases 0.000 description 1
- 208000001132 Osteoporosis Diseases 0.000 description 1
- 241000282579 Pan Species 0.000 description 1
- 241000282320 Panthera leo Species 0.000 description 1
- 241000282376 Panthera tigris Species 0.000 description 1
- 208000031481 Pathologic Constriction Diseases 0.000 description 1
- 235000019483 Peanut oil Nutrition 0.000 description 1
- 241001494479 Pecora Species 0.000 description 1
- 241000721454 Pemphigus Species 0.000 description 1
- 208000031845 Pernicious anaemia Diseases 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical group [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 239000004793 Polystyrene Substances 0.000 description 1
- 241000282405 Pongo abelii Species 0.000 description 1
- 241000288906 Primates Species 0.000 description 1
- 208000024777 Prion disease Diseases 0.000 description 1
- 208000004403 Prostatic Hyperplasia Diseases 0.000 description 1
- 208000033464 Reiter syndrome Diseases 0.000 description 1
- 208000013616 Respiratory Distress Syndrome Diseases 0.000 description 1
- 208000017442 Retinal disease Diseases 0.000 description 1
- 206010038923 Retinopathy Diseases 0.000 description 1
- PYMYPHUHKUWMLA-LMVFSUKVSA-N Ribose Natural products OC[C@@H](O)[C@@H](O)[C@@H](O)C=O PYMYPHUHKUWMLA-LMVFSUKVSA-N 0.000 description 1
- 241000283984 Rodentia Species 0.000 description 1
- 208000021811 Sandhoff disease Diseases 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N Silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 208000021386 Sjogren Syndrome Diseases 0.000 description 1
- 108010061312 Sphingomyelin Phosphodiesterase Proteins 0.000 description 1
- 208000036834 Spinocerebellar ataxia type 3 Diseases 0.000 description 1
- 229920002472 Starch Polymers 0.000 description 1
- 229930006000 Sucrose Natural products 0.000 description 1
- CZMRCDWAGMRECN-UGDNZRGBSA-N Sucrose Chemical compound O[C@H]1[C@H](O)[C@@H](CO)O[C@@]1(CO)O[C@@H]1[C@H](O)[C@@H](O)[C@H](O)[C@@H](CO)O1 CZMRCDWAGMRECN-UGDNZRGBSA-N 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- 201000009594 Systemic Scleroderma Diseases 0.000 description 1
- 206010042953 Systemic sclerosis Diseases 0.000 description 1
- 208000022292 Tay-Sachs disease Diseases 0.000 description 1
- 206010043395 Thalassaemia sickle cell Diseases 0.000 description 1
- 208000002903 Thalassemia Diseases 0.000 description 1
- 108091028113 Trans-activating crRNA Proteins 0.000 description 1
- 201000006704 Ulcerative Colitis Diseases 0.000 description 1
- 206010046851 Uveitis Diseases 0.000 description 1
- 208000024248 Vascular System injury Diseases 0.000 description 1
- 208000012339 Vascular injury Diseases 0.000 description 1
- 206010053648 Vascular occlusion Diseases 0.000 description 1
- 206010047642 Vitiligo Diseases 0.000 description 1
- 208000027418 Wounds and injury Diseases 0.000 description 1
- 238000002679 ablation Methods 0.000 description 1
- 230000002159 abnormal effect Effects 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 102000010126 acid sphingomyelin phosphodiesterase activity proteins Human genes 0.000 description 1
- 230000002378 acidificating effect Effects 0.000 description 1
- 208000009621 actinic keratosis Diseases 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 208000011341 adult acute respiratory distress syndrome Diseases 0.000 description 1
- 201000000028 adult respiratory distress syndrome Diseases 0.000 description 1
- 206010064930 age-related macular degeneration Diseases 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 208000026935 allergic disease Diseases 0.000 description 1
- HMFHBZSHGGEWLO-UHFFFAOYSA-N alpha-D-Furanose-Ribose Natural products OCC1OC(O)C(O)C1O HMFHBZSHGGEWLO-UHFFFAOYSA-N 0.000 description 1
- 108010030291 alpha-Galactosidase Proteins 0.000 description 1
- 108010028144 alpha-Glucosidases Proteins 0.000 description 1
- 102000012086 alpha-L-Fucosidase Human genes 0.000 description 1
- 108010061314 alpha-L-Fucosidase Proteins 0.000 description 1
- 108010012864 alpha-Mannosidase Proteins 0.000 description 1
- 102000019199 alpha-Mannosidase Human genes 0.000 description 1
- 108010015684 alpha-N-Acetylgalactosaminidase Proteins 0.000 description 1
- 108010009380 alpha-N-acetyl-D-glucosaminidase Proteins 0.000 description 1
- 201000008333 alpha-mannosidosis Diseases 0.000 description 1
- 208000007502 anemia Diseases 0.000 description 1
- 238000002399 angioplasty Methods 0.000 description 1
- 239000003125 aqueous solvent Substances 0.000 description 1
- 125000003118 aryl group Chemical group 0.000 description 1
- 125000004429 atom Chemical group 0.000 description 1
- 230000003416 augmentation Effects 0.000 description 1
- 108010055059 beta-Mannosidase Proteins 0.000 description 1
- 201000006486 beta-mannosidosis Diseases 0.000 description 1
- 230000001588 bifunctional effect Effects 0.000 description 1
- 210000000013 bile duct Anatomy 0.000 description 1
- 210000000601 blood cell Anatomy 0.000 description 1
- 210000002798 bone marrow cell Anatomy 0.000 description 1
- 239000007853 buffer solution Substances 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 230000000747 cardiac effect Effects 0.000 description 1
- 239000000969 carrier Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000004113 cell culture Methods 0.000 description 1
- 230000004700 cellular uptake Effects 0.000 description 1
- 125000003636 chemical group Chemical group 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 239000003153 chemical reaction reagent Substances 0.000 description 1
- 230000007882 cirrhosis Effects 0.000 description 1
- 208000019425 cirrhosis of liver Diseases 0.000 description 1
- 230000000052 comparative effect Effects 0.000 description 1
- 238000010668 complexation reaction Methods 0.000 description 1
- 239000007891 compressed tablet Substances 0.000 description 1
- 230000021615 conjugation Effects 0.000 description 1
- 201000003046 cornea plana Diseases 0.000 description 1
- 206010011005 corneal dystrophy Diseases 0.000 description 1
- 238000012937 correction Methods 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 235000013365 dairy product Nutrition 0.000 description 1
- 230000006378 damage Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000001934 delay Effects 0.000 description 1
- 239000000412 dendrimer Substances 0.000 description 1
- 229920000736 dendritic polymer Polymers 0.000 description 1
- 239000007933 dermal patch Substances 0.000 description 1
- 238000011161 development Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 239000008121 dextrose Substances 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 239000003085 diluting agent Substances 0.000 description 1
- 238000004090 dissolution Methods 0.000 description 1
- 239000002552 dosage form Substances 0.000 description 1
- 239000006196 drop Substances 0.000 description 1
- 239000003937 drug carrier Substances 0.000 description 1
- 239000000890 drug combination Substances 0.000 description 1
- 238000012377 drug delivery Methods 0.000 description 1
- 229940126534 drug product Drugs 0.000 description 1
- 238000010410 dusting Methods 0.000 description 1
- 230000008482 dysregulation Effects 0.000 description 1
- 230000009881 electrostatic interaction Effects 0.000 description 1
- 210000002257 embryonic structure Anatomy 0.000 description 1
- 239000003995 emulsifying agent Substances 0.000 description 1
- 239000003623 enhancer Substances 0.000 description 1
- 210000002919 epithelial cell Anatomy 0.000 description 1
- 230000008029 eradication Effects 0.000 description 1
- 125000001033 ether group Chemical group 0.000 description 1
- 238000011156 evaluation Methods 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 238000002474 experimental method Methods 0.000 description 1
- 229940012413 factor vii Drugs 0.000 description 1
- 229960000301 factor viii Drugs 0.000 description 1
- 229940012426 factor x Drugs 0.000 description 1
- 206010016629 fibroma Diseases 0.000 description 1
- 206010049444 fibromatosis Diseases 0.000 description 1
- 235000013312 flour Nutrition 0.000 description 1
- 235000013305 food Nutrition 0.000 description 1
- 239000012634 fragment Substances 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 239000012520 frozen sample Substances 0.000 description 1
- 201000008049 fucosidosis Diseases 0.000 description 1
- 201000006440 gangliosidosis Diseases 0.000 description 1
- 210000001035 gastrointestinal tract Anatomy 0.000 description 1
- 239000000499 gel Substances 0.000 description 1
- 239000008273 gelatin Substances 0.000 description 1
- 229920000159 gelatin Polymers 0.000 description 1
- 235000019322 gelatine Nutrition 0.000 description 1
- 235000011852 gelatine desserts Nutrition 0.000 description 1
- 238000003209 gene knockout Methods 0.000 description 1
- 230000030279 gene silencing Effects 0.000 description 1
- 238000012226 gene silencing method Methods 0.000 description 1
- 238000012637 gene transfection Methods 0.000 description 1
- 210000004602 germ cell Anatomy 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- YQEMORVAKMFKLG-UHFFFAOYSA-N glycerine monostearate Natural products CCCCCCCCCCCCCCCCCC(=O)OC(CO)CO YQEMORVAKMFKLG-UHFFFAOYSA-N 0.000 description 1
- SVUQHVRAGMNPLW-UHFFFAOYSA-N glycerol monostearate Natural products CCCCCCCCCCCCCCCCC(=O)OCC(O)CO SVUQHVRAGMNPLW-UHFFFAOYSA-N 0.000 description 1
- 201000004502 glycogen storage disease II Diseases 0.000 description 1
- 201000008977 glycoproteinosis Diseases 0.000 description 1
- 230000012010 growth Effects 0.000 description 1
- 239000001963 growth medium Substances 0.000 description 1
- 230000036541 health Effects 0.000 description 1
- 208000009429 hemophilia B Diseases 0.000 description 1
- 108010089932 heparan sulfate sulfatase Proteins 0.000 description 1
- 208000006359 hepatoblastoma Diseases 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 230000003463 hyperproliferative effect Effects 0.000 description 1
- 208000022368 idiopathic cardiomyopathy Diseases 0.000 description 1
- 210000002865 immune cell Anatomy 0.000 description 1
- 210000000987 immune system Anatomy 0.000 description 1
- 230000007813 immunodeficiency Effects 0.000 description 1
- 230000001771 impaired effect Effects 0.000 description 1
- 230000004054 inflammatory process Effects 0.000 description 1
- 208000030309 inherited neurodegenerative disease Diseases 0.000 description 1
- 230000002401 inhibitory effect Effects 0.000 description 1
- 239000003999 initiator Substances 0.000 description 1
- 238000002743 insertional mutagenesis Methods 0.000 description 1
- 230000010354 integration Effects 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000007794 irritation Effects 0.000 description 1
- 230000003780 keratinization Effects 0.000 description 1
- 210000003292 kidney cell Anatomy 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 210000000265 leukocyte Anatomy 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 229940057995 liquid paraffin Drugs 0.000 description 1
- 208000019423 liver disease Diseases 0.000 description 1
- 208000007903 liver failure Diseases 0.000 description 1
- 231100000835 liver failure Toxicity 0.000 description 1
- 230000033001 locomotion Effects 0.000 description 1
- 230000004777 loss-of-function mutation Effects 0.000 description 1
- 239000006210 lotion Substances 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 230000002132 lysosomal effect Effects 0.000 description 1
- 206010025482 malaise Diseases 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000010034 metabolic health Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 230000015689 metaplastic ossification Effects 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 239000011859 microparticle Substances 0.000 description 1
- 239000002480 mineral oil Substances 0.000 description 1
- 235000010446 mineral oil Nutrition 0.000 description 1
- 238000009126 molecular therapy Methods 0.000 description 1
- 208000025919 mucopolysaccharidosis type 7 Diseases 0.000 description 1
- 201000006417 multiple sclerosis Diseases 0.000 description 1
- 210000003205 muscle Anatomy 0.000 description 1
- 201000000585 muscular atrophy Diseases 0.000 description 1
- 238000002703 mutagenesis Methods 0.000 description 1
- 231100000350 mutagenesis Toxicity 0.000 description 1
- 206010028417 myasthenia gravis Diseases 0.000 description 1
- 229920005615 natural polymer Polymers 0.000 description 1
- 239000013642 negative control Substances 0.000 description 1
- 210000001577 neostriatum Anatomy 0.000 description 1
- 230000004770 neurodegeneration Effects 0.000 description 1
- 208000015122 neurodegenerative disease Diseases 0.000 description 1
- 201000004931 neurofibromatosis Diseases 0.000 description 1
- 230000016273 neuron death Effects 0.000 description 1
- 239000002773 nucleotide Substances 0.000 description 1
- 125000003729 nucleotide group Chemical group 0.000 description 1
- 235000018343 nutrient deficiency Nutrition 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 238000005457 optimization Methods 0.000 description 1
- 208000025661 ovarian cyst Diseases 0.000 description 1
- RPQRDASANLAFCM-UHFFFAOYSA-N oxiran-2-ylmethyl prop-2-enoate Chemical compound C=CC(=O)OCC1CO1 RPQRDASANLAFCM-UHFFFAOYSA-N 0.000 description 1
- 229910052760 oxygen Inorganic materials 0.000 description 1
- 125000004430 oxygen atom Chemical group O* 0.000 description 1
- 239000006179 pH buffering agent Substances 0.000 description 1
- 239000012188 paraffin wax Substances 0.000 description 1
- 230000001575 pathological effect Effects 0.000 description 1
- 230000007170 pathology Effects 0.000 description 1
- 239000000312 peanut oil Substances 0.000 description 1
- 230000035515 penetration Effects 0.000 description 1
- 239000003208 petroleum Substances 0.000 description 1
- 239000000825 pharmaceutical preparation Substances 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 206010035116 pityriasis rubra pilaris Diseases 0.000 description 1
- 239000013612 plasmid Substances 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 231100000572 poisoning Toxicity 0.000 description 1
- 230000000607 poisoning effect Effects 0.000 description 1
- 229920002189 poly(glycerol 1-O-monomethacrylate) polymer Polymers 0.000 description 1
- 229920000058 polyacrylate Polymers 0.000 description 1
- 208000030761 polycystic kidney disease Diseases 0.000 description 1
- 229920000155 polyglutamine Polymers 0.000 description 1
- 108010040003 polyglutamine Proteins 0.000 description 1
- 239000002861 polymer material Substances 0.000 description 1
- 230000000379 polymerizing effect Effects 0.000 description 1
- 208000005987 polymyositis Diseases 0.000 description 1
- 229920002223 polystyrene Polymers 0.000 description 1
- 239000013641 positive control Substances 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 150000003141 primary amines Chemical class 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 230000001681 protective effect Effects 0.000 description 1
- 208000005069 pulmonary fibrosis Diseases 0.000 description 1
- 208000002574 reactive arthritis Diseases 0.000 description 1
- 230000006798 recombination Effects 0.000 description 1
- 238000005215 recombination Methods 0.000 description 1
- 238000003168 reconstitution method Methods 0.000 description 1
- 230000009467 reduction Effects 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 208000037803 restenosis Diseases 0.000 description 1
- 230000000284 resting effect Effects 0.000 description 1
- 206010039073 rheumatoid arthritis Diseases 0.000 description 1
- 235000009566 rice Nutrition 0.000 description 1
- 229920002477 rna polymer Polymers 0.000 description 1
- 201000000306 sarcoidosis Diseases 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 201000000980 schizophrenia Diseases 0.000 description 1
- 239000008159 sesame oil Substances 0.000 description 1
- 235000011803 sesame oil Nutrition 0.000 description 1
- 208000011985 sialidosis Diseases 0.000 description 1
- 239000000741 silica gel Substances 0.000 description 1
- 229910002027 silica gel Inorganic materials 0.000 description 1
- 235000020183 skimmed milk Nutrition 0.000 description 1
- 210000004927 skin cell Anatomy 0.000 description 1
- 210000002460 smooth muscle Anatomy 0.000 description 1
- RYYKJJJTJZKILX-UHFFFAOYSA-M sodium octadecanoate Chemical compound [Na+].CCCCCCCCCCCCCCCCCC([O-])=O RYYKJJJTJZKILX-UHFFFAOYSA-M 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 210000001082 somatic cell Anatomy 0.000 description 1
- 239000003549 soybean oil Substances 0.000 description 1
- 235000012424 soybean oil Nutrition 0.000 description 1
- 230000002269 spontaneous effect Effects 0.000 description 1
- 239000007921 spray Substances 0.000 description 1
- 230000006641 stabilisation Effects 0.000 description 1
- 239000003381 stabilizer Substances 0.000 description 1
- 239000008107 starch Substances 0.000 description 1
- 235000019698 starch Nutrition 0.000 description 1
- 230000036262 stenosis Effects 0.000 description 1
- 208000037804 stenosis Diseases 0.000 description 1
- 238000003860 storage Methods 0.000 description 1
- 238000007920 subcutaneous administration Methods 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 239000005720 sucrose Substances 0.000 description 1
- 230000009469 supplementation Effects 0.000 description 1
- 238000001356 surgical procedure Methods 0.000 description 1
- 230000004083 survival effect Effects 0.000 description 1
- 238000013268 sustained release Methods 0.000 description 1
- 239000012730 sustained-release form Substances 0.000 description 1
- 208000011580 syndromic disease Diseases 0.000 description 1
- 230000009885 systemic effect Effects 0.000 description 1
- 239000000454 talc Substances 0.000 description 1
- 229910052623 talc Inorganic materials 0.000 description 1
- 238000011200 topical administration Methods 0.000 description 1
- 230000001988 toxicity Effects 0.000 description 1
- 231100000419 toxicity Toxicity 0.000 description 1
- 239000012096 transfection reagent Substances 0.000 description 1
- 238000004627 transmission electron microscopy Methods 0.000 description 1
- 230000008733 trauma Effects 0.000 description 1
- 208000021331 vascular occlusion disease Diseases 0.000 description 1
- 235000013311 vegetables Nutrition 0.000 description 1
- 239000003981 vehicle Substances 0.000 description 1
- 238000009736 wetting Methods 0.000 description 1
- 239000000080 wetting agent Substances 0.000 description 1
- 229940045860 white wax Drugs 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/58—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/59—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
- A61K47/595—Polyamides, e.g. nylon
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/59—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
- A61K47/60—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/61—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
- A61K47/6921—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
- A61K47/6927—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
- A61K47/6929—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
- A61K47/6931—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
- A61K47/6933—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained by reactions only involving carbon to carbon, e.g. poly(meth)acrylate, polystyrene, polyvinylpyrrolidone or polyvinylalcohol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
- A61K47/6921—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
- A61K47/6927—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
- A61K47/6929—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
- A61K47/6931—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
- A61K47/6935—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being obtained otherwise than by reactions involving carbon to carbon unsaturated bonds, e.g. polyesters, polyamides or polyglycerol
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
- A61K47/6921—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
- A61K47/6927—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
- A61K47/6929—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
- A61K47/6931—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
- A61K47/6939—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer the polymer being a polysaccharide, e.g. starch, chitosan, chitin, cellulose or pectin
-
- 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
- C12N15/87—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
- C12N15/88—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
Definitions
- Nanoparticulate compositions for gene therapy are described.
- the present invention relates to a nanoparticulate composition for gene therapy. Also contemplated are methods of treating skin genetic disorders such as Recessive dystrophic epidermolysis bullosa (RDEB).
- RDEB Recessive dystrophic epidermolysis bullosa
- polymer-based and lipid-based vectors are the two most common vectors, which carry distinct advantages, but both contribute to gene therapy development.
- PAEs poly( ⁇ -amino ester)s
- RNA also called large RNA, mainly include messenger RNA (mRNA) and long non-coding RNA (IncRNA), are the typical nucleic acids susceptible to auto- hydrolysis, even without the presence of enzymes or any specific condition.
- mRNA messenger RNA
- IncRNA long non-coding RNA
- RNA and double strands DNA are much more stable due to double strands structures or lack of 2' OH groups, but they still can be susceptible to hydrolysis under acidic conditions with the presence of enzymes or other particular conditions.
- Polymers for delivery of DNA are described in WANG YAO ET AL ( “Effects of Branching Strategy on the Gene Transfection of Highly Branched Poly(-amino ester)s”, CHINESE JOURNAL OF POLYMER SCIENCE, CHINESE CHEMICAL SOCIETY AND INSTITUTE OF CHEMISTRY, CAS, BEIJING, vol. 38, no.
- anti-cleavage units which have not been described before, incorporated into polymers, can encapsulate nucleic acids and form a stable-particle that presents a high stability against acidic hydrolysis, enzyme hydrolysis, and especially, auto- hydrolysis, without the need for inclusion of lipids to form a hydrophobic environment.
- the stable-particle has been found to be capable of delivering long RNA and, similarly, stabilised delivery of other nucleic acids.
- the polymers specifically carry diamine moieties (anti-cleavage units) that can both condense the nucleic acids via electrostatic interaction and protect each phosphodiester bond from hydrolysis and loss of function.
- Example of suitable anti-cleavage units are provided in Figure 1.
- the Applicant has also discovered that polymers having about 10 to about 150 diamine moieties per polymer backbone provides for excellent mRNA protection and transfection efficacy (Figure 9).
- the invention provides a composition comprising a nucleic acid complexed within a polymer, wherein the polymer comprises a diamine moiety incorporated into the polymer as a terminal group, and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone.
- the hydrocarbon backbone is a saturated hydrocarbon backbone. In any embodiment, the hydrocarbon backbone has 3, 4 or 5 carbon atoms, with or without hydrocarbon branching from the hydrocarbon backbone.
- the polymer comprises the plurality of diamine moieties as terminal endcap groups.
- the polymer comprises the plurality of diamine moieties as polymer side-chain terminal groups.
- the polymer comprises 5 to 200 diamine moieties per polymer backbone.
- the polymer comprises about 10 to about 150 diamine moieties per polymer backbone.
- the polymer is a cationic polymer.
- the polymer is a linear polymer.
- the polymer is a branched polymer.
- the polymer is a hyperbranched polymer.
- the hyperbranched polymer is a 3-branching or 4-branching hyperbranched polymer.
- the polymer is a comb polymer.
- the polymer is a brush polymer.
- the polymer is a poly(beta amino ester) polymer (hereafter “PAE polymer”).
- the polymer is a PEG polymer. In any embodiment, the polymer is a poly(glycidyl methacrylate) polymer.
- the polymer is a hyaluronic acid polymer.
- the polymer is a poly(amidoamine) polymer (hereafter “PAMAM polymer”).
- the polymer is an oligomer combination of a PAE and PEG polymers.
- each diamine terminal group each, independently, has a chemical formula R1-L1-R2: in which:
- R1 and R2 are each independently selected from a primary, secondary or tertiary amine group
- L1 is a linker
- L1 is a straight or branched, optionally substituted, hydrocarbon chain having 3, 4 or 5 carbon atoms.
- L1 is saturated.
- L1 is selected from the group consisting of: CH 2 -(CH 2 )y-CH(X1)-CH 2 ; CH 2 -CH(X2)-(CH 2 )y-CH 2 ; CH 2 -CH(X2)-CH(X1)-CH 2 ; CH(X3)-CH(X2)-CH(X1)-CH 2 ; and, CH(X3)-CH(X2)-CH(X1)-CH(X4); wherein, y is at each occurrence 0, 1 or 2;
- X1 is independently selected from the group consisting of: hydrogen, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 and -COOH;
- X2 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 and -COOH;
- X3 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 and -COOH;
- X4 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 and -COOH.
- L1 is selected from the group consisting of: (CH 2 ) 3 ; (CH 2 ) 4 , (CH 2 ) 5 ,CH 2 - CH 2 -CH(CH 3 )-CH 2 ; CH 2 -CH(CH 3 )-CH(CH 3 )-CH 2 ; CH(CH 3 )-CH(CH 3 )-CH(CH 3 )-CH 2 ; CH(CH 3 )-CH(CH 3 )-CH(CH 3 )-CH(CH 3 ); and, CH(CH 3 )-CH 2 -CH 2 -CH 2 .
- L1 is selected from the group consisting of: (CH 2 ) 3 ; CH 2 -CH 2 -CH(CH 3 )- CH 2 .
- R1 has the structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group consisting of: H and CH 2 Z1 ; wherein
- Z1 is independent selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH 2 OH, -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 , -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R1 ’ and R1 ” are the same or different.
- R1 has a structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group consisting of: H; CH 3 ; CH 2 OH; CH 2 CH 2 OH; and a polymer, wherein R1 ’ and R1 ” are the same or different.
- R2 has the structure R2’-N-R2”, in which R2’ and R2” are each independently selected form the group consisting of: H and CH 2 Z2; wherein
- Z2 is independently selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH 2 OH, -F, -Cl, -Br, -I, -SH, -N 3 , -NO 2 , -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R2’ and R2” are the same or different.
- R2 has a structure selected from:
- R2’-N-R2 in which R2’ and R2” are each independently selected from the group consisting of: H; CH 3 ; CH 2 OH; CH 2 CH 2 OH; and a polymer; and a heterocyclic group containing two ring nitrogen atoms; wherein R2’ and R2” are the same or different.
- R2, or R1 , or R2 and R1 have a structure selected from: a substituted or unsubstituted piperazinyl group; a substituted or unsubstituted piperidinyl group; a substituted or unsubstituted pyrrolidinyl group; a substituted or unsubstituted pyrazolidinyl group; and a substituted or unsubstituted imidazolidinyl group.
- each diamine terminal group each, independently, has a chemical formula R1-L1-R2: in which:
- R1 has a structure R1 ’-N-R1 ”, in which R1’ and R1 ” are each independently selected from the group consisting of: H; CH 3 ; CH 2 OH; CH 2 CH 2 OH; and a polymer;
- L1 is selected from the group consisting of: (CH 2 ) 3 ; and CH 2 -CH 2 -CH(CH 3 )CH 2 ;
- R2 has a structure selected from:
- R2’-N-R2 in which R2’ and R2” are each independently selected from the group consisting of: H; CH 3 ; CH 2 OH; CH 2 CH 2 OH; and a polymer; and a heterocyclic group containing two ring nitrogen atoms.
- the heterocyclic group containing two ring nitrogen atoms has a structure:
- L1 does not contain an ether group.
- the diamine terminal group has a molecular weight of less than 2000 Da, 1500 Da, 1000 Da, 500 Da, 400 Da, 300 Da, 250 Da or 200 Da.
- the composition is a non-viral gene delivery vector. In any embodiment, the composition is substantially free of lipid.
- the nucleic acid is a single stranded nucleic acid.
- the nucleic acid is an RNA molecule.
- the RNA molecule is a long RNA molecule.
- the long RNA molecule is selected from a messenger RNA molecule (mRNA) and a long non-coding RNA molecule (IncRNA).
- mRNA messenger RNA molecule
- IncRNA long non-coding RNA molecule
- the composition is a pharmaceutical composition and comprises a a suitable pharmaceutical excipient.
- the composition is microparticulate.
- the composition is nanoparticulate.
- the nucleic acid is provided in the form of a gene editing ribonucleoprotein system.
- the gene editing ribonucleoprotein system is a Cas9-gRNA ribonucleoprotein system, such as a CRISPR-Cas9 gene editing system, which is typically configured to induce deletion of a targeted genomic sequence including excision of a mutation or exon in a gene, replace a mutation in a gene, or produce a knock-down or knock-out of a gene.
- CRISPR-Cas9 gene editing system such as a CRISPR-Cas9 gene editing system
- Other gene editing ribonucleoprotein systems include for example alternative CRISPR-Cas derivatives such as Cas12a, Cas14, CRISPR Base editors, zinc finger nuclease systems and TALEN systems.
- the gene editing ribonucleoprotein system is a CRISPR- Cas9 gene editing system.
- the gene editing ribonucleoprotein system is configured for exon- excision.
- the gene editing ribonucleoprotein system is a CRISPR-Cas9 gene editing system.
- the gene editing ribonucleoprotein system is configured to excise exon 80 of the COL7A1 gene encoding for Collagen VII protein.
- the nanoparticulate composition has an average dimension of 50-500, 50-400, 50-300, 100-400, 100-300, 150-250, and ideally about 200 nm.
- Methods of measuring the average size of the nanoparticulate compositions are for example a dynamic light scattering system or transmission electron microscopy.
- PDI polydispersity index
- a Malvern Zetasizer Nano ZS equipped with a scattering angle of 173° can be used. Nanoparticulate size measurements are performed in a clear plastic disposable cuvette.
- the invention also provides a method of making a composition of the invention comprising the steps of: providing a solution of nucleic acid in a buffer; providing a solution of polymer in a suitable non-aqueous solvent, in which the polymer comprises a diamine moiety incorporated into the polymer as a terminal group; and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone; and mixing the solutions such that there is optionally an excess of mass of the polymer over that of the nucleic acid in the mixture; and typically resting the mixture to allow the composition to form.
- the polymer solution is prepared by dissolving the polymer in an alcohol such as ethanol.
- the polymer is dissolved in the solvent at a concentration of 10-200 mg/ml, preferably 50-150 mg/ml, and ideally about 100 mg/ml.
- the polymer solution comprises 0.1 to 100 g of polymer. system typically containing 0.1 to 100 ⁇ g of ribonucleoprotein complex.
- the nucleic acid buffer is sodium acetate.
- the first and second solutions are mixed at a volumetric ratio of about 1-100:1-100, such that there is an excess of mass of the polymer over that of the nucleic acid.
- the amount of polymer in the second solution is 1 to 100 times more than the nucleic acid in terms of mass, and in which the first and second solutions are mixed at a volumetric ratio of about 1-100:1-100.
- the polymer comprises 10 to 150 diamine moieties per polymer backbone.
- the invention also provides a composition of the invention, for use in a method of treatment of a genetic disease in a subject, in which a therapeutically effective amount of the composition is administered to the subject.
- the genetic disease is characterised by a mutation in a gene of the individual.
- the nucleic acid is configured to edit a gene of the individual. Editing may comprise non-homologous end joining (NHEJ) (i.e. for large or small genomic deletions or exon excision), knock down or knock out a gene, and homology direct repair (HDR).
- NHEJ non-homologous end joining
- HDR homology direct repair
- the editing comprises deleting or replacing the mutation or a section of the gene including the mutation.
- the composition is administered topically or by sub-dermal injection.
- the genetic disease is selected from a skin genetic disorder.
- skin genetic disorder examples include Epidermolysis Bullosa (EB), Recessive dystrophic epidermolysis bullosa (RDEB), Epidermolytic Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease, Localized Autosomal Recessive Hypotrichosis.
- Additional skin diseases may include: alternative EB subtypes such as Simplex EB and Junctional EB, Epidermolytic Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease and, Localized Autosomal Recessive Hypotrichosis,
- the genetic disease is Recessive Dystrophic subtype of Epidermolysis Bullosa (RDEB), and wherein the gene editing ribonucleoprotein system is configured for collagen VII exon 80 excision.
- the gene editing ribonucleoprotein system is a CRISPR-Cas9 gene editing system.
- the invention provides a method of treating a genetic disease in a subject comprising a step of administering a composition according to the invention to the individual.
- the composition of the invention comprises a gene editing ribonucleoprotein system typically comprising a CRISPR nuclease complexed with a cationic polymer.
- the CRISPR nuclease protein is Cas9 or a Cas9 derivative.
- the invention provides a method of treating a skin genetic disease in a subject comprising a step of administering a composition according to the invention to the skin of the individual by topical administration or sub-dermal injection, in which the composition of the invention comprises a gene editing ribonucleoprotein system typically comprising a CRISPR nuclease complexed with a cationic polymer.
- the CRISPR nuclease protein is Cas9 or a Cas9 derivative.
- the invention provides a method of genetically modifying a cell ex-vivo or in-vitro comprising a step of incubating the cell with a composition according to the invention, whereby the nucleic acid forming part of the composition genetically modifies the cell.
- the method comprises a step of isolating the cell from a subject, and then implanting the genetically modified cell into the subject.
- the subject has a genetic disease characterised by a mutation in a gene in the cell, wherein the nucleic acid is configured to correct the mutation or delete the mutation or all or part of an exon containing the mutation.
- the invention provides a method of genetically modifying a sample of tissue ex-vivo or in-vitro comprising a step of incubating the tissue with a composition according to the invention, whereby the nucleic acid genetically modifies at least some of the cells of the tissue.
- the method comprises a step of isolating the tissue from a subject, and then implanting the genetically modified tissue into the subject.
- the subject has a genetic disease characterised by a mutation in a gene in a cell of the tissue, wherein the nucleic acid is configured to correct the mutation or delete the mutation or all or part of an exon containing the mutation.
- the invention provides a cell or tissue genetically modified in-vitro or ex- vivo according to a method of the invention.
- FIGURE 1 Scheme defining the concept of Stable-Particles, and examples of the example library of specific anti-cleavage units .
- R can be the same or different and selected from the above structures, while at least one of the R groups links to polymer moiety.that can be integrated into polymers.
- FIGURE 2 Route of action of the Stable-Particles.
- FIGURE 3 Formation of the Stable-Particles for transfection
- FIGURE 4 Figure 4.
- the mRNA entrapment of HPAE-stable was higher and more stable than HPAE-Control and Xfect, although all groups had a high mRNA entrapment close to 100% after a short time incubation ( ⁇ 10 min) (Fig. 4a).
- the nanoparticle sizes of Stable-Particles before and after lyophilization were similar and both slightly smaller than HPAE-Control- and Xfect- formed mRNA-complex (Fig. 4b).
- the sizes of Stable-Particles before and after lyophilization were stable in aqueous solutions after 4 h incubation (Fig. 4c).
- FIGURE 5 Cell viability of HEK293 after 48 hours transfection with Stable-Particles.
- Transfected Human Embryonic Kidney cells (HEK293) with Stable-Particles at different doses (0.1 to 0.5 ⁇ g mRNA, 20:1 w/w) for luciferase expression showed high viability, comparable to untreated ones (set as 100% viability) and superior to Lipo MessengerMAX.
- FIGURE 6 Luminescence 48-hour post-transfection of HEK293 with Stable-Particles. The luminescence intensities represent the expression of luciferase protein after 48-hour post- transfection of HEK293. As different doses with 0.1 to 0.5 ⁇ g mRNA, the Stable-Particles showed excellent efficacy similar to or higher than Lipo MessengerMAX and one to two orders of magnitude superior to Xfect (Fig. 7).
- FIGURE 7 Cellular uptake of HPAE-stable formulated Stable-Particle and Xfect formulated mRNA-complex. The uptake and intracellular transportation of the Stable-Particle was visualized using fluorescently labelled red mRNA, showing excellent uptake and intracellular allocation of Stable-Particle (Fig. 7). On the contrary, the commercial linear PAE transfection reagent Xfect also successfully delivered mRNA into cells but failed to produce high efficacy compared to the Stable-Particle, presenting the unique and robust mRNA protection from the Stable-Particle comparable with leading lipid-based vectors (Fig. 6 and Fig. 7).
- FIGURE 8 Efficacy of liquid and lyophilized Stable-Particle store at different temperatures.
- Stable-Particles in the solution can be stored at -20 and -80 degrees Celsius for long terms, but not at room temperature or 4 degrees Celsius due to the spontaneous hydrolysis/degradation of abundant ester bonds in HPAE-stable polymer material, besides mRNA hydrolysis. After lyophilization, Stable-Particles remained 100% efficacy for long terms when stored at 4, -20 and -80 degrees Celsius, and remained majority efficacy at room temperature even after 24 weeks, which is considered stable.
- FIGURE 9 (a) Luminescence 48-hour post-transfection of HEK293 with HPAEs with different amounts of anti-cleavage units, (b) mRNA entrapment of HPAEs with different amounts of anti-cleavage units.
- the term “comprise,” or variations thereof such as “comprises” or “comprising,” are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, element, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers.
- the term “comprising” is inclusive or open-ended and does not exclude additional, unrecited integers or method/process steps.
- the term “disease” is used to define any abnormal condition that impairs physiological function and is associated with specific symptoms.
- the term is used broadly to encompass any disorder, illness, abnormality, pathology, sickness, condition or syndrome in which physiological function is impaired irrespective of the nature of the aetiology (or indeed whether the aetiological basis for the disease is established). It therefore encompasses conditions arising from infection, trauma, injury, surgery, radiological ablation, age, poisoning or nutritional deficiencies.
- treatment refers to an intervention (e.g. the administration of an agent to a subject) which cures, ameliorates or lessens the symptoms of a disease or removes (or lessens the impact of) its cause(s) (for example, the reduction in accumulation of pathological levels of lysosomal enzymes).
- the term is used synonymously with the term “therapy”.
- treatment refers to an intervention (e.g. the administration of an agent to a subject) which prevents or delays the onset or progression of a disease or reduces (or eradicates) its incidence within a treated population.
- the term treatment is used synonymously with the term “prophylaxis”.
- an effective amount or a therapeutically effective amount of an agent defines an amount that can be administered to a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, but one that is sufficient to provide the desired effect, e.g. the treatment or prophylaxis manifested by a permanent or temporary improvement in the subject's condition.
- the amount will vary from subject to subject, depending on the age and general condition of the individual, mode of administration and other factors. Thus, while it is not possible to specify an exact effective amount, those skilled in the art will be able to determine an appropriate "effective" amount in any individual case using routine experimentation and background general knowledge.
- a therapeutic result in this context includes eradication or lessening of symptoms, reduced pain or discomfort, prolonged survival, improved mobility and other markers of clinical improvement.
- a therapeutic result need not be a complete cure. Improvement may be observed in biological I molecular markers, clinical or observational improvements.
- the methods of the invention are applicable to humans, large racing animals (horses, camels, dogs), and domestic companion animals (cats and dogs).
- the term subject defines any subject, particularly a mammalian subject, for whom treatment is indicated.
- Mammalian subjects include, but are not limited to, humans, domestic animals, farm animals, zoo animals, sport animals, pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, camels, bison, cattle, cows; primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; and rodents such as mice, rats, hamsters and guinea pigs.
- the subject is a human.
- the term “equine” refers to mammals of the family Equidae, which includes horses, donkeys, asses, kiang and zebra.
- “Nucleic acid” as used herein refers to ribonucleic acid molecules, deoxyribonucleic acid molecules, and artificial nucleic acid molecules, that comprise phosphodiester bonds and are generally susceptible to auto-hydrolysis, acid hydrolysis or enzyme hydrolysis.
- the nucleic acid molecule is typically single stranded.
- Exemplary nucleic acids for use in the present invention include long RNA molecules, including messenger RNA (mRNA) and long non-coding RNA (IncRNA).
- nucleic acid containing complexes such as nucleoproteins including gene editing ribonucleotproteins.
- the nucleic acid is capable of editing a gene in a cell (gene editing nucleic acid), for example by deletion, excision or replacement of a mutation, a section of a gene, or an exon, knock down or knock out a gene, and homology directed repair (HDR).
- Polymer The polymer for use in the present invention may be a linear, branched, hyperbranched, comb or brush polymer.
- the polymer may be selected from a poly-beta amino ester (PAE) polymer, polyethylene glycol (PEG) polymer, an oligomer combination of PAE and PEG, a poly(amidoamine) polymer, a polyacrylate polymer, a polyaspartamide polymer, a natural polymer such as chitosan, or any derivative thereof.
- PAE poly-beta amino ester
- PEG polyethylene glycol
- PEG poly(amidoamine) polymer
- polyacrylate polymer a polyaspartamide polymer
- a natural polymer such as chitosan, or any derivative thereof.
- Diamine moiety means a chemical group having two terminal amine groups linked by a hydrocarbon backbone that may be linear or branched and is optionally substituted.
- the amine groups may each, independently, be primary, secondary or tertiary amine groups.
- Examples of diamine moieties suitable for use in the present invention are provided in Figure 1.
- the diamine moieties can be incorporated into polymers as terminal groups (i.e. the backbone, endcaps or side chains) to acquire optimal interaction with nucleic acids, or other polymer parts.
- the anti-cleavage units are optimally based on specific diamines, ideally with three to five carbons between two amine groups, and optionally including substitutions that do not hinder intramolecular affinity between the anti-cleavage units and phosphodiester bonds.
- the optimal carbon numbers depend on the structures of amine groups. With the optimal amine distance and structure, the anti- cleavage units can bind to negatively charged phosphodiester bonds of nucleic acids, restrict strand movement, and explicitly block the attack from 2'OH to prevent hydrolysis, if any.
- the structures of the anti-cleavage units should fit into the space around phosphodiester bonds to present the most effective hydrolysis-blocking ability, which can be varied with different nucleic acids.
- the units shall not carry structure/groups or too many charged atoms/groups that hinder optimal interaction with secondary/condensed secondary structures of the nucleic acids (Fig. 1). Structures of polymers carrying anti-cleavage units should be flexible and can have enough units interacting with as much as possible phosphodiester bonds within the nucleic acids for optimal stabilisation.
- the anti-cleavage units can be the examples given in Fig. 1.
- Terminal group as applied to the position in which the diamine moieties are incorporated into the polymer refers to diamine moieties being incorporated as endcap and side-chain terminal groups.
- X diamine moieties per polymer backbone refers to the average amounts of diamine moieties conjugated on polymer backbones after the endcapping reaction. Due to the nature of the polymerisation reaction, the molecular weights and available terminals for endcap conjugation vary slightly between each polymer molecule. Therefore, the average amounts of diamine moieties are controlled by endcap monomer feeding ratios and backbone branching degrees and can be measured and calculated by the Nuclear Magnetic Resonance (NMR) interpretation of the final polymer solution.
- NMR Nuclear Magnetic Resonance
- Comb polymer refers to polymer molecules consists of a main chain with linear side chains, and where there are two or more three-way branch points 1 .
- “Brush polymer” refers to polymer molecules consists of a main chain with linear side chain and where one or more of the branch points are at least four-way 2 .
- “Cationic polymer” refers to polymers with positive charges.
- Poly-beta amino ester or “PAE” polymer refers to the polymer formed from the Michael- type addition of bifunctional amines to diacrylate esters 3 .
- Poly-beta amino ester hyperbranched polymer refers to a cationic polymer formed by random polymerisation between branched monomers (for example monomers having three, four or more reactive sites that can react with acrylate or amine groups), diacrylate groups and first and second amine components to provide a highly branched poly(B-amino ester) (HPAE) having a 3-D structure and multiple end groups.
- HPAE highly branched poly(B-amino ester)
- the term includes 3- branching hyperbranched polymers and 4-branching hyperbranched polymers.
- Polyethylene glycol or “PEG” polymer refers to polymers obtained by polymerizing with monomers containing PEG units (eg. polyethylene glycol) diacrylate), where PEG refer to a oligomer or polymer of ethylene oxide 4 .
- “3-branching hyperbranched polymer” refers to polymers formed by reacting a monomer with three reacting sites that can react with acrylate or amine groups (three-branching monomer) with a diacrylate and first and second amine components.
- the polymer is formed using a oligomer combination approach, in which the diacrylate and first amine components are reacted together to form a first oligomer, the first oligomer and second amine component are reacted together to form a second oligomer, and the second oligomer and four branching monomer are reacted together to form the hyperbranched polymer of the invention.
- This oligomer combination approach is described in detail in Zeng et al (Nano. Lett.
- the four-branching monomer, diacrylate component, and first amine are reacted together in a Michael Addition reaction to form a first polymer, and the first polymer and second amine component (endcapping amine) are reacted together in a Michael Addition reaction to form the hyperbranched polymer of the invention.
- Examples of 3-branching hyperbranched polymers are described in US2017216455 and Zeng et al.
- “4-branching hyperbranched polymer” refers to polymers formed by reacting a monomer with four reacting sites that can react with acrylate or amine groups (four-branching monomer) with a diacrylate and first and second amine components. They are described in
- Gene editing ribonucleoprotein system or “gene editing RNP system” refers to a complex formed by a ribosomal protein bound to one or more sequences of nucleic acid that is capable of editing a gene in a mammal, for example by deleting or replacing a mutation in a gene or a segment of a gene (such as an exon), inserting an oligonucleotide into a gene (insertional mutagenesis), or modulating the expression of a gene (knock-down or knock- out mutation).
- the nucleic acid may be RNA in a format consisting of but no limited to, crRNA, tracRNA, sgRNA.
- the nucleic acid is a sgRNA comprising crRNA and tracrRNA.
- the ribosomal protein may be a CRISPR nuclease protein, e.g. Cas9, Cas12a, Cas14 or a Cas variant, for example modified versions of nuclease dead (dCas9).
- the gene editing ribonucleoprotein system can be further complimented with the addition of nucleic acids in the form of DNA or RNA or a combination of both. Complimentary nucleic acids can be incorporated into the gene editing ribonucleoprotein system to induce gene augmentation, gene silencing, gene addition, gene knockdown, gene knockout, gene editing via homology directed repair.
- the nucleic acid may be employed in a format consisting of but not limited to RNA oligonucleotides, antisense oligonucleotides.
- DNA may be employed in a format consisting of but not limited to DNA oligonucleotides, antisense oligonucleotides, single-strand DNA donor oligo, plasmid DNA.
- the gene editing system may be a CRISPR-associated Cas system (Sander and Joung (2014) CRISPR-Cas systems for editing, regulating and targeting genomes Nature Biotechnology 32(4): 347-355)), a TALEN system (Boch J (February 2011); "TALES of genome targeting". Nature Biotechnology. 29 (2): 135-6.
- the gene editing system is configured to perform insertational mutagenesis on a cell, for example OBLIGARE systems, and CRISPR-Cpf1 systems (Maresca et al.
- Obligate Ligation-Gated Recombination (ObLiGaRe): Custom-designed nuclease- mediated targeted integration through nonhomologous end joining Genome Res. 23: 539- 546; see also WO2014/033644), Fagerlund et al. (2015) The Cpf1 CRISPR-Cas protein expands genome-editing tools Genome Biology 16: 251-253; Ledford (2015) Bacteria yield new gene cutter Smaller CRISPR enzyme should simplify genome editing Nature 526: 17).
- the gene editing ribonucleoprotein system of the invention may be employed in gene addition, gene replacement, gene knockdown and gene editing.
- Gene replacement is defined as the provision of a functional healthy copy of a gene to replace a dysfunctional mutant containing gene which has given rise to a disease.
- Gene addition is defined as the supplementation of therapeutic genes that target a specific aspect of a disease mechanism.
- Gene knockdown is defined as the process of inhibiting a target genes capability to synthesize a toxic/dysfunctional protein which gives rise to a disease.
- Gene editing is defined as the process whereby a target genes nucleotide sequence is altered resulting in either a loss of function/correction/manipulation of gene expression.
- Such gene editing systems consists of but are not limited to i) clustered, regularly interspaced, palindromic repeats (CRISPR)-associated (Cas) system; (ii) a transcription activator-like effector nuclease (TALEN) system; or (iii) a zinc finger nuclease (ZFN) system.
- CRISPR clustered, regularly interspaced, palindromic repeats
- TALEN transcription activator-like effector nuclease
- ZFN zinc finger nuclease
- Linker means any linker group, including an aryl or alkyl group. Preferred linkers include O, NH, CH 2 , alkyl, lower alkyl, alkoxy, lower alkoxy, O-alkyl, CH 2 O, CH 2 NH, and CH 2 NHCOCH 2 , CO, COO. “Nanoparticulate composition” refers to a composition is the nano-size range.
- the particulate composition has a particle size of less than 2 pm, 1 .5 pm, 1000 nm, for example 20-900 nm, 50-800 nm, 50-700 nm, 50-600 nm, 50-500 nm, 50-400 nm, 50-300 nm, 100-300 nm, 150-250 nm, or about 200 nm.
- Gene therapy/editing The present invention may be used to edit a portion of the genome of a cell or replace a portion of the genome of a cell with an exogenous DNA insert in an orientation-specific manner.
- the invention may be used to edit or replace a defective portion of a disease-causing gene (i.e. for gene repair), or to insertionally inactivate (i.e. silence) a gene the expression of which is associated with a disease, or to edit or modify a gene for example to delete disease causing mutations or modify or add in residues required for normal functioning of a gene.
- a disease-causing gene i.e. for gene repair
- insertionally inactivate i.e. silence
- the invention finds application in gene therapy, as herein defined.
- Gene therapy according to the invention may target all of the cells in an organism, or may be targeted to a subset of cells (e.g. to selected organs, tissues or cells).
- Gene therapy according to the invention may target somatic cells specifically.
- Gene therapy according to the invention may exclude the targeting of germ line cells. It may exclude the targeting of totipotent cells. It may exclude the targeting of human embryos.
- the method may be applied ex vivo to isolated organs, tissues or cells (e.g. to blood, blood cells, immune cells, bone marrow cells, skin cells, nervous tissue, muscle etc.).
- isolated organs, tissues or cells e.g. to blood, blood cells, immune cells, bone marrow cells, skin cells, nervous tissue, muscle etc.
- Gene therapy finds application in the treatment of any genetically inherited disorder, particularly those arising from single gene mutations.
- gene therapy finds particular application in the treatment of lysosomal storage diseases, muscular dystrophies, cystic fibrosis, Marfan syndrome, sickle cell anaemia, dwarfism, phenylketonuria, neurofibromatosis, Huntington’s disease, osteogenesis imperfecta, thalassemia and hemochromatosis.
- diseases which may be suitable for gene therapy according to the invention include diseases and disorders of: blood, coagulation, heterogenous skin disease, cell proliferation and dysregulation, neoplasia (including cancer), inflammatory processes, immune system (including autoimmune diseases), metabolism, liver, kidney, musculoskeletal, neurological, neuronal and ocular tissues.
- Exemplary skin diseases include recessive dystrophic epidermolysis bullosa (RDEB), a rare heterogenous skin disease caused by biallelic loss-of-function mutations in the COL7A1 gene.
- Additional skin diseases may include: alternative EB subtypes such as Simplex EB and Junctional EB, Epidermolytic Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease and Localized Autosomal Recessive Hypotrichosis.
- Exemplary blood and coagulation diseases and disorders include: anaemia, bare lymphocyte syndrome, bleeding disorders, deficiencies of factor H, factor H-like 1 , factor V, factor VIII, factor VII, factor X, factor XI, factor XII, factor XIIIA, factor XIIIB, Fanconi anaemia, haemophagocytic lymphohistiocytosis, haemophilia A, haemophilia B, haemorrhagic disorder, leukocyte deficiency, sickle cell anaemia and thalassemia.
- immune related diseases and disorders include: AIDS; autoimmune lymphoproliferative syndrome; combined immunodeficiency; HIV -1 ; HIV susceptibility or infection; immunodeficiency and severe combined immunodeficiency (SCIDs).
- Autoimmune diseases which can be treated according to the invention include Grave’s disease, rheumatoid arthritis, Hashimoto’s thyroiditis, vitiligo, type I (early onset) diabetes, pernicious anaemia, multiple sclerosis, glomerulonephritis, systemic lupus E (SLE, lupus) and Sjogren syndrome.
- autoimmune diseases include scleroderma, psoriasis, ankylosing spondilitis, myasthenia gravis, pemphigus, polymyositis, dermomyositis, uveitis, Guillain-Barre syndrome, Crohn's disease and ulcerative colitis (frequently referred to collectively as inflammatory bowel disease (IBD)).
- IBD inflammatory bowel disease
- exemplary diseases include: amyloid neuropathy; amyloidosis; cystic fibrosis; lysosomal storage diseases; hepatic adenoma; hepatic failure; neurologic disorders; hepatic lipase deficiency; hepatoblastoma, cancer or carcinoma; medullary cystic kidney disease; phenylketonuria; polycystic kidney; or hepatic disease.
- Exemplary musculoskeletal diseases and disorders include: muscular dystrophy (e.g.
- Exemplary neurological and neuronal diseases and disorders include: ALS, Alzheimer's disease; autism; fragile X syndrome, Huntington's disease, Parkinson's disease, Schizophrenia, secretase related disorders, trinucleotide repeat disorders, Kennedy's disease, Friedrich's ataxia, Machado-Joseph's disease, spinocerebellar ataxia, myotonic dystrophy and dentatorubral pallidoluysian atrophy (DRPLA).
- ALS Alzheimer's disease
- autism fragile X syndrome
- Huntington's disease Huntington's disease
- Parkinson's disease Parkinson's disease
- Schizophrenia secretase related disorders
- trinucleotide repeat disorders Kennedy's disease, Friedrich's ataxia, Machado-Joseph's disease, spinocerebellar ataxia, myotonic dystrophy and dentatorubral pallidoluysian atrophy (DRPLA).
- Exemplary ocular diseases include: age related macular degeneration, corneal clouding and dystrophy, cornea plana congenital, glaucoma, Leber’s congenital amaurosis and macular dystrophy.
- Gene therapy according to the invention finds particular application in the treatment of lysosomal storage disorders. Listed below are exemplary lysosomal storage disorders and the corresponding defective enzymes:
- Gaucher disease Acid beta-glucosidase or glucocerebrosidase
- GMI-gangliosidosis Acid beta-galactosidase
- Tay-Sachs disease beta-Hexosaminidase A
- Hurler-Scheie disease alpha-L-lduronidase
- Morquio disease A N-Acetylgalactosamine-6-sulfate sulfatase
- Gene therapy according to the invention also finds particular application in the treatment of proteostatic diseases including both aggregative and misfolding proteostatic diseases, for example prion diseases, various amyloidoses and neurodegenerative disorders (e.g. Parkinson’s disease, Alzheimer’s disease and Huntington’s disease), certain forms of diabetes, emphysema, cancer and cystic fibrosis.
- proteostatic diseases including both aggregative and misfolding proteostatic diseases, for example prion diseases, various amyloidoses and neurodegenerative disorders (e.g. Parkinson’s disease, Alzheimer’s disease and Huntington’s disease), certain forms of diabetes, emphysema, cancer and cystic fibrosis.
- Cystic fibrosis occurs when there is a mutation in the CFTR gene leading to reduced ion channel activity (via increased clearance of the misfolded CFTR proteins).
- Gene therapy according to the invention finds particular application in the treatment of expanded CAG repeat diseases. These diseases stem from the expansion of CAG repeats in particular genes with the encoded proteins having corresponding polyglutamine tracts which lead to aggregation and accumulation in the nuclei and cytoplasm of neurons.
- Aggregated amino-terminal fragments of mutant huntingtin are toxic to neuronal cells and are thought to mediate neurodegeneration. Examples include Huntington's disease (HD), which is characterized by selective neuronal cell death primarily in the cortex and striatum. CAG expansions have also been found in at least seven other inherited neurodegenerative disorders, including for example spinal and bulbar muscular atrophy (SBMA), Kennedy’s disease, some forms of amyotrophic lateral sclerosis (ALS), dentatorubral pallidoluysian atrophy (DRPLA) and spinocerebellar ataxia (SCA) types 1 , 2, 3, 6 and 7.
- SBMA spinal and bulbar muscular atrophy
- ALS amyotrophic lateral sclerosis
- DRPLA dentatorubral pallidoluysian atrophy
- SCA spinocerebellar ataxia
- Gene therapy according to the invention finds particular application in the treatment of any neoplasia, including proliferative disorders, benign, pre-cancerous and malignant neoplasia, hyperlasia, metaplasia and dysplasia.
- the invention therefore finds application in the treatment of proliferative disorders which include, but are not limited to cancer, cancer metastasis, smooth muscle cell proliferation, systemic sclerosis, cirrhosis of the liver, adult respiratory distress syndrome, idiopathic cardiomyopathy, lupus erythematosus, retinopathy (e.g.
- Neoplasia involving smooth muscle cell proliferation include hyperproliferation of cells in the vasculature (e.g. intimal smooth muscle cell hyperplasia, restenosis and vascular occlusion, including in particular stenosis following biologically- or mechanically-mediated vascular injury, such as angioplasty).
- intimal smooth muscle cell hyperplasia can include hyperplasia in smooth muscle other than the vasculature (e.g.
- Non-cancerous proliferative disorders also include hyperproliferation of cells in the skin such as psoriasis and its varied clinical forms, Reiter's syndrome, pityriasis rubra pilaris and hyperproliferative variants of disorders of keratinization (including actinic keratosis, senile keratosis and scleroderma). Particularly preferred is the treatment of malignant neoplasia (cancer).
- cancer malignant neoplasia
- composition of the invention may be adapted for topical, oral, rectal, parenteral, intramuscular, intraperitoneal, intra-arterial, intrabronchial, subcutaneous, subdermal, intradermal, intravenous, nasal, vaginal, buccal, ocular or sublingual routes of administration.
- parenteral intramuscular, intraperitoneal, intra-arterial, intrabronchial, subcutaneous, subdermal, intradermal, intravenous, nasal, vaginal, buccal, ocular or sublingual routes of administration.
- intramuscular intraperitoneal
- intra-arterial intrabronchial
- subcutaneous, subdermal, intradermal, intravenous, nasal, vaginal, buccal, ocular or sublingual routes of administration for oral administration, particular use is made of compressed tablets, pills, tablets, drops, and capsules.
- these compositions contain from 0.01 to 250 mg and more preferably from 0.1-10 mg, of active ingredient per dose.
- Other forms of administration comprise solutions or emulsions which
- compositions of the present invention may also be in form of suspensions, emulsions, lotions, ointments, creams, gels, sprays, nebulizers, solutions or dusting powders.
- the composition of the invention may be formulated for topical delivery.
- Topical delivery generally means delivery to the skin, but can also mean delivery to a body lumen lined with epithelial cells, for example the lungs or airways, the gastrointestinal tract, the buccal cavity.
- formulations for topical delivery are described in Topical drug delivery formulations edited by David Osborne and Antonio Aman, Taylor & Francis, the complete contents of which are incorporated herein by reference.
- compositions or formulations for delivery to the airways are described in O’Riordan et al (Respir Care, 2002, Nov. 47), EP2050437, W02005023290, US2010098660, and US20070053845.
- Composition and formulations for delivering active agents to the ileum, especially the proximal ileum include microparticles and microencapsulates where the active agent is encapsulated within a protecting matrix formed of polymer or dairy protein that is acid resistant but prone to dissolution in the more alkaline environment of the ileum. Examples of such delivery systems are described in EP1072600.2 and EP13171757.1 .
- An alternative means of transdermal administration is by use of a skin patch.
- the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin or into a hydrogel.
- the active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
- Injectable forms may contain between 10-1000 mg, preferably between 10-250 mg, of active ingredient per dose.
- compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
- the agent may be administered at a dose of from 0.01 to 50 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
- pharmaceutically acceptable excipient refers to a diluent, adjuvant, excipient, or vehicle with which the polyplex is administered.
- Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
- Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like.
- the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents, or skin penetration enhancers. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
- a base polymer (PGMA) can be obtained through controlled radical polymerisation.
- the base polymer is then further capped with diamine 103 to obtain PGMA-103.
- the base polymer HA-A is prepared by reacting hyaluronic acid and glycidyl acrylate. The base polymer is then further capped with diamine 103 to obtain HA-A-103.
- Example 3 Synthesis of poly(amidomine) polymer with terminal diamine stabilising group
- An embodiment of a PAMAM polymer with terminal diamine stabilising groups was prepared according to Scheme 3 below.
- the PAMAM is prepared by divergent synthesis, which refers to the sequential “growth” of a dendrimer layer by layer.
- the example PAMAM polymer uses diamine 103 as initiator core to react with methyl acrylate via Michael addition. More diamine 103 is then coupled with ester-termination to form generation 1 PAMAM.
- a hyperbranched-based polymer (HB-PEG) can be obtained through controlled radical polymerisation.
- the base polymer is then further capped with diamine 103 to obtain Hyperbranched-PEG-103.
- a brush-shaped based polymer (Brush-PEG) can be obtained through controlled radical polymerisation.
- the base polymer is then further capped with diamine 103 to obtain Brush- PEG-103.
- the arms are prepared as linear polymer or long monomers (eg. PEGDA). Then the arms are attached to the core (eg. 103) to obtain the base polymer. The base polymer is then further capped with diamine 103 to obtain Star-PEG-103.
- PEGDA long monomers
- the base polymer is then further capped with diamine 103 to obtain Star-PEG-103.
- PEG polymer with terminal diamine stabilising groups was prepared according to Scheme 7 below. Through Michael addition of diamine and diacrylate monomers, a linear based polymer can be obtained. The base polymer is then further capped with diamine 103 to obtain linear- PEG-103.
- Backbone monomers are mixed in DMSO for polymerization at 90 °C to form hyperbranched backbones via Michael addition, and then 103 diamines are added and serve as endcap monomers to coupling with acrylate terminations to obtain hyperbranched PAE polymer with terminal stabilising groups.
- HPAEs contain different amounts of anti-cleavage units.
- Example 9 (comparative) - Synthesis of a hyperbranched PAE polymer without terminal diamine stabilising group (HPAE control)
- a hyperbranched HPAE was synthesized with a non-stabilising terminal diamine endcap in which the linker connecting the two amines contains carbon atoms and three oxygen atoms, according to Scheme 9 below.
- Backbone monomers are mixed in DMSO for polymerization at 90 °C to form hyperbranched backbones via Michael addition. Then non-stabilising terminal 122 diamines are added and serve as endcap monomer to coupling with acrylate terminations to obtain the control hyperbranched PAE polymer.
- the luciferase mRNA (Luc mRNA) was formulated with the HPAE-stable and HPAE-control polymers in different mixture buffer options, then either lyophilized with presenting of sugar lyoprotectant or used directly (Fig. 3).
- the mixture buffer combinations for the Stable- Particles were compelling, and extended buffer choices can be suitable for different uses.
- the Stable-Particles formed by mixing polymer ethanol solution and mRNA sodium acetate solution was selected as the example in the following sections.
- RiboGreenTM Assay is used to quantify mRNA entrapment. Luc mRNA and polymers were formulated to form polyplex solutions. Then, the polyplex solution samples were incubated with RiboGreenTM working solution at room temperature for 5 min and measured at 485 nm for fluorescence. After the first measurement, the RiboGreenTM mixed samples are then incubated at 37 °C for 1 , 2, 3 and 4 hours before fluorescence reading at 485 nm. Negative controls containing the same amount of polymers and RiboGreenTM and the positive control containing the same amount of mRNA and RiboGreenTM were also measured simultaneously. The mRNA entrapment is then calculated by:
- the sizes of polyplexes formulated by the different polymers and Luc mRNA were measured using Zetasizer Pro (Malvern Panalytical).
- the Zetasizer Pro contains He-Ne (633 nm) laser with 4 mW max powder. The temperature of the samples was controlled at 25 °C. 100 pl of the polyplexes solution was prepared and then measured freshly or after lyophilisation, stored at room temperature for 24 hours and final reconstitution. The lyophilisation and reconstitution methods are described in sections below. After loading polyplex solution samples in the disposable polystyrene cuvettes (Malvern Panalytical, ZEN0040), the samples were measured immediately and then incubated at 37°C for 1 , 2, 3 and 4 hours. The dynamic light scattering (DLS) with back angle was used to determine the particle size at each time point.
- DLS dynamic light scattering
- alamarBlueTM assay which provided a quantitative measurement of cell proliferation and metabolic health.
- Cell viability has been assessed 48-72 hrs post transfection experiments in cells.
- Culture media is removed from cells in a well plate and cells are washed with (hanks balanced salt solution) HBSS per well.
- 100 ⁇ l of alamarBlueTM working solution (10% alamarBlueTM in HBSS) is added to each well and allowed to incubate under normal cell culture conditions for 2 hrs protected from light.
- the alamarBlueTM solution is transferred to a fresh flat bottomed 96 well plate and absorbance at 570 nm and 600 nm is recorded on a SpectraMax M3 multi-plate reader. Wells containing alamarBlueTM reagent only are subtracted from each sample as a background reading. Untreated cells are used to normalize fluorescence values and plotted as 100% viable.
- Cells are seeded 24hr-48hrs prior to transfections to allow attachment to well plates and flasks. Cells are seeded at optimized cell densities.
- polymer- DNA complexes are prepared and after complexation, are mixed with the appropriate cell media such that the final polyplex solution is no more than 20% of the overall media volume.
- Cell media containing polymer-DNA complexes are added to cells and after 4hrs is removed and replaced with fresh media to remove complexes.
- trehalose is added to polyplex solution samples to the final trehalose concentration of 10% (w/w). All samples are frozen at -80 °C overnight and then immediately subjected to freeze-dry with a Christ Alpha 1 -2 LDplus Freeze-Dryer at -55 °C for 24 h. Afterwards, the polyplexes are stored at room temperature, 4 °C, -20 °C or -80 °C for 24 hours, or 1 , 2, 4, 6, 12, 24 weeks, respectively. Meanwhile, trehalose is added to the other group of polyplex solution samples to 10% (w/w), and then directly stored at room temperature, 4 °C, -20 °C or -80 °C for indicated periods. The lyophilised samples are reconstituted with the original volume of pure water and used for transfection along with thawed frozen samples to identify the change of size or change of transfection efficacy.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Engineering & Computer Science (AREA)
- Life Sciences & Earth Sciences (AREA)
- Bioinformatics & Cheminformatics (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Public Health (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Nanotechnology (AREA)
- Genetics & Genomics (AREA)
- Immunology (AREA)
- Organic Chemistry (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- General Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biophysics (AREA)
- Physics & Mathematics (AREA)
- Molecular Biology (AREA)
- Plant Pathology (AREA)
- Microbiology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- General Chemical & Material Sciences (AREA)
- Biochemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Dermatology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
A nanoparticulate composition comprising a nucleic acid complexed within a polymer, wherein the polymer comprises a diamine moiety incorporated into the polymer as a terminal group, and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone having 3 to 5 carbon atoms with or without hydrocarbon branching from the hydrocarbon backbone. The polymer comprises the plurality of diamine moieties as terminal endcap groups or polymer side-chain terminal groups. The or each diamine terminal group each, independently, has a chemical formula R1-L1-R2 in which R1 and R2 are each independently selected from a primary, secondary or tertiary amine group and L1 is a straight or branched, optionally substituted, hydrocarbon chain having 3, 4 or 5 carbon atoms with or without hydrocarbon branching from the hydrocarbon backbone. The use of the nanoparticulate composition in gene therapy of a subject is also disclosed.
Description
TITLE
Nanoparticulate compositions for gene therapy
Field of the Invention
The present invention relates to a nanoparticulate composition for gene therapy. Also contemplated are methods of treating skin genetic disorders such as Recessive dystrophic epidermolysis bullosa (RDEB).
Background to the Invention
Among non-viral gene delivery vectors, polymer-based and lipid-based vectors are the two most common vectors, which carry distinct advantages, but both contribute to gene therapy development.
Although many polymeric gene delivery vectors usually carry much larger payloads but can lack protective efficacy for cargos, exceptionally long RNA, which is sensitive to hydrolysis or other damaging factors, compared to lipid-based vectors. Even the poly(β -amino ester)s (PAEs), one of the most promising candidates, were not ideal for delivering mRNA susceptible to auto-hydrolysis, unless the PAEs are designed to be lipid-like and co- formulated with lipid-based components to form a more hydrophobic structure to protect mRNA from hydrolysis.
Hydrolysis is a crucial process for the intracellular and extracellular manipulation of genetic materials, but also the main reason for the function loss of nucleic acids during transfection. Long RNA, also called large RNA, mainly include messenger RNA (mRNA) and long non-coding RNA (IncRNA), are the typical nucleic acids susceptible to auto- hydrolysis, even without the presence of enzymes or any specific condition. The auto- hydrolysis happens because the ribose in RNA has a hydroxyl group at the 2' position, which can be deprotonated in aqueous solutions and attack the adjacent phosphorus in the phosphodiester bond of the RNA backbone, leading to backbone cleavage and subsequently loss of functions. Compared to long RNA, short double strands RNA and double strands DNA are much more stable due to double strands structures or lack of 2' OH groups, but they still can be susceptible to hydrolysis under acidic conditions with the
presence of enzymes or other particular conditions. Polymers for delivery of DNA are described in WANG YAO ET AL ( “Effects of Branching Strategy on the Gene Transfection of Highly Branched Poly(-amino ester)s”, CHINESE JOURNAL OF POLYMER SCIENCE, CHINESE CHEMICAL SOCIETY AND INSTITUTE OF CHEMISTRY, CAS, BEIJING, vol. 38, no. 8, 17 April 2020 (2020-04-17), pages 830-839), DEZHONG ZHOU ET AL ( SCIENCE ADVANCES, vol. 2, no. 6, 1 June 2016 (2016-06-01) page e1600102), WO2021/058492A1 , WO2010/132879A2 and GREGORY T ZUGATES ET AL (“Rapid Optimization of Gene Delivery by Parallel End-modification of Poly(-amino ester)s”, MOLECULAR THERAPY, vol. 15, no. 7, 1 July 2007 (2007-07-01), pages 1306-1312).
It is an object of the invention to overcome at least one of the above-referenced problems.
Summary of the Invention
The Applicant has unexpectedly discovered that anti-cleavage units, which have not been described before, incorporated into polymers, can encapsulate nucleic acids and form a stable-particle that presents a high stability against acidic hydrolysis, enzyme hydrolysis, and especially, auto- hydrolysis, without the need for inclusion of lipids to form a hydrophobic environment. The stable-particle has been found to be capable of delivering long RNA and, similarly, stabilised delivery of other nucleic acids. The polymers specifically carry diamine moieties (anti-cleavage units) that can both condense the nucleic acids via electrostatic interaction and protect each phosphodiester bond from hydrolysis and loss of function. Example of suitable anti-cleavage units are provided in Figure 1. The Applicant has also discovered that polymers having about 10 to about 150 diamine moieties per polymer backbone provides for excellent mRNA protection and transfection efficacy (Figure 9).
In a first aspect, the invention provides a composition comprising a nucleic acid complexed within a polymer, wherein the polymer comprises a diamine moiety incorporated into the polymer as a terminal group, and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone.
In any embodiment, the hydrocarbon backbone is a saturated hydrocarbon backbone.
In any embodiment, the hydrocarbon backbone has 3, 4 or 5 carbon atoms, with or without hydrocarbon branching from the hydrocarbon backbone.
In any embodiment, the polymer comprises the plurality of diamine moieties as terminal endcap groups.
In any embodiment, the polymer comprises the plurality of diamine moieties as polymer side-chain terminal groups.
In any embodiment, the polymer comprises 5 to 200 diamine moieties per polymer backbone.
In any embodiment, the polymer comprises about 10 to about 150 diamine moieties per polymer backbone.
In any embodiment, the polymer is a cationic polymer.
In any embodiment, the polymer is a linear polymer.
In any embodiment, the polymer is a branched polymer.
In any embodiment, the polymer is a hyperbranched polymer.
In one embodiment, the hyperbranched polymer is a 3-branching or 4-branching hyperbranched polymer.
In any embodiment, the polymer is a comb polymer.
In any embodiment, the polymer is a brush polymer.
In any embodiment, the polymer is a poly(beta amino ester) polymer (hereafter “PAE polymer”).
In any embodiment, the polymer is a PEG polymer.
In any embodiment, the polymer is a poly(glycidyl methacrylate) polymer.
In any embodiment, the polymer is a hyaluronic acid polymer.
In any embodiment, the polymer is a poly(amidoamine) polymer (hereafter “PAMAM polymer”).
In any embodiment, the polymer is an oligomer combination of a PAE and PEG polymers.
In any embodiment, the or each diamine terminal group each, independently, has a chemical formula R1-L1-R2: in which:
R1 and R2 are each independently selected from a primary, secondary or tertiary amine group; and
L1 is a linker.
In any embodiment, L1 is a straight or branched, optionally substituted, hydrocarbon chain having 3, 4 or 5 carbon atoms.
In any embodiment, L1 is saturated.
In any embodiment, L1 is selected from the group consisting of: CH2-(CH2)y-CH(X1)-CH2; CH2-CH(X2)-(CH2)y-CH2; CH2-CH(X2)-CH(X1)-CH2; CH(X3)-CH(X2)-CH(X1)-CH2; and, CH(X3)-CH(X2)-CH(X1)-CH(X4); wherein, y is at each occurrence 0, 1 or 2;
X1 is independently selected from the group consisting of: hydrogen, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X2 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X3 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X4 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH.
In any embodiment, L1 is selected from the group consisting of: (CH2)3; (CH2)4, (CH2)5,CH2- CH2-CH(CH3)-CH2; CH2-CH(CH3)-CH(CH3)-CH2; CH(CH3)-CH(CH3)-CH(CH3)-CH2; CH(CH3)-CH(CH3)-CH(CH3)-CH(CH3); and, CH(CH3)-CH2-CH2-CH2.
In any embodiment, L1 is selected from the group consisting of: (CH2)3; CH2-CH2-CH(CH3)- CH2.
In any embodiment, R1 has the structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group consisting of: H and CH2Z1 ; wherein
Z1 is independent selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH2OH, -F, -Cl, -Br, -I, -SH, -N3, -NO2, -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R1 ’ and R1 ” are the same or different.
In any embodiment, R1 has a structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer, wherein R1 ’ and R1 ” are the same or different.
In any embodiment, R2 has the structure R2’-N-R2”, in which R2’ and R2” are each independently selected form the group consisting of: H and CH2Z2; wherein
Z2 is independently selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH2OH, -F, -Cl, -Br, -I, -SH, -N3, -NO2, -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R2’ and R2” are the same or different.
In any embodiment, R2 has a structure selected from:
R2’-N-R2”, in which R2’ and R2” are each independently selected from the group consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer; and a heterocyclic group containing two ring nitrogen atoms;
wherein R2’ and R2” are the same or different.
In any embodiment, R2, or R1 , or R2 and R1 , have a structure selected from: a substituted or unsubstituted piperazinyl group; a substituted or unsubstituted piperidinyl group; a substituted or unsubstituted pyrrolidinyl group; a substituted or unsubstituted pyrazolidinyl group; and a substituted or unsubstituted imidazolidinyl group.
In any embodiment, the or each diamine terminal group each, independently, has a chemical formula R1-L1-R2: in which:
R1 has a structure R1 ’-N-R1 ”, in which R1’ and R1 ” are each independently selected from the group consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer;
L1 is selected from the group consisting of: (CH2)3; and CH2-CH2-CH(CH3)CH2;
R2 has a structure selected from:
R2’-N-R2”, in which R2’ and R2” are each independently selected from the group consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer; and a heterocyclic group containing two ring nitrogen atoms.
In any embodiment, L1 does not contain an ether group.
In any embodiment, the diamine terminal group has a molecular weight of less than 2000 Da, 1500 Da, 1000 Da, 500 Da, 400 Da, 300 Da, 250 Da or 200 Da.
In any embodiment, the composition is a non-viral gene delivery vector.
In any embodiment, the composition is substantially free of lipid.
In any embodiment, the nucleic acid is a single stranded nucleic acid.
In any embodiment, the nucleic acid is an RNA molecule.
In any embodiment, the RNA molecule is a long RNA molecule.
In any embodiment, the long RNA molecule is selected from a messenger RNA molecule (mRNA) and a long non-coding RNA molecule (IncRNA).
In any embodiment, the composition is a pharmaceutical composition and comprises a a suitable pharmaceutical excipient.
In any embodiment, the composition is microparticulate.
In any embodiment, the composition is nanoparticulate.
In any embodiment, the nucleic acid is provided in the form of a gene editing ribonucleoprotein system.
In any embodiment, the gene editing ribonucleoprotein system is a Cas9-gRNA ribonucleoprotein system, such as a CRISPR-Cas9 gene editing system, which is typically configured to induce deletion of a targeted genomic sequence including excision of a mutation or exon in a gene, replace a mutation in a gene, or produce a knock-down or knock-out of a gene. Other gene editing ribonucleoprotein systems that may be employed with the present invention include for example alternative CRISPR-Cas derivatives such as Cas12a, Cas14, CRISPR Base editors, zinc finger nuclease systems and TALEN systems.
In any preferred embodiment, the gene editing ribonucleoprotein system is a CRISPR- Cas9 gene editing system.
In any embodiment, the gene editing ribonucleoprotein system is configured for exon- excision. In one embodiment, the gene editing ribonucleoprotein system is a CRISPR-Cas9
gene editing system. In one embodiment, the gene editing ribonucleoprotein system is configured to excise exon 80 of the COL7A1 gene encoding for Collagen VII protein.
In any embodiment, the nanoparticulate composition has an average dimension of 50-500, 50-400, 50-300, 100-400, 100-300, 150-250, and ideally about 200 nm. Methods of measuring the average size of the nanoparticulate compositions are for example a dynamic light scattering system or transmission electron microscopy. To measure nanoparticulate size, and polydispersity index (PDI), which provides a measurement of nanoparticle uniformity in solution, a Malvern Zetasizer Nano ZS (Malvern Instrument) equipped with a scattering angle of 173° can be used. Nanoparticulate size measurements are performed in a clear plastic disposable cuvette.
The invention also provides a method of making a composition of the invention comprising the steps of: providing a solution of nucleic acid in a buffer; providing a solution of polymer in a suitable non-aqueous solvent, in which the polymer comprises a diamine moiety incorporated into the polymer as a terminal group; and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone; and mixing the solutions such that there is optionally an excess of mass of the polymer over that of the nucleic acid in the mixture; and typically resting the mixture to allow the composition to form.
In one embodiment, the polymer solution is prepared by dissolving the polymer in an alcohol such as ethanol.
In one embodiment, the polymer is dissolved in the solvent at a concentration of 10-200 mg/ml, preferably 50-150 mg/ml, and ideally about 100 mg/ml.
In one embodiment, the polymer solution comprises 0.1 to 100 g of polymer.
system typically containing 0.1 to 100 μg of ribonucleoprotein complex.
In one embodiment, the nucleic acid buffer is sodium acetate.
In one embodiment, the first and second solutions are mixed at a volumetric ratio of about 1-100:1-100, such that there is an excess of mass of the polymer over that of the nucleic acid.
In one embodiment, the amount of polymer in the second solution is 1 to 100 times more than the nucleic acid in terms of mass, and in which the first and second solutions are mixed at a volumetric ratio of about 1-100:1-100.
In one embodiment, the polymer comprises 10 to 150 diamine moieties per polymer backbone.
The invention also provides a composition of the invention, for use in a method of treatment of a genetic disease in a subject, in which a therapeutically effective amount of the composition is administered to the subject.
In any embodiment, the genetic disease is characterised by a mutation in a gene of the individual.
In any embodiment, the nucleic acid is configured to edit a gene of the individual. Editing may comprise non-homologous end joining (NHEJ) (i.e. for large or small genomic deletions or exon excision), knock down or knock out a gene, and homology direct repair (HDR). In a preferred embodiment, the editing comprises deleting or replacing the mutation or a section of the gene including the mutation.
In one embodiment, the composition is administered topically or by sub-dermal injection.
In one embodiment, the genetic disease is selected from a skin genetic disorder. Examples include Epidermolysis Bullosa (EB), Recessive dystrophic epidermolysis bullosa (RDEB), Epidermolytic Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease, Localized Autosomal Recessive Hypotrichosis. Additional skin diseases may include: alternative EB subtypes such as Simplex EB and Junctional EB, Epidermolytic
Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease and, Localized Autosomal Recessive Hypotrichosis,
In one embodiment, the genetic disease is Recessive Dystrophic subtype of Epidermolysis Bullosa (RDEB), and wherein the gene editing ribonucleoprotein system is configured for collagen VII exon 80 excision. Preferably, the gene editing ribonucleoprotein system is a CRISPR-Cas9 gene editing system.
In another aspect, the invention provides a method of treating a genetic disease in a subject comprising a step of administering a composition according to the invention to the individual. In any embodiment, the composition of the invention comprises a gene editing ribonucleoprotein system typically comprising a CRISPR nuclease complexed with a cationic polymer. Typically, the CRISPR nuclease protein is Cas9 or a Cas9 derivative.
In another aspect, the invention provides a method of treating a skin genetic disease in a subject comprising a step of administering a composition according to the invention to the skin of the individual by topical administration or sub-dermal injection, in which the composition of the invention comprises a gene editing ribonucleoprotein system typically comprising a CRISPR nuclease complexed with a cationic polymer. Typically, the CRISPR nuclease protein is Cas9 or a Cas9 derivative.
In another aspect, the invention provides a method of genetically modifying a cell ex-vivo or in-vitro comprising a step of incubating the cell with a composition according to the invention, whereby the nucleic acid forming part of the composition genetically modifies the cell. In one embodiment, the method comprises a step of isolating the cell from a subject, and then implanting the genetically modified cell into the subject. In one embodiment, the subject has a genetic disease characterised by a mutation in a gene in the cell, wherein the nucleic acid is configured to correct the mutation or delete the mutation or all or part of an exon containing the mutation.
In another aspect, the invention provides a method of genetically modifying a sample of tissue ex-vivo or in-vitro comprising a step of incubating the tissue with a composition according to the invention, whereby the nucleic acid genetically modifies at least some of the cells of the tissue. In one embodiment, the method comprises a step of isolating the tissue from a subject, and then implanting the genetically modified tissue into the subject.
In one embodiment, the subject has a genetic disease characterised by a mutation in a gene in a cell of the tissue, wherein the nucleic acid is configured to correct the mutation or delete the mutation or all or part of an exon containing the mutation.
In another aspect, the invention provides a cell or tissue genetically modified in-vitro or ex- vivo according to a method of the invention.
Other aspects and preferred embodiments of the invention are defined and described in the other claims set out below.
Brief Description of the Figures
FIGURE 1 : Scheme defining the concept of Stable-Particles, and examples of the example library of specific anti-cleavage units . R can be the same or different and selected from the above structures, while at least one of the R groups links to polymer moiety.that can be integrated into polymers.
FIGURE 2: Route of action of the Stable-Particles.
FIGURE 3: Formation of the Stable-Particles for transfection
FIGURE 4: Figure 4. (a) mRNA entrapment of HPAE-stable and control groups; (b) Nanoparticle sizes of HPAE-stable and control groups; (c) Nanoparticle stability of HPAE- stable before and after lyophilization. The mRNA entrapment of HPAE-stable was higher and more stable than HPAE-Control and Xfect, although all groups had a high mRNA entrapment close to 100% after a short time incubation (<10 min) (Fig. 4a). The nanoparticle sizes of Stable-Particles before and after lyophilization were similar and both slightly smaller than HPAE-Control- and Xfect- formed mRNA-complex (Fig. 4b). The sizes of Stable-Particles before and after lyophilization were stable in aqueous solutions after 4 h incubation (Fig. 4c).
FIGURE 5: Cell viability of HEK293 after 48 hours transfection with Stable-Particles. Transfected Human Embryonic Kidney cells (HEK293) with Stable-Particles at different doses (0.1 to 0.5 μg mRNA, 20:1 w/w) for luciferase expression showed high viability, comparable to untreated ones (set as 100% viability) and superior to Lipo MessengerMAX.
FIGURE 6: Luminescence 48-hour post-transfection of HEK293 with Stable-Particles. The luminescence intensities represent the expression of luciferase protein after 48-hour post- transfection of HEK293. As different doses with 0.1 to 0.5 μg mRNA, the Stable-Particles showed excellent efficacy similar to or higher than Lipo MessengerMAX and one to two orders of magnitude superior to Xfect (Fig. 7).
FIGURE 7: Cellular uptake of HPAE-stable formulated Stable-Particle and Xfect formulated mRNA-complex. The uptake and intracellular transportation of the Stable-Particle was visualized using fluorescently labelled red mRNA, showing excellent uptake and intracellular allocation of Stable-Particle (Fig. 7). On the contrary, the commercial linear PAE transfection reagent Xfect also successfully delivered mRNA into cells but failed to produce high efficacy compared to the Stable-Particle, presenting the unique and robust mRNA protection from the Stable-Particle comparable with leading lipid-based vectors (Fig. 6 and Fig. 7).
FIGURE 8: Efficacy of liquid and lyophilized Stable-Particle store at different temperatures. Stable-Particles in the solution can be stored at -20 and -80 degrees Celsius for long terms, but not at room temperature or 4 degrees Celsius due to the spontaneous hydrolysis/degradation of abundant ester bonds in HPAE-stable polymer material, besides mRNA hydrolysis. After lyophilization, Stable-Particles remained 100% efficacy for long terms when stored at 4, -20 and -80 degrees Celsius, and remained majority efficacy at room temperature even after 24 weeks, which is considered stable.
FIGURE 9: (a) Luminescence 48-hour post-transfection of HEK293 with HPAEs with different amounts of anti-cleavage units, (b) mRNA entrapment of HPAEs with different amounts of anti-cleavage units.
Detailed Description of the Invention
All publications, patents, patent applications and other references mentioned herein are hereby incorporated by reference in their entireties for all purposes as if each individual publication, patent or patent application were specifically and individually indicated to be incorporated by reference and the content thereof recited in full.
Definitions and general preferences
Where used herein and unless specifically indicated otherwise, the following terms are intended to have the following meanings in addition to any broader (or narrower) meanings the terms might enjoy in the art:
Unless otherwise required by context, the use herein of the singular is to be read to include the plural and vice versa. The term "a" or "an" used in relation to an entity is to be read to refer to one or more of that entity. As such, the terms "a" (or "an"), "one or more," and "at least one" are used interchangeably herein.
As used herein, the term "comprise," or variations thereof such as "comprises" or "comprising," are to be read to indicate the inclusion of any recited integer (e.g. a feature, element, characteristic, property, method/process step or limitation) or group of integers (e.g. features, element, characteristics, properties, method/process steps or limitations) but not the exclusion of any other integer or group of integers. Thus, as used herein the term "comprising" is inclusive or open-ended and does not exclude additional, unrecited integers or method/process steps.
As used herein, the term “disease” is used to define any abnormal condition that impairs physiological function and is associated with specific symptoms. The term is used broadly to encompass any disorder, illness, abnormality, pathology, sickness, condition or syndrome in which physiological function is impaired irrespective of the nature of the aetiology (or indeed whether the aetiological basis for the disease is established). It therefore encompasses conditions arising from infection, trauma, injury, surgery, radiological ablation, age, poisoning or nutritional deficiencies.
As used herein, the term "treatment" or "treating" refers to an intervention (e.g. the administration of an agent to a subject) which cures, ameliorates or lessens the symptoms of a disease or removes (or lessens the impact of) its cause(s) (for example, the reduction in accumulation of pathological levels of lysosomal enzymes). In this case, the term is used synonymously with the term “therapy”.
Additionally, the terms "treatment" or "treating" refers to an intervention (e.g. the administration of an agent to a subject) which prevents or delays the onset or progression of a disease or reduces (or eradicates) its incidence within a treated population. In this case, the term treatment is used synonymously with the term “prophylaxis”.
As used herein, an effective amount or a therapeutically effective amount of an agent defines an amount that can be administered to a subject without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio, but one that is sufficient to provide the desired effect, e.g. the treatment or prophylaxis manifested by a permanent or temporary improvement in the subject's condition. The amount will vary from subject to subject, depending on the age and general condition of the individual, mode of administration and other factors. Thus, while it is not possible to specify an exact effective amount, those skilled in the art will be able to determine an appropriate "effective" amount in any individual case using routine experimentation and background general knowledge. A therapeutic result in this context includes eradication or lessening of symptoms, reduced pain or discomfort, prolonged survival, improved mobility and other markers of clinical improvement. A therapeutic result need not be a complete cure. Improvement may be observed in biological I molecular markers, clinical or observational improvements. In a preferred embodiment, the methods of the invention are applicable to humans, large racing animals (horses, camels, dogs), and domestic companion animals (cats and dogs).
In the context of treatment and effective amounts as defined above, the term subject (which is to be read to include "individual", "animal", "patient" or "mammal" where context permits) defines any subject, particularly a mammalian subject, for whom treatment is indicated. Mammalian subjects include, but are not limited to, humans, domestic animals, farm animals, zoo animals, sport animals, pet animals such as dogs, cats, guinea pigs, rabbits, rats, mice, horses, camels, bison, cattle, cows; primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; and rodents such as mice, rats, hamsters and guinea pigs. In preferred embodiments, the subject is a human. As used herein, the term “equine” refers to mammals of the family Equidae, which includes horses, donkeys, asses, kiang and zebra.
“Nucleic acid” as used herein refers to ribonucleic acid molecules, deoxyribonucleic acid molecules, and artificial nucleic acid molecules, that comprise phosphodiester bonds and are generally susceptible to auto-hydrolysis, acid hydrolysis or enzyme hydrolysis. The nucleic acid molecule is typically single stranded. Exemplary nucleic acids for use in the present invention include long RNA molecules, including messenger RNA (mRNA) and long non-coding RNA (IncRNA). The term also encompasses nucleic acid containing complexes such as nucleoproteins including gene editing ribonucleotproteins. Typically, the nucleic acid is capable of editing a gene in a cell (gene editing nucleic acid), for example by deletion, excision or replacement of a mutation, a section of a gene, or an exon, knock down or knock out a gene, and homology directed repair (HDR).
“Polymer”: The polymer for use in the present invention may be a linear, branched, hyperbranched, comb or brush polymer. The polymer may be selected from a poly-beta amino ester (PAE) polymer, polyethylene glycol (PEG) polymer, an oligomer combination of PAE and PEG, a poly(amidoamine) polymer, a polyacrylate polymer, a polyaspartamide polymer, a natural polymer such as chitosan, or any derivative thereof.
“Diamine moiety” means a chemical group having two terminal amine groups linked by a hydrocarbon backbone that may be linear or branched and is optionally substituted. The amine groups may each, independently, be primary, secondary or tertiary amine groups. Examples of diamine moieties suitable for use in the present invention are provided in Figure 1. The diamine moieties can be incorporated into polymers as terminal groups (i.e. the backbone, endcaps or side chains) to acquire optimal interaction with nucleic acids, or other polymer parts. The inventors discovered that the anti-cleavage units are optimally based on specific diamines, ideally with three to five carbons between two amine groups, and optionally including substitutions that do not hinder intramolecular affinity between the anti-cleavage units and phosphodiester bonds. The optimal carbon numbers depend on the structures of amine groups. With the optimal amine distance and structure, the anti- cleavage units can bind to negatively charged phosphodiester bonds of nucleic acids, restrict strand movement, and explicitly block the attack from 2'OH to prevent hydrolysis, if any. Furthermore, the structures of the anti-cleavage units should fit into the space around phosphodiester bonds to present the most effective hydrolysis-blocking ability, which can be varied with different nucleic acids. In addition, the units shall not carry structure/groups or too many charged atoms/groups that hinder optimal interaction with secondary/condensed secondary structures of the nucleic acids (Fig. 1). Structures of
polymers carrying anti-cleavage units should be flexible and can have enough units interacting with as much as possible phosphodiester bonds within the nucleic acids for optimal stabilisation. In some embodiments, the anti-cleavage units can be the examples given in Fig. 1.
“Terminal group” as applied to the position in which the diamine moieties are incorporated into the polymer refers to diamine moieties being incorporated as endcap and side-chain terminal groups.
“X diamine moieties per polymer backbone” refers to the average amounts of diamine moieties conjugated on polymer backbones after the endcapping reaction. Due to the nature of the polymerisation reaction, the molecular weights and available terminals for endcap conjugation vary slightly between each polymer molecule. Therefore, the average amounts of diamine moieties are controlled by endcap monomer feeding ratios and backbone branching degrees and can be measured and calculated by the Nuclear Magnetic Resonance (NMR) interpretation of the final polymer solution.
“Comb polymer” refers to polymer molecules consists of a main chain with linear side chains, and where there are two or more three-way branch points1.
“Brush polymer” refers to polymer molecules consists of a main chain with linear side chain and where one or more of the branch points are at least four-way2.
“Cationic polymer” refers to polymers with positive charges. E.g. LPAE, HPAE, LBPAE, poly-beta amino ester hyperbranched polymers, hyperbranched polymers, hyperbranched poly-beta amino ester polymers, and hyperbranched PEG polymers.
“Poly-beta amino ester” or “PAE” polymer refers to the polymer formed from the Michael- type addition of bifunctional amines to diacrylate esters3.
“Poly-beta amino ester hyperbranched polymer” refers to a cationic polymer formed by random polymerisation between branched monomers (for example monomers having three, four or more reactive sites that can react with acrylate or amine groups), diacrylate groups and first and second amine components to provide a highly branched poly(B-amino ester) (HPAE) having a 3-D structure and multiple end groups. The term includes 3- branching hyperbranched polymers and 4-branching hyperbranched polymers.
“Polyethylene glycol” or “PEG” polymer refers to polymers obtained by polymerizing with monomers containing PEG units (eg. polyethylene glycol) diacrylate), where PEG refer to a oligomer or polymer of ethylene oxide4.
“3-branching hyperbranched polymer” refers to polymers formed by reacting a monomer with three reacting sites that can react with acrylate or amine groups (three-branching monomer) with a diacrylate and first and second amine components. In one embodiment, the polymer is formed using a oligomer combination approach, in which the diacrylate and first amine components are reacted together to form a first oligomer, the first oligomer and second amine component are reacted together to form a second oligomer, and the second oligomer and four branching monomer are reacted together to form the hyperbranched polymer of the invention. This oligomer combination approach is described in detail in Zeng et al (Nano. Lett. 2019 19, 381-391). In another embodiment, the four-branching monomer, diacrylate component, and first amine are reacted together in a Michael Addition reaction to form a first polymer, and the first polymer and second amine component (endcapping amine) are reacted together in a Michael Addition reaction to form the hyperbranched polymer of the invention. Examples of 3-branching hyperbranched polymers are described in US2017216455 and Zeng et al.
“4-branching hyperbranched polymer” refers to polymers formed by reacting a monomer with four reacting sites that can react with acrylate or amine groups (four-branching monomer) with a diacrylate and first and second amine components. They are described in
“Gene editing ribonucleoprotein system” or “gene editing RNP system” refers to a complex formed by a ribosomal protein bound to one or more sequences of nucleic acid that is capable of editing a gene in a mammal, for example by deleting or replacing a mutation in a gene or a segment of a gene (such as an exon), inserting an oligonucleotide into a gene (insertional mutagenesis), or modulating the expression of a gene (knock-down or knock- out mutation). The nucleic acid may be RNA in a format consisting of but no limited to, crRNA, tracRNA, sgRNA. Generally the nucleic acid is a sgRNA comprising crRNA and tracrRNA. The ribosomal protein may be a CRISPR nuclease protein, e.g. Cas9, Cas12a, Cas14 or a Cas variant, for example modified versions of nuclease dead (dCas9). The gene editing ribonucleoprotein system can be further complimented with the addition of nucleic acids in the form of DNA or RNA or a combination of both. Complimentary nucleic acids can be incorporated into the gene editing ribonucleoprotein system to induce gene
augmentation, gene silencing, gene addition, gene knockdown, gene knockout, gene editing via homology directed repair. In some embodiments the nucleic acid may be employed in a format consisting of but not limited to RNA oligonucleotides, antisense oligonucleotides. DNA may be employed in a format consisting of but not limited to DNA oligonucleotides, antisense oligonucleotides, single-strand DNA donor oligo, plasmid DNA. The gene editing system may be a CRISPR-associated Cas system (Sander and Joung (2014) CRISPR-Cas systems for editing, regulating and targeting genomes Nature Biotechnology 32(4): 347-355)), a TALEN system (Boch J (February 2011); "TALES of genome targeting". Nature Biotechnology. 29 (2): 135-6. doi:10.1038/nbt.1767. PMID 21301438), a meganuclease system, or a zinc finger nuclease (ZFN) system (Carroll, D (2011) "Genome engineering with zinc-finger nucleases". Genetics Society of America. 188 (4): 773 78doi:10.1534/genetics.111.131433. PMC 3176093. PMID 21828278). In one embodiment, the gene editing system is configured to perform insertational mutagenesis on a cell, for example OBLIGARE systems, and CRISPR-Cpf1 systems (Maresca et al. (2013) Obligate Ligation-Gated Recombination (ObLiGaRe): Custom-designed nuclease- mediated targeted integration through nonhomologous end joining Genome Res. 23: 539- 546; see also WO2014/033644), Fagerlund et al. (2015) The Cpf1 CRISPR-Cas protein expands genome-editing tools Genome Biology 16: 251-253; Ledford (2015) Bacteria yield new gene cutter Smaller CRISPR enzyme should simplify genome editing Nature 526: 17). The gene editing ribonucleoprotein system of the invention may be employed in gene addition, gene replacement, gene knockdown and gene editing. Gene replacement is defined as the provision of a functional healthy copy of a gene to replace a dysfunctional mutant containing gene which has given rise to a disease. Gene addition is defined as the supplementation of therapeutic genes that target a specific aspect of a disease mechanism. Gene knockdown is defined as the process of inhibiting a target genes capability to synthesize a toxic/dysfunctional protein which gives rise to a disease. Gene editing is defined as the process whereby a target genes nucleotide sequence is altered resulting in either a loss of function/correction/manipulation of gene expression. Such gene editing systems consists of but are not limited to i) clustered, regularly interspaced, palindromic repeats (CRISPR)-associated (Cas) system; (ii) a transcription activator-like effector nuclease (TALEN) system; or (iii) a zinc finger nuclease (ZFN) system.
“Linker” means any linker group, including an aryl or alkyl group. Preferred linkers include O, NH, CH2, alkyl, lower alkyl, alkoxy, lower alkoxy, O-alkyl, CH2O, CH2NH, and CH2NHCOCH2, CO, COO.
“Nanoparticulate composition” refers to a composition is the nano-size range. In one embodiment, the particulate composition has a particle size of less than 2 pm, 1 .5 pm, 1000 nm, for example 20-900 nm, 50-800 nm, 50-700 nm, 50-600 nm, 50-500 nm, 50-400 nm, 50-300 nm, 100-300 nm, 150-250 nm, or about 200 nm.
“Gene therapy/editing”: The present invention may be used to edit a portion of the genome of a cell or replace a portion of the genome of a cell with an exogenous DNA insert in an orientation-specific manner.
Thus, the invention may be used to edit or replace a defective portion of a disease-causing gene (i.e. for gene repair), or to insertionally inactivate (i.e. silence) a gene the expression of which is associated with a disease, or to edit or modify a gene for example to delete disease causing mutations or modify or add in residues required for normal functioning of a gene.
Thus, the invention finds application in gene therapy, as herein defined.
Gene therapy according to the invention may target all of the cells in an organism, or may be targeted to a subset of cells (e.g. to selected organs, tissues or cells).
Gene therapy according to the invention may target somatic cells specifically.
Gene therapy according to the invention may exclude the targeting of germ line cells. It may exclude the targeting of totipotent cells. It may exclude the targeting of human embryos.
In cases where gene therapy according to the invention is applied to selected organs, tissues or cells, the method may be applied ex vivo to isolated organs, tissues or cells (e.g. to blood, blood cells, immune cells, bone marrow cells, skin cells, nervous tissue, muscle etc.).
Gene therapy finds application in the treatment of any genetically inherited disorder, particularly those arising from single gene mutations. Thus, gene therapy finds particular application in the treatment of lysosomal storage diseases, muscular dystrophies, cystic
fibrosis, Marfan syndrome, sickle cell anaemia, dwarfism, phenylketonuria, neurofibromatosis, Huntington’s disease, osteogenesis imperfecta, thalassemia and hemochromatosis.
Other diseases which may be suitable for gene therapy according to the invention include diseases and disorders of: blood, coagulation, heterogenous skin disease, cell proliferation and dysregulation, neoplasia (including cancer), inflammatory processes, immune system (including autoimmune diseases), metabolism, liver, kidney, musculoskeletal, neurological, neuronal and ocular tissues.
Exemplary skin diseases include recessive dystrophic epidermolysis bullosa (RDEB), a rare heterogenous skin disease caused by biallelic loss-of-function mutations in the COL7A1 gene. Additional skin diseases may include: alternative EB subtypes such as Simplex EB and Junctional EB, Epidermolytic Palmoplantar Keratoderma, Hailey-Hailey’s disease, Darier’s disease and Localized Autosomal Recessive Hypotrichosis.
Exemplary blood and coagulation diseases and disorders include: anaemia, bare lymphocyte syndrome, bleeding disorders, deficiencies of factor H, factor H-like 1 , factor V, factor VIII, factor VII, factor X, factor XI, factor XII, factor XIIIA, factor XIIIB, Fanconi anaemia, haemophagocytic lymphohistiocytosis, haemophilia A, haemophilia B, haemorrhagic disorder, leukocyte deficiency, sickle cell anaemia and thalassemia.
Examples of immune related diseases and disorders include: AIDS; autoimmune lymphoproliferative syndrome; combined immunodeficiency; HIV -1 ; HIV susceptibility or infection; immunodeficiency and severe combined immunodeficiency (SCIDs). Autoimmune diseases which can be treated according to the invention include Grave’s disease, rheumatoid arthritis, Hashimoto’s thyroiditis, vitiligo, type I (early onset) diabetes, pernicious anaemia, multiple sclerosis, glomerulonephritis, systemic lupus E (SLE, lupus) and Sjogren syndrome. Other autoimmune diseases include scleroderma, psoriasis, ankylosing spondilitis, myasthenia gravis, pemphigus, polymyositis, dermomyositis, uveitis, Guillain-Barre syndrome, Crohn's disease and ulcerative colitis (frequently referred to collectively as inflammatory bowel disease (IBD)).
Other exemplary diseases include: amyloid neuropathy; amyloidosis; cystic fibrosis; lysosomal storage diseases; hepatic adenoma; hepatic failure; neurologic disorders; hepatic lipase deficiency; hepatoblastoma, cancer or carcinoma; medullary cystic kidney disease; phenylketonuria; polycystic kidney; or hepatic disease.
Exemplary musculoskeletal diseases and disorders include: muscular dystrophy (e.g.
Duchenne and Becker muscular dystrophies), osteoporosis and muscular atrophy.
Exemplary neurological and neuronal diseases and disorders include: ALS, Alzheimer's disease; autism; fragile X syndrome, Huntington's disease, Parkinson's disease, Schizophrenia, secretase related disorders, trinucleotide repeat disorders, Kennedy's disease, Friedrich's ataxia, Machado-Joseph's disease, spinocerebellar ataxia, myotonic dystrophy and dentatorubral pallidoluysian atrophy (DRPLA).
Exemplary ocular diseases include: age related macular degeneration, corneal clouding and dystrophy, cornea plana congenital, glaucoma, Leber’s congenital amaurosis and macular dystrophy.
Gene therapy according to the invention finds particular application in the treatment of lysosomal storage disorders. Listed below are exemplary lysosomal storage disorders and the corresponding defective enzymes:
Pompe disease: Acid alpha-glucosidase
Gaucher disease: Acid beta-glucosidase or glucocerebrosidase
Fabry disease: alpha-Galactosidase A
GMI-gangliosidosis: Acid beta-galactosidase
Tay-Sachs disease: beta-Hexosaminidase A
Sandhoff disease: beta-Hexosaminidase B
Niemann-Pick disease: Acid sphingomyelinase
Krabbe disease: Galactocerebrosidase
Farber disease: Acid ceramidase
Metachromatic leukodystrophy: Arylsulfatase A
Hurler-Scheie disease: alpha-L-lduronidase
Hunter disease: lduronate-2-sulfatase
Sanfilippo disease A: Heparan N-sulfatase
Sanfilippo disease B: alpha-N-Acetylglucosaminidase
Sanfilippo disease C: Acetyl-CoA: alpha-glucosaminide N-acetyltransferase
Sanfilippo disease D: N-Acetylglucosamine-6-sulfate sulfatase
Morquio disease A: N-Acetylgalactosamine-6-sulfate sulfatase
Morquio disease B: Acid beta-galactosidase
Maroteaux-Lamy disease: Arylsulfatase B
Sly disease: beta-Glucuronidase alpha-Mannosidosis: Acid alpha-mannosidase beta-Mannosidosis: Acid beta-mannosidase
Fucosidosis: Acid alpha-L-fucosidase
Sialidosis: Sialidase
Schindler-Kanzaki disease: alpha-N-acetylgalactosaminidase
Gene therapy according to the invention also finds particular application in the treatment of proteostatic diseases including both aggregative and misfolding proteostatic diseases, for example prion diseases, various amyloidoses and neurodegenerative disorders (e.g. Parkinson’s disease, Alzheimer’s disease and Huntington’s disease), certain forms of diabetes, emphysema, cancer and cystic fibrosis.
Gene therapy according to the invention finds particular application in the treatment of cystic fibrosis. Cystic fibrosis occurs when there is a mutation in the CFTR gene leading to reduced ion channel activity (via increased clearance of the misfolded CFTR proteins).
Gene therapy according to the invention finds particular application in the treatment of expanded CAG repeat diseases. These diseases stem from the expansion of CAG repeats in particular genes with the encoded proteins having corresponding polyglutamine tracts which lead to aggregation and accumulation in the nuclei and cytoplasm of neurons.
Aggregated amino-terminal fragments of mutant huntingtin are toxic to neuronal cells and are thought to mediate neurodegeneration. Examples include Huntington's disease (HD), which is characterized by selective neuronal cell death primarily in the cortex and striatum. CAG expansions have also been found in at least seven other inherited neurodegenerative disorders, including for example spinal and bulbar muscular atrophy (SBMA), Kennedy’s disease, some forms of amyotrophic lateral sclerosis (ALS), dentatorubral pallidoluysian atrophy (DRPLA) and spinocerebellar ataxia (SCA) types 1 , 2, 3, 6 and 7.
Gene therapy according to the invention finds particular application in the treatment of any neoplasia, including proliferative disorders, benign, pre-cancerous and malignant neoplasia, hyperlasia, metaplasia and dysplasia. The invention therefore finds application in the treatment of proliferative disorders which include, but are not limited to cancer, cancer metastasis, smooth muscle cell proliferation, systemic sclerosis, cirrhosis of the liver, adult respiratory distress syndrome, idiopathic cardiomyopathy, lupus erythematosus, retinopathy (e.g. diabetic retinopathy), cardiac hyperplasia, benign prostatic hyperplasia, ovarian cysts, pulmonary fibrosis, endometriosis, fibromatosis, hematomas, lymphangiomatosis, sarcoidosis and desmoid tumours. Neoplasia involving smooth muscle cell proliferation include hyperproliferation of cells in the vasculature (e.g. intimal smooth muscle cell hyperplasia, restenosis and vascular occlusion, including in particular stenosis following biologically- or mechanically-mediated vascular injury, such as angioplasty). Moreover, intimal smooth muscle cell hyperplasia can include hyperplasia in smooth muscle other than the vasculature (e.g. blockage of the bile duct, bronchial airways and in the kidneys of patients with renal interstitial fibrosis). Non-cancerous proliferative disorders also include hyperproliferation of cells in the skin such as psoriasis and its varied clinical forms, Reiter's syndrome, pityriasis rubra pilaris and hyperproliferative variants of disorders of keratinization (including actinic keratosis, senile keratosis and scleroderma). Particularly preferred is the treatment of malignant neoplasia (cancer).
Administration
The composition of the invention may be adapted for topical, oral, rectal, parenteral, intramuscular, intraperitoneal, intra-arterial, intrabronchial, subcutaneous, subdermal, intradermal, intravenous, nasal, vaginal, buccal, ocular or sublingual routes of administration. For oral administration, particular use is made of compressed tablets, pills, tablets, drops, and capsules. Preferably, these compositions contain from 0.01 to 250 mg and more preferably from 0.1-10 mg, of active ingredient per dose. Other forms of administration comprise solutions or emulsions which may be injected intravenously, intra- arterial, subcutaneously, intradermally, intraperitoneally or intramuscularly, and which are prepared from sterile or sterilisable solutions. The pharmaceutical compositions of the present invention may also be in form of suspensions, emulsions, lotions, ointments, creams, gels, sprays, nebulizers, solutions or dusting powders. The composition of the invention may be formulated for topical delivery. Topical delivery generally means delivery to the skin, but can also mean delivery to a body lumen lined with epithelial cells, for example the lungs or airways, the gastrointestinal tract, the buccal cavity. In particular,
formulations for topical delivery are described in Topical drug delivery formulations edited by David Osborne and Antonio Aman, Taylor & Francis, the complete contents of which are incorporated herein by reference. Compositions or formulations for delivery to the airways are described in O’Riordan et al (Respir Care, 2002, Nov. 47), EP2050437, W02005023290, US2010098660, and US20070053845. Composition and formulations for delivering active agents to the ileum, especially the proximal ileum, include microparticles and microencapsulates where the active agent is encapsulated within a protecting matrix formed of polymer or dairy protein that is acid resistant but prone to dissolution in the more alkaline environment of the ileum. Examples of such delivery systems are described in EP1072600.2 and EP13171757.1 . An alternative means of transdermal administration is by use of a skin patch. For example, the active ingredient can be incorporated into a cream consisting of an aqueous emulsion of polyethylene glycols or liquid paraffin or into a hydrogel. The active ingredient can also be incorporated, at a concentration of between 1 and 10% by weight, into an ointment consisting of a white wax or white soft paraffin base together with such stabilisers and preservatives as may be required.
Injectable forms may contain between 10-1000 mg, preferably between 10-250 mg, of active ingredient per dose.
Compositions may be formulated in unit dosage form, i.e., in the form of discrete portions containing a unit dose, or a multiple or sub-unit of a unit dose.
A person of ordinary skill in the art can easily determine an appropriate dose of one of the instant compositions to administer to a subject without undue experimentation. Typically, a physician will determine the actual dosage which will be most suitable for an individual patient and it will depend on a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual undergoing therapy. The dosages disclosed herein are exemplary of the average case. There can of course be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention. Depending upon the need, the agent may be administered at a dose of from 0.01 to 50 mg/kg body weight, such as from 0.1 to 10 mg/kg, more preferably from 0.1 to 1 mg/kg body weight.
The term "pharmaceutically acceptable excipient" refers to a diluent, adjuvant, excipient, or vehicle with which the polyplex is administered. Such pharmaceutical carriers can be sterile
liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents, or skin penetration enhancers. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
Exemplification
The invention will now be described with reference to specific examples. These are merely exemplary and for illustrative purposes only: they are not intended to be limiting in any way to the scope of the monopoly claimed or to the invention described. These examples constitute the best mode currently contemplated for practicing the invention.
Example 1 - Synthesis of polvtalycidyl methacrylate) polymer with terminal diamine stabilising group
An embodiment of a poly(glycidyl methacrylate) polymer with terminal diamine stabilising groups was prepared according to Scheme 1 below.
A base polymer (PGMA) can be obtained through controlled radical polymerisation. The base polymer is then further capped with diamine 103 to obtain PGMA-103.
Example 2 - Synthesis of hyaluronic acid polymer with terminal diamine stabilising group
An embodiment of a hyaluronic acid polymer with terminal diamine stabilising groups was prepared according to Scheme 2 below.
The base polymer HA-A is prepared by reacting hyaluronic acid and glycidyl acrylate. The base polymer is then further capped with diamine 103 to obtain HA-A-103.
Example 3 - Synthesis of poly(amidomine) polymer with terminal diamine stabilising group
An embodiment of a PAMAM polymer with terminal diamine stabilising groups was prepared according to Scheme 3 below.
The PAMAM is prepared by divergent synthesis, which refers to the sequential “growth” of a dendrimer layer by layer. The example PAMAM polymer uses diamine 103 as initiator core to react with methyl acrylate via Michael addition. More diamine 103 is then coupled with ester-termination to form generation 1 PAMAM.
Example 4 - Synthesis of a hyperbranched PEG polymer with terminal diamine stabilising group
An embodiment of a PEG polymer with terminal diamine stabilising groups was prepared according to Scheme 4 below.
A hyperbranched-based polymer (HB-PEG) can be obtained through controlled radical polymerisation. The base polymer is then further capped with diamine 103 to obtain Hyperbranched-PEG-103.
Example 5 - Synthesis of a brush PEG polymer with terminal diamine stabilising group
An embodiment of a PEG polymer with terminal diamine stabilising groups was prepared according to Scheme 5 below.
A brush-shaped based polymer (Brush-PEG) can be obtained through controlled radical polymerisation. The base polymer is then further capped with diamine 103 to obtain Brush- PEG-103.
Example 6 - Synthesis of a star PEG polymer with terminal diamine stabilising group
An embodiment of a PEG polymer with terminal diamine stabilising groups was prepared according to Scheme 6 below.
The arms are prepared as linear polymer or long monomers (eg. PEGDA). Then the arms are attached to the core (eg. 103) to obtain the base polymer. The base polymer is then further capped with diamine 103 to obtain Star-PEG-103.
Example 7 - Synthesis of a linear PEG polymer with terminal diamine stabilising group
An embodiment of a PEG polymer with terminal diamine stabilising groups was prepared according to Scheme 7 below.
Through Michael addition of diamine and diacrylate monomers, a linear based polymer can be obtained. The base polymer is then further capped with diamine 103 to obtain linear- PEG-103.
Example 8 - Synthesis of a hyperbranched PAE polymer with terminal diamine stabilising group (HPAE stable)
An embodiment of a hyperbranched PAE polymer with terminal diamine stabilising groups was prepared according to Scheme 8 below.
Backbone monomers are mixed in DMSO for polymerization at 90 °C to form hyperbranched backbones via Michael addition, and then 103 diamines are added and serve as endcap monomers to coupling with acrylate terminations to obtain hyperbranched PAE polymer with terminal stabilising groups.
Using the example monomers, a series of HPAE polymers containing different amounts of 103 endcap moieties have been synthesised (as seen in the table below).
The polymers were tested in HEK293 cells for the transfection of luciferase mRNA and their mRNA entrapment efficacy. Figure 9 shows that a specific range, about 10 to about 150 moieties, of anti-cleavage unit amounts provides for optimal mRNA protection and transfection efficacy.
Example 9 (comparative) - Synthesis of a hyperbranched PAE polymer without terminal diamine stabilising group (HPAE control)
As a control, a hyperbranched HPAE was synthesized with a non-stabilising terminal diamine endcap in which the linker connecting the two amines contains carbon atoms and three oxygen atoms, according to Scheme 9 below.
Backbone monomers are mixed in DMSO for polymerization at 90 °C to form hyperbranched backbones via Michael addition. Then non-stabilising terminal 122
diamines are added and serve as endcap monomer to coupling with acrylate terminations to obtain the control hyperbranched PAE polymer.
The luciferase mRNA (Luc mRNA) was formulated with the HPAE-stable and HPAE-control polymers in different mixture buffer options, then either lyophilized with presenting of sugar lyoprotectant or used directly (Fig. 3). The mixture buffer combinations for the Stable- Particles were compelling, and extended buffer choices can be suitable for different uses. The Stable-Particles formed by mixing polymer ethanol solution and mRNA sodium acetate solution was selected as the example in the following sections.
As an example:
1. Dilute mRNA in sodium acetate solution to desired concentrations;
2. Dilute polymers in ethanol in a separate tube to desired concentrations;
3. Add diluted polymer solution into diluted mRNA solution with a volume ratio of 1 :1 ;
4. Incubate for the required time (i.e. 1-10 min) to form Stable-Particles;
5. Use for transfection freshly or lyophilization for storage as drug products. mRNA entrapment, size and stability
RiboGreen™ Assay is used to quantify mRNA entrapment. Luc mRNA and polymers were formulated to form polyplex solutions. Then, the polyplex solution samples were incubated with RiboGreen™ working solution at room temperature for 5 min and measured at 485 nm for fluorescence. After the first measurement, the RiboGreen™ mixed samples are then incubated at 37 °C for 1 , 2, 3 and 4 hours before fluorescence reading at 485 nm. Negative controls containing the same amount of polymers and RiboGreen™ and the positive control containing the same amount of mRNA and RiboGreen™ were also measured simultaneously. The mRNA entrapment is then calculated by:
The sizes of polyplexes formulated by the different polymers and Luc mRNA were measured using Zetasizer Pro (Malvern Panalytical). The Zetasizer Pro contains He-Ne (633 nm) laser with 4 mW max powder. The temperature of the samples was controlled at 25 °C. 100 pl of the polyplexes solution was prepared and then measured freshly or after lyophilisation, stored at room temperature for 24 hours and final reconstitution. The lyophilisation and reconstitution methods are described in sections below. After loading
polyplex solution samples in the disposable polystyrene cuvettes (Malvern Panalytical, ZEN0040), the samples were measured immediately and then incubated at 37°C for 1 , 2, 3 and 4 hours. The dynamic light scattering (DLS) with back angle was used to determine the particle size at each time point.
Transfected Cell Viability
Evaluation of cell cytotoxicity induced by different polyplex conditions has been assessed using the alamarBlue™ assay, which provided a quantitative measurement of cell proliferation and metabolic health. Cell viability has been assessed 48-72 hrs post transfection experiments in cells. Culture media is removed from cells in a well plate and cells are washed with (hanks balanced salt solution) HBSS per well. Following this, 100 μl of alamarBlue™ working solution (10% alamarBlue™ in HBSS) is added to each well and allowed to incubate under normal cell culture conditions for 2 hrs protected from light. After incubation, the alamarBlue™ solution is transferred to a fresh flat bottomed 96 well plate and absorbance at 570 nm and 600 nm is recorded on a SpectraMax M3 multi-plate reader. Wells containing alamarBlue™ reagent only are subtracted from each sample as a background reading. Untreated cells are used to normalize fluorescence values and plotted as 100% viable.
Cell Transfection Efficiency
Cells are seeded 24hr-48hrs prior to transfections to allow attachment to well plates and flasks. Cells are seeded at optimized cell densities. On the day of transfection, polymer- DNA complexes are prepared and after complexation, are mixed with the appropriate cell media such that the final polyplex solution is no more than 20% of the overall media volume. Cell media containing polymer-DNA complexes are added to cells and after 4hrs is removed and replaced with fresh media to remove complexes.
Thermal stability of fresh and lyophilized Stable-Particles
For lyophilisation, trehalose is added to polyplex solution samples to the final trehalose concentration of 10% (w/w). All samples are frozen at -80 °C overnight and then immediately subjected to freeze-dry with a Christ Alpha 1 -2 LDplus Freeze-Dryer at -55 °C for 24 h. Afterwards, the polyplexes are stored at room temperature, 4 °C, -20 °C or -80 °C for 24 hours, or 1 , 2, 4, 6, 12, 24 weeks, respectively. Meanwhile, trehalose is added to the other group of polyplex solution samples to 10% (w/w), and then directly stored at room temperature, 4 °C, -20 °C or -80 °C for indicated periods.
The lyophilised samples are reconstituted with the original volume of pure water and used for transfection along with thawed frozen samples to identify the change of size or change of transfection efficacy.
Equivalents
The foregoing description details presently preferred embodiments of the present invention. Numerous modifications and variations in practice thereof are expected to occur to those skilled in the art upon consideration of these descriptions. Those modifications and variations are intended to be encompassed within the claims appended hereto.
References
1 . Abbasi, M., Faust, L. & Wilhelm, M. Comb and Bottlebrush Polymers with Superior Rheological and Mechanical Properties. Adv. Mater. 31 , 1806484 (2019).
2. Milner, S. T. Polymer brushes. Science (80-. ). 251 , 905-914 (1991).
3. Cordeiro, R. A., Serra, A., Coelho, J. F. J. & Faneca, H. Poly(β -amino ester)-based gene delivery systems: From discovery to therapeutic applications. Journal of Controlled Release vol. 310 155-187 (2019).
4. D’souza, A., delivery, R. S.-E. opinion on drug & 2016, undefined. Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications. Taylor Fr. 13, 1257- 1275 (2016).
Claims
1. A nanoparticulate composition comprising a nucleic acid complexed within a polymer, wherein the polymer comprises a diamine moiety incorporated into the polymer as a terminal group, and in which the or each diamine terminal group each, independently, has a chemical formula R1-L1-R2: in which:
R1 and R2 are each independently selected from a primary, secondary or tertiary amine group; and
L1 is a straight or branched, optionally substituted, hydrocarbon chain having 3, 4 or 5 carbon atoms with or without hydrocarbon branching from the hydrocarbon chain.
2. A nanoparticulate composition according to Claim 1 , in which the nucleic acid is a long RNA molecule.
3. A nanoparticulate composition according to Claim 1 , in which the nucleic acid is a long RNA molecule selected from a messenger RNA molecule (mRNA) and a long non-coding RNA molecule (IncRNA).
4. A nanoparticulate composition according to any preceding Claim, in which the hydrocarbon chain is a saturated hydrocarbon chain.
5. A nanoparticulate composition according to any preceding Claim, in which the polymer comprises a plurality of diamine moieties as terminal endcap groups or polymer side-chain terminal groups.
6. A nanoparticulate composition according to any preceding Claim, in which the polymer is Selected from a hyperbranched polymer, a comb polymer and a brush polymer.
7. A nanoparticulate composition according to any preceding Claim, in which the polymer is selected from the group consisting of: a poly(beta amino ester) polymer; a PEG polymer; a poly(glycidyl methacrylate) polymer; a hyaluronic acid polymer; a poly(amidoamine) polymer; and an oligomer combination of a PAE and PEG polymers.
8. A nanoparticulate composition according to any preceding Claim, in which L1 is selected from the group consisting of: CH2-(CH2)y-CH(X1)-CH2; CH2-CH(X2)-(CH2)y-CH2; CH2- CH(X2)-CH(X1)-CH2; CH(X3)-CH(X2)-CH(X1)-CH2; and, CH(X3)-CH(X2)-CH(X1)-CH(X4); wherein, y is at each occurrence 0, 1 or 2;
X1 is independently selected from the group consisting of: hydrogen, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X2 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X3 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH;
X4 is independently selected from the group consisting of: methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH -F, -Cl, -Br, -I, -SH, -N3, -NO2 and -COOH.
9. A nanoparticulate composition according to any preceding Claim, in which L1 is selected from the group consisting of: (CH2)3; (CH2)4, (CH2)5,CH2-CH2-CH(CH3)-CH2; CH2-CH(CH3)- CH(CH3)-CH2; CH(CH3)-CH(CH3)-CH(CH3)-CH2; CH(CH3)-CH(CH3)-CH(CH3)-CH(CH3); and, CH(CH3)-CH2-CH2-CH2.
10. A nanoparticulate composition according to any preceding Claims, in which R1 has the structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group consisting of: H and CH2Z1 ; wherein
Z1 is independent selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH2OH, -F, -Cl, -Br, -I, -SH, -N3, -NO2, -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R1 ’ and R1 ” are the same or different.
1 1 . A nanoparticulate composition according to any preceding Claim, in which R1 has a structure R1 ’-N-R1 ”, in which R1 ’ and R1 ” are each independently selected from the group
consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer, wherein R1’ and R1 ” are the same or different.
12. A nanoparticulate composition according to any preceding Claim, in which R2 has the structure R2’-N-R2”, in which R2’ and R2” are each independently selected form the group consisting of: H and CH2Z2; wherein
Z2 is independently selected from the group consisting of: H, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl (straight and branched), hexyl (straight and branched), -OH, -CH2OH, -F, -Cl, -Br, -I, -SH, -N3, -NO2, -COOH and a polymer, wherein at least one of R1 and R2 is linked to the polymer, wherein R2’ and R2” are the same or different.
13. A nanoparticulate composition according to any preceding Claim, in which R2 has a structure selected from:
R2’-N-R2”, in which R2’ and R2” are each independently selected from the group consisting of: H; CH3; CH2OH; CH2CH2OH; and a polymer; and a heterocyclic group containing two ring nitrogen atoms; wherein R2’ and R2” are the same or different.
14. A nanoparticulate composition according to any preceding Claim, in which R2, or R1 , or R2 and R1 , have a structure selected from: a substituted or unsubstituted piperazinyl group; a substituted or unsubstituted piperidinyl group; a substituted or unsubstituted pyrrolidinyl group; a substituted or unsubstituted pyrazolidinyl group; and a substituted or unsubstituted imidazolidinyl group.
15. A nanoparticulate composition according to Claim 1 , in which: the nucleic acid is a long RNA molecule; and the polymer is selected from the group consisting of: a poly(beta amino ester) polymer; a PEG polymer; a poly(glycidyl methacrylate) polymer; a hyaluronic acid polymer; a poly(amidoamine) polymer; and an oligomer combination of a PAE and PEG polymers.
16. A nanoparticulate composition according to any preceding Claim, in which: the nucleic acid is a mRNA molecule; and the polymer is a poly(beta amino ester) polymer.
17. A nanoparticulate composition comprising a nucleic acid complexed within a polymer, wherein the polymer comprises a diamine moiety incorporated into the polymer as a terminal group, and in which the diamine moiety comprises two terminal amine groups separated by a linear or branched, optionally substituted, hydrocarbon backbone having 3 to 5 carbon atoms with or without hydrocarbon branching from the hydrocarbon backbone, wherein the nucleic acid is a long RNA molecule.
18. A nanoparticulate composition according to Claim 17, in which the polymer comprises about 10 to about 150 diamine moieties per polymer backbone.
19. A nanoparticulate composition according to any preceding Claim, in which the long RNA molecule is a mRNA molecule.
20. A pharmaceutical composition comprising a nanoparticulate composition according to any of Claims 1 to 19 in combination with a suitable pharmaceutical excipient.
21. A pharmaceutical composition according to Claim 20, for use in a method of treating a genetic disorder in a subject by gene therapy.
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP22150378 | 2022-01-05 | ||
EP22150378.2 | 2022-01-05 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2023131648A1 true WO2023131648A1 (en) | 2023-07-13 |
Family
ID=80218443
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/EP2023/050181 WO2023131648A1 (en) | 2022-01-05 | 2023-01-05 | Nanoparticulate compositions for gene therapy |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2023131648A1 (en) |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2005023290A2 (en) | 2003-05-23 | 2005-03-17 | Pestka Biomedical Laboratories, Inc. | Uses of interferons for the treatment of severe acute respiratory syndrome and other viral infections |
US20070053845A1 (en) | 2004-03-02 | 2007-03-08 | Shiladitya Sengupta | Nanocell drug delivery system |
EP2050437A1 (en) | 2007-10-15 | 2009-04-22 | Laboratoires SMB | Improved pharmaceutical dry powder compositions for inhalation. |
US20100098660A1 (en) | 2007-05-18 | 2010-04-22 | Research Foundation Of State University Of New York And New York University | Method of treating tuberculosis with interferons |
WO2010132879A2 (en) | 2009-05-15 | 2010-11-18 | The Johns Hopkins University | Multicomponent degradable cationic polymers |
WO2014033644A2 (en) | 2012-08-28 | 2014-03-06 | Novartis Ag | Methods of nuclease-based genetic engineering |
US20170216455A1 (en) | 2014-08-06 | 2017-08-03 | National University Of Ireland, Galway | HYPERBRANCHED POLY (ß-AMINO ESTER) FOR GENE THERAPY |
WO2021058492A1 (en) | 2019-09-25 | 2021-04-01 | University College Dublin | Nanoparticle compositions for gene therapy |
-
2023
- 2023-01-05 WO PCT/EP2023/050181 patent/WO2023131648A1/en active Application Filing
Patent Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2005023290A2 (en) | 2003-05-23 | 2005-03-17 | Pestka Biomedical Laboratories, Inc. | Uses of interferons for the treatment of severe acute respiratory syndrome and other viral infections |
US20070053845A1 (en) | 2004-03-02 | 2007-03-08 | Shiladitya Sengupta | Nanocell drug delivery system |
US20100098660A1 (en) | 2007-05-18 | 2010-04-22 | Research Foundation Of State University Of New York And New York University | Method of treating tuberculosis with interferons |
EP2050437A1 (en) | 2007-10-15 | 2009-04-22 | Laboratoires SMB | Improved pharmaceutical dry powder compositions for inhalation. |
WO2010132879A2 (en) | 2009-05-15 | 2010-11-18 | The Johns Hopkins University | Multicomponent degradable cationic polymers |
WO2014033644A2 (en) | 2012-08-28 | 2014-03-06 | Novartis Ag | Methods of nuclease-based genetic engineering |
US20170216455A1 (en) | 2014-08-06 | 2017-08-03 | National University Of Ireland, Galway | HYPERBRANCHED POLY (ß-AMINO ESTER) FOR GENE THERAPY |
WO2021058492A1 (en) | 2019-09-25 | 2021-04-01 | University College Dublin | Nanoparticle compositions for gene therapy |
Non-Patent Citations (20)
Title |
---|
ABBASI, M.FAUST, L.WILHELM, M.: "Comb and Bottlebrush Polymers with Superior Rheological and Mechanical Properties", ADV. MATER., vol. 31, 2019, pages 1806484 |
BOCH J: "TALEs of genome targeting", NATURE BIOTECHNOLOGY, vol. 29, no. 2, February 2011 (2011-02-01), pages 135 - 6, XP055077654, DOI: 10.1038/nbt.1767 |
CARROLL, D: "Genome engineering with zinc-finger nucleases", GENETICS SOCIETY OF AMERICA, vol. 188, no. 4, 2011, pages 773 - 78, XP055171682, DOI: 10.1534/genetics.111.131433 |
CORDEIRO, R. A.SERRA, A.COELHO, J. F. J.FANECA, H.: "Poly([3-amino ester)-based gene delivery systems: From discovery to therapeutic applications", JOURNAL OF CONTROLLED RELEASE, vol. 310, 2019, pages 155 - 187 |
DEZHONG ZHOU ET AL, SCIENCE ADVANCES, vol. 2, no. 6, 1 June 2016 (2016-06-01), pages e1600102, XP055539263, DOI: 10.1126/sciadv.1600102 * |
DEZHONG ZHOU ET AL., SCIENCE ADVANCES, vol. 2, no. 6, 1 June 2016 (2016-06-01), pages e1600102 |
D'SOUZA, A.: "Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications", vol. 13, 2016, TAYLOR FR, article "delivery, R. S.-E. opinion on drug & 2016, undefined", pages: 1257 - 1275 |
FAGERLUND ET AL.: "The Cpf1 CRISPR-Cas protein expands genome-editing tools", GENOME BIOLOGY, vol. 16, 2015, pages 251 - 253 |
GREGORY T ZUGATES ET AL.: "Rapid Optimization of Gene Delivery by Parallel End-modification of Poly(-amino ester)s", MOLECULAR THERAPY, vol. 15, no. 7, 1 July 2007 (2007-07-01), pages 1306 - 1312, XP055261837, DOI: 10.1038/sj.mt.6300132 |
GREGORY T ZUGATES ET AL: "Rapid Optimization of Gene Delivery by Parallel End-modification of Poly(β-amino ester)s", MOLECULAR THERAPY, vol. 15, no. 7, 1 July 2007 (2007-07-01), US, pages 1306 - 1312, XP055261837, ISSN: 1525-0016, DOI: 10.1038/sj.mt.6300132 * |
KACZMAREK JAMES C ET AL: "Systemic delivery of mRNA and DNA to the lung using polymer-lipid nanoparticles", BIOMATERIALS, ELSEVIER, AMSTERDAM, NL, vol. 275, 10 June 2021 (2021-06-10), XP086706259, ISSN: 0142-9612, [retrieved on 20210610], DOI: 10.1016/J.BIOMATERIALS.2021.120966 * |
LEDFORD: "Bacteria yield new gene cutter Smaller CRISPR enzyme should simplify genome editing", NATURE, vol. 526, 2015, pages 17 |
MARESCA: "Obligate Ligation-Gated Recombination (ObLiGaRe): Custom-designed nuclease-mediated targeted integration through nonhomologous end joining", GENOME RES, vol. 23, 2013, pages 539 - 546, XP055077484, DOI: 10.1101/gr.145441.112 |
MILNER, S. T.: "Polymer brushes", SCIENCE, vol. 251, no. 80, 1991, pages 905 - 914, XP000941345, DOI: 10.1126/science.251.4996.905 |
O'RIORDAN ET AL., RESPIR CARE, November 2002 (2002-11-01) |
SANDER, JOUNG: "CRISPR-Cas systems for editing, regulating and targeting genomes", NATURE BIOTECHNOLOGY, vol. 32, no. 4, 2014, pages 347 - 355, XP055481941, DOI: 10.1038/nbt.2842 |
WANG YAO ET AL.: "CHINESE JOURNAL OF POLYMER SCIENCE", vol. 38, 17 April 2020, CHINESE CHEMICAL SOCIETY AND INSTITUTE OF CHEMISTRY, article "Effects of Branching Strategy on the Gene Transfection of Highly Branched Poly(-amino ester)s", pages: 830 - 839 |
WANG YAO ET AL: "Effects of Branching Strategy on the Gene Transfection of Highly Branched Poly(-amino ester)s", CHINESE JOURNAL OF POLYMER SCIENCE, CHINESE CHEMICAL SOCIETY AND INSTITUTE OF CHEMISTRY, CAS, BEIJING, vol. 38, no. 8, 17 April 2020 (2020-04-17), pages 830 - 839, XP037165668, ISSN: 0256-7679, [retrieved on 20200417], DOI: 10.1007/S10118-020-2393-Y * |
ZENG ET AL., NANO. LETT., vol. 19, 2019, pages 381 - 391 |
ZENG, 4-BRANCHING HYPERBRANCHED POLYMER |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
Luo et al. | Arginine functionalized peptide dendrimers as potential gene delivery vehicles | |
Qiu et al. | Developing biodegradable lipid nanoparticles for intracellular mRNA delivery and genome editing | |
Tang et al. | Efficient in vitro siRNA delivery and intramuscular gene silencing using PEG-modified PAMAM dendrimers | |
EP4034587B1 (en) | Hyperbranched cationic polymers useful as nucleic acid delivery vectors for transfecting cells | |
Lin et al. | An acid-labile block copolymer of PDMAEMA and PEG as potential carrier for intelligent gene delivery systems | |
Gao et al. | Gene-carried chitosan-linked-PEI induced high gene transfection efficiency with low toxicity and significant tumor-suppressive activity | |
Guo et al. | Image-guided and tumor-targeted drug delivery with radiolabeled unimolecular micelles | |
US10172956B2 (en) | Polymeric nanoparticles | |
US20130149783A1 (en) | Cleavable modifications to reducible poly (amido ethylenimines)s to enhance nucleotide delivery | |
Tanaka et al. | Development of lipid-like materials for RNA delivery based on intracellular environment-responsive membrane destabilization and spontaneous collapse | |
AU2014300980A1 (en) | Compositions for introducing RNA into cells | |
Zhao et al. | Tumor acidity-induced sheddable polyethylenimine-poly (trimethylene carbonate)/DNA/polyethylene glycol-2, 3-dimethylmaleicanhydride ternary complex for efficient and safe gene delivery | |
WO2008058457A1 (en) | A biodegradable crosslinked polyethyleneimine and its uses | |
Ravina et al. | Core− shell dendriplexes with sterically induced stoichiometry for gene delivery | |
Lee et al. | General structure–activity relationship for poly (glycoamidoamine) s: the effect of amine density on cytotoxicity and DNA delivery efficiency | |
US20220259597A1 (en) | Oligonucleotide antagonists for rna guided genome editing | |
Wang et al. | Tris [2-(acryloyloxy) ethyl] isocyanurate cross-linked low-molecular-weight polyethylenimine as gene delivery carriers in cell culture and dystrophic mdx mice | |
Cao et al. | Divalent folate modification on PEG: an effective strategy for improving the cellular uptake and targetability of PEGylated polyamidoamine–polyethylenimine copolymer | |
Wang et al. | Circumvent PEGylation dilemma by implementing matrix metalloproteinase-responsive chemistry for promoted tumor gene therapy | |
Boyle et al. | Molecular additives significantly enhance glycopolymer-mediated transfection of large plasmids and functional CRISPR-Cas9 transcription activation ex vivo in primary human fibroblasts and induced pluripotent stem cells | |
US20220340933A1 (en) | Nanoparticle compositions for gene therapy | |
Fang et al. | Inhalable Bottlebrush Polymer Bioconjugates as Vectors for Efficient Pulmonary Delivery of Oligonucleotides | |
WO2023131648A1 (en) | Nanoparticulate compositions for gene therapy | |
EP2907876B1 (en) | Reduction stimuli-responsive gene vector system and preparation and use thereof | |
EP4433031A1 (en) | A composition comprising a therapeutically active agent packaged within a drug delivery vehicle |
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: 23700446 Country of ref document: EP Kind code of ref document: A1 |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2023700446 Country of ref document: EP |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 2023700446 Country of ref document: EP Effective date: 20240805 |