WO2005113765A2 - Procedes d'activation de sulfatases, methodes et compositions d'utilisation associees - Google Patents
Procedes d'activation de sulfatases, methodes et compositions d'utilisation associees Download PDFInfo
- Publication number
- WO2005113765A2 WO2005113765A2 PCT/US2005/014097 US2005014097W WO2005113765A2 WO 2005113765 A2 WO2005113765 A2 WO 2005113765A2 US 2005014097 W US2005014097 W US 2005014097W WO 2005113765 A2 WO2005113765 A2 WO 2005113765A2
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- protein
- arylsulfatase
- sulfatase
- seq
- cells
- Prior art date
Links
- 108060007951 sulfatase Proteins 0.000 title claims abstract description 233
- 102000005262 Sulfatase Human genes 0.000 title claims abstract description 194
- 238000000034 method Methods 0.000 title claims abstract description 138
- 239000000203 mixture Substances 0.000 title claims abstract description 92
- 230000004913 activation Effects 0.000 title description 29
- 210000004027 cell Anatomy 0.000 claims description 200
- 102100031491 Arylsulfatase B Human genes 0.000 claims description 109
- 108010027520 N-Acetylgalactosamine-4-Sulfatase Proteins 0.000 claims description 107
- 108090000623 proteins and genes Proteins 0.000 claims description 103
- 102000004169 proteins and genes Human genes 0.000 claims description 95
- 235000018102 proteins Nutrition 0.000 claims description 92
- LSGOVYNHVSXFFJ-UHFFFAOYSA-N vanadate(3-) Chemical compound [O-][V]([O-])([O-])=O LSGOVYNHVSXFFJ-UHFFFAOYSA-N 0.000 claims description 78
- 230000014509 gene expression Effects 0.000 claims description 52
- 238000002360 preparation method Methods 0.000 claims description 44
- 102000009133 Arylsulfatases Human genes 0.000 claims description 39
- 102100022146 Arylsulfatase A Human genes 0.000 claims description 37
- 108010036867 Cerebroside-Sulfatase Proteins 0.000 claims description 37
- MTCFGRXMJLQNBG-UHFFFAOYSA-N Serine Natural products OCC(N)C(O)=O MTCFGRXMJLQNBG-UHFFFAOYSA-N 0.000 claims description 37
- 230000000694 effects Effects 0.000 claims description 36
- 210000004978 chinese hamster ovary cell Anatomy 0.000 claims description 30
- UGJBHEZMOKVTIM-UHFFFAOYSA-N N-formylglycine Chemical compound OC(=O)CNC=O UGJBHEZMOKVTIM-UHFFFAOYSA-N 0.000 claims description 29
- 239000013598 vector Substances 0.000 claims description 23
- 239000001963 growth medium Substances 0.000 claims description 19
- 239000008194 pharmaceutical composition Substances 0.000 claims description 18
- 125000003607 serino group Chemical group [H]N([H])[C@]([H])(C(=O)[*])C(O[H])([H])[H] 0.000 claims description 18
- 235000001014 amino acid Nutrition 0.000 claims description 17
- 125000000151 cysteine group Chemical group N[C@@H](CS)C(=O)* 0.000 claims description 17
- 150000007523 nucleic acids Chemical class 0.000 claims description 16
- 230000035772 mutation Effects 0.000 claims description 14
- 125000003275 alpha amino acid group Chemical group 0.000 claims description 12
- 108020004707 nucleic acids Proteins 0.000 claims description 12
- 102000039446 nucleic acids Human genes 0.000 claims description 12
- 239000003937 drug carrier Substances 0.000 claims description 11
- 230000001678 irradiating effect Effects 0.000 claims description 10
- 101000923070 Homo sapiens Arylsulfatase B Proteins 0.000 claims description 9
- 229910052720 vanadium Inorganic materials 0.000 claims description 9
- LEONUFNNVUYDNQ-UHFFFAOYSA-N vanadium atom Chemical compound [V] LEONUFNNVUYDNQ-UHFFFAOYSA-N 0.000 claims description 9
- 239000002265 redox agent Substances 0.000 claims description 8
- 102100029199 Iduronate 2-sulfatase Human genes 0.000 claims description 7
- 239000003085 diluting agent Substances 0.000 claims description 7
- 238000011534 incubation Methods 0.000 claims description 7
- 239000000546 pharmaceutical excipient Substances 0.000 claims description 7
- 102000043401 human ARSB Human genes 0.000 claims description 5
- 230000028327 secretion Effects 0.000 claims description 5
- 102100021655 Extracellular sulfatase Sulf-1 Human genes 0.000 claims description 4
- 102100021654 Extracellular sulfatase Sulf-2 Human genes 0.000 claims description 4
- 101000820630 Homo sapiens Extracellular sulfatase Sulf-1 Proteins 0.000 claims description 4
- 101000820626 Homo sapiens Extracellular sulfatase Sulf-2 Proteins 0.000 claims description 4
- 102100023282 N-acetylglucosamine-6-sulfatase Human genes 0.000 claims description 4
- 102100027661 N-sulphoglucosamine sulphohydrolase Human genes 0.000 claims description 4
- 238000012258 culturing Methods 0.000 claims description 4
- MSWZFWKMSRAUBD-GASJEMHNSA-N 2-amino-2-deoxy-D-galactopyranose Chemical compound N[C@H]1C(O)O[C@H](CO)[C@H](O)[C@@H]1O MSWZFWKMSRAUBD-GASJEMHNSA-N 0.000 claims description 3
- 102100033887 Arylsulfatase D Human genes 0.000 claims description 3
- 101710115233 Arylsulfatase D Proteins 0.000 claims description 3
- 102100033886 Arylsulfatase F Human genes 0.000 claims description 3
- 101710115249 Arylsulfatase F Proteins 0.000 claims description 3
- 102100033890 Arylsulfatase G Human genes 0.000 claims description 3
- 101710115232 Arylsulfatase G Proteins 0.000 claims description 3
- 102100023943 Arylsulfatase L Human genes 0.000 claims description 3
- 101710115246 Arylsulfatase L Proteins 0.000 claims description 3
- 101000901140 Homo sapiens Arylsulfatase A Proteins 0.000 claims description 3
- 101000829992 Homo sapiens N-acetylglucosamine-6-sulfatase Proteins 0.000 claims description 3
- 125000003047 N-acetyl group Chemical group 0.000 claims description 3
- 108010006140 N-sulfoglucosamine sulfohydrolase Proteins 0.000 claims description 3
- 108010087999 Steryl-Sulfatase Proteins 0.000 claims description 3
- 102100038021 Steryl-sulfatase Human genes 0.000 claims description 3
- MSWZFWKMSRAUBD-UHFFFAOYSA-N beta-D-galactosamine Natural products NC1C(O)OC(CO)C(O)C1O MSWZFWKMSRAUBD-UHFFFAOYSA-N 0.000 claims description 3
- 229930183167 cerebroside Natural products 0.000 claims description 3
- RIZIAUKTHDLMQX-UHFFFAOYSA-N cerebroside D Natural products CCCCCCCCCCCCCCCCC(O)C(=O)NC(C(O)C=CCCC=C(C)CCCCCCCCC)COC1OC(CO)C(O)C(O)C1O RIZIAUKTHDLMQX-UHFFFAOYSA-N 0.000 claims description 3
- JSPNNZKWADNWHI-PNANGNLXSA-N (2r)-2-hydroxy-n-[(2s,3r,4e,8e)-3-hydroxy-9-methyl-1-[(2r,3r,4s,5s,6r)-3,4,5-trihydroxy-6-(hydroxymethyl)oxan-2-yl]oxyoctadeca-4,8-dien-2-yl]heptadecanamide Chemical compound CCCCCCCCCCCCCCC[C@@H](O)C(=O)N[C@H]([C@H](O)\C=C\CC\C=C(/C)CCCCCCCCC)CO[C@@H]1O[C@H](CO)[C@@H](O)[C@H](O)[C@H]1O JSPNNZKWADNWHI-PNANGNLXSA-N 0.000 claims description 2
- 108010053927 Iduronate Sulfatase Proteins 0.000 claims 1
- 102200001976 rs121908465 Human genes 0.000 claims 1
- 102000004190 Enzymes Human genes 0.000 description 110
- 108090000790 Enzymes Proteins 0.000 description 110
- FAPWRFPIFSIZLT-UHFFFAOYSA-M Sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 description 71
- 108090000765 processed proteins & peptides Proteins 0.000 description 60
- 239000002609 medium Substances 0.000 description 36
- 229960001153 serine Drugs 0.000 description 36
- 235000004400 serine Nutrition 0.000 description 36
- 238000006243 chemical reaction Methods 0.000 description 35
- HEMHJVSKTPXQMS-UHFFFAOYSA-M Sodium hydroxide Chemical compound [OH-].[Na+] HEMHJVSKTPXQMS-UHFFFAOYSA-M 0.000 description 33
- 239000011780 sodium chloride Substances 0.000 description 33
- 239000000243 solution Substances 0.000 description 30
- MTCFGRXMJLQNBG-REOHCLBHSA-N (2S)-2-Amino-3-hydroxypropansäure Chemical compound OC[C@H](N)C(O)=O MTCFGRXMJLQNBG-REOHCLBHSA-N 0.000 description 29
- 238000001994 activation Methods 0.000 description 29
- 102000004196 processed proteins & peptides Human genes 0.000 description 29
- 238000004519 manufacturing process Methods 0.000 description 25
- 239000000872 buffer Substances 0.000 description 24
- 230000008569 process Effects 0.000 description 22
- PEDCQBHIVMGVHV-UHFFFAOYSA-N Glycerine Chemical compound OCC(O)CO PEDCQBHIVMGVHV-UHFFFAOYSA-N 0.000 description 21
- 230000001225 therapeutic effect Effects 0.000 description 21
- 238000011282 treatment Methods 0.000 description 21
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 20
- 229920002684 Sepharose Polymers 0.000 description 19
- 239000003795 chemical substances by application Substances 0.000 description 19
- 239000002299 complementary DNA Substances 0.000 description 17
- 239000000047 product Substances 0.000 description 17
- 238000003556 assay Methods 0.000 description 16
- 238000004113 cell culture Methods 0.000 description 16
- 239000003814 drug Substances 0.000 description 16
- 239000012530 fluid Substances 0.000 description 16
- 230000010412 perfusion Effects 0.000 description 16
- 102000007056 Recombinant Fusion Proteins Human genes 0.000 description 15
- 108010008281 Recombinant Fusion Proteins Proteins 0.000 description 15
- 238000004587 chromatography analysis Methods 0.000 description 15
- 229920001184 polypeptide Polymers 0.000 description 15
- 230000001419 dependent effect Effects 0.000 description 14
- 239000013604 expression vector Substances 0.000 description 14
- 238000001802 infusion Methods 0.000 description 14
- 238000000746 purification Methods 0.000 description 14
- XLYOFNOQVPJJNP-UHFFFAOYSA-N water Substances O XLYOFNOQVPJJNP-UHFFFAOYSA-N 0.000 description 14
- 235000018417 cysteine Nutrition 0.000 description 13
- 230000004048 modification Effects 0.000 description 13
- 238000012986 modification Methods 0.000 description 13
- -1 vanadate ions Chemical class 0.000 description 13
- XUJNEKJLAYXESH-UHFFFAOYSA-N cysteine Natural products SCC(N)C(O)=O XUJNEKJLAYXESH-UHFFFAOYSA-N 0.000 description 12
- 238000002641 enzyme replacement therapy Methods 0.000 description 12
- 230000012010 growth Effects 0.000 description 12
- 238000003860 storage Methods 0.000 description 12
- 239000000758 substrate Substances 0.000 description 12
- 102220565761 Arylsulfatase A_C69S_mutation Human genes 0.000 description 11
- 238000004458 analytical method Methods 0.000 description 11
- 108020004414 DNA Proteins 0.000 description 10
- 208000015439 Lysosomal storage disease Diseases 0.000 description 10
- 241001465754 Metazoa Species 0.000 description 10
- 229940024606 amino acid Drugs 0.000 description 10
- 201000010099 disease Diseases 0.000 description 10
- 208000035475 disorder Diseases 0.000 description 10
- 238000011156 evaluation Methods 0.000 description 10
- 238000002741 site-directed mutagenesis Methods 0.000 description 10
- 238000002965 ELISA Methods 0.000 description 9
- 229920002683 Glycosaminoglycan Polymers 0.000 description 9
- VMHLLURERBWHNL-UHFFFAOYSA-M Sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 description 9
- 150000001413 amino acids Chemical class 0.000 description 9
- 238000003776 cleavage reaction Methods 0.000 description 9
- 238000009472 formulation Methods 0.000 description 9
- 238000003306 harvesting Methods 0.000 description 9
- 230000001965 increasing effect Effects 0.000 description 9
- 238000011012 sanitization Methods 0.000 description 9
- 230000007017 scission Effects 0.000 description 9
- 231100000419 toxicity Toxicity 0.000 description 9
- 230000001988 toxicity Effects 0.000 description 9
- WEVYAHXRMPXWCK-UHFFFAOYSA-N Acetonitrile Chemical compound CC#N WEVYAHXRMPXWCK-UHFFFAOYSA-N 0.000 description 8
- 229920002271 DEAE-Sepharose Polymers 0.000 description 8
- 238000000338 in vitro Methods 0.000 description 8
- 238000001727 in vivo Methods 0.000 description 8
- 238000010647 peptide synthesis reaction Methods 0.000 description 8
- 125000001997 phenyl group Chemical group [H]C1=C([H])C([H])=C(*)C([H])=C1[H] 0.000 description 8
- 230000002829 reductive effect Effects 0.000 description 8
- 230000014616 translation Effects 0.000 description 8
- 230000002485 urinary effect Effects 0.000 description 8
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 7
- 108091028043 Nucleic acid sequence Proteins 0.000 description 7
- 238000007792 addition Methods 0.000 description 7
- QVGXLLKOCUKJST-UHFFFAOYSA-N atomic oxygen Chemical compound [O] QVGXLLKOCUKJST-UHFFFAOYSA-N 0.000 description 7
- 238000010923 batch production Methods 0.000 description 7
- 230000015572 biosynthetic process Effects 0.000 description 7
- 238000011210 chromatographic step Methods 0.000 description 7
- 229940079593 drug Drugs 0.000 description 7
- 238000002347 injection Methods 0.000 description 7
- 239000007924 injection Substances 0.000 description 7
- 239000007788 liquid Substances 0.000 description 7
- 230000033001 locomotion Effects 0.000 description 7
- 239000000463 material Substances 0.000 description 7
- 230000002018 overexpression Effects 0.000 description 7
- 229910052760 oxygen Inorganic materials 0.000 description 7
- 239000001301 oxygen Substances 0.000 description 7
- 239000013612 plasmid Substances 0.000 description 7
- 230000009467 reduction Effects 0.000 description 7
- 238000012360 testing method Methods 0.000 description 7
- 238000001262 western blot Methods 0.000 description 7
- FHVDTGUDJYJELY-UHFFFAOYSA-N 6-{[2-carboxy-4,5-dihydroxy-6-(phosphanyloxy)oxan-3-yl]oxy}-4,5-dihydroxy-3-phosphanyloxane-2-carboxylic acid Chemical compound O1C(C(O)=O)C(P)C(O)C(O)C1OC1C(C(O)=O)OC(OP)C(O)C1O FHVDTGUDJYJELY-UHFFFAOYSA-N 0.000 description 6
- CSCPPACGZOOCGX-UHFFFAOYSA-N Acetone Chemical compound CC(C)=O CSCPPACGZOOCGX-UHFFFAOYSA-N 0.000 description 6
- 101710096421 Iduronate 2-sulfatase Proteins 0.000 description 6
- 241000700605 Viruses Species 0.000 description 6
- 229940072056 alginate Drugs 0.000 description 6
- 235000010443 alginic acid Nutrition 0.000 description 6
- 229920000615 alginic acid Polymers 0.000 description 6
- 230000008901 benefit Effects 0.000 description 6
- 239000011575 calcium Substances 0.000 description 6
- 239000003153 chemical reaction reagent Substances 0.000 description 6
- 230000007423 decrease Effects 0.000 description 6
- 238000002474 experimental method Methods 0.000 description 6
- 238000001914 filtration Methods 0.000 description 6
- OVBPIULPVIDEAO-LBPRGKRZSA-N folic acid Chemical compound C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-LBPRGKRZSA-N 0.000 description 6
- 238000007913 intrathecal administration Methods 0.000 description 6
- 239000012528 membrane Substances 0.000 description 6
- 229910021645 metal ion Inorganic materials 0.000 description 6
- 235000015097 nutrients Nutrition 0.000 description 6
- 238000005457 optimization Methods 0.000 description 6
- 230000003647 oxidation Effects 0.000 description 6
- 238000007254 oxidation reaction Methods 0.000 description 6
- 239000012071 phase Substances 0.000 description 6
- 150000003839 salts Chemical class 0.000 description 6
- 239000000523 sample Substances 0.000 description 6
- 239000006228 supernatant Substances 0.000 description 6
- 210000001519 tissue Anatomy 0.000 description 6
- 238000013518 transcription Methods 0.000 description 6
- 230000035897 transcription Effects 0.000 description 6
- 230000003612 virological effect Effects 0.000 description 6
- 102100031688 N-acetylgalactosamine-6-sulfatase Human genes 0.000 description 5
- 239000004480 active ingredient Substances 0.000 description 5
- 239000011324 bead Substances 0.000 description 5
- 230000037396 body weight Effects 0.000 description 5
- 230000015556 catabolic process Effects 0.000 description 5
- 230000007812 deficiency Effects 0.000 description 5
- 238000006731 degradation reaction Methods 0.000 description 5
- 238000011026 diafiltration Methods 0.000 description 5
- 238000010790 dilution Methods 0.000 description 5
- 239000012895 dilution Substances 0.000 description 5
- 239000006185 dispersion Substances 0.000 description 5
- 238000004895 liquid chromatography mass spectrometry Methods 0.000 description 5
- 210000004185 liver Anatomy 0.000 description 5
- 230000002132 lysosomal effect Effects 0.000 description 5
- 239000003550 marker Substances 0.000 description 5
- 238000012544 monitoring process Methods 0.000 description 5
- 239000002773 nucleotide Substances 0.000 description 5
- 125000003729 nucleotide group Chemical group 0.000 description 5
- 239000000843 powder Substances 0.000 description 5
- 239000011347 resin Substances 0.000 description 5
- 229920005989 resin Polymers 0.000 description 5
- 210000003705 ribosome Anatomy 0.000 description 5
- 238000013341 scale-up Methods 0.000 description 5
- CMZUMMUJMWNLFH-UHFFFAOYSA-N sodium metavanadate Chemical compound [Na+].[O-][V](=O)=O CMZUMMUJMWNLFH-UHFFFAOYSA-N 0.000 description 5
- 239000007790 solid phase Substances 0.000 description 5
- 230000002459 sustained effect Effects 0.000 description 5
- 208000024891 symptom Diseases 0.000 description 5
- 238000003786 synthesis reaction Methods 0.000 description 5
- 229940124597 therapeutic agent Drugs 0.000 description 5
- 238000002560 therapeutic procedure Methods 0.000 description 5
- 231100000331 toxic Toxicity 0.000 description 5
- 230000002588 toxic effect Effects 0.000 description 5
- 238000012546 transfer Methods 0.000 description 5
- 238000000108 ultra-filtration Methods 0.000 description 5
- 125000005287 vanadyl group Chemical group 0.000 description 5
- 238000005406 washing Methods 0.000 description 5
- 108091003079 Bovine Serum Albumin Proteins 0.000 description 4
- 108091026890 Coding region Proteins 0.000 description 4
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N Ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 4
- 241000282326 Felis catus Species 0.000 description 4
- 229930182816 L-glutamine Natural products 0.000 description 4
- 208000000149 Multiple Sulfatase Deficiency Disease Diseases 0.000 description 4
- 208000035032 Multiple sulfatase deficiency Diseases 0.000 description 4
- 108010021466 Mutant Proteins Proteins 0.000 description 4
- 102000008300 Mutant Proteins Human genes 0.000 description 4
- ISWSIDIOOBJBQZ-UHFFFAOYSA-N Phenol Chemical compound OC1=CC=CC=C1 ISWSIDIOOBJBQZ-UHFFFAOYSA-N 0.000 description 4
- 239000004793 Polystyrene Substances 0.000 description 4
- IQFYYKKMVGJFEH-XLPZGREQSA-N Thymidine Chemical compound O=C1NC(=O)C(C)=CN1[C@@H]1O[C@H](CO)[C@@H](O)C1 IQFYYKKMVGJFEH-XLPZGREQSA-N 0.000 description 4
- 230000001154 acute effect Effects 0.000 description 4
- 210000004102 animal cell Anatomy 0.000 description 4
- 238000013459 approach Methods 0.000 description 4
- 239000002585 base Substances 0.000 description 4
- 229910052791 calcium Inorganic materials 0.000 description 4
- 230000010261 cell growth Effects 0.000 description 4
- 230000001413 cellular effect Effects 0.000 description 4
- 210000003169 central nervous system Anatomy 0.000 description 4
- 230000009920 chelation Effects 0.000 description 4
- 238000013375 chromatographic separation Methods 0.000 description 4
- 238000010511 deprotection reaction Methods 0.000 description 4
- 238000001514 detection method Methods 0.000 description 4
- 239000002612 dispersion medium Substances 0.000 description 4
- 239000012149 elution buffer Substances 0.000 description 4
- 238000005516 engineering process Methods 0.000 description 4
- 239000012634 fragment Substances 0.000 description 4
- 239000011521 glass Substances 0.000 description 4
- 230000036541 health Effects 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 238000006460 hydrolysis reaction Methods 0.000 description 4
- FDGQSTZJBFJUBT-UHFFFAOYSA-N hypoxanthine Chemical compound O=C1NC=NC2=C1NC=N2 FDGQSTZJBFJUBT-UHFFFAOYSA-N 0.000 description 4
- 238000001990 intravenous administration Methods 0.000 description 4
- 230000007246 mechanism Effects 0.000 description 4
- 244000005700 microbiome Species 0.000 description 4
- 229920001542 oligosaccharide Polymers 0.000 description 4
- 150000002482 oligosaccharides Chemical class 0.000 description 4
- 230000008488 polyadenylation Effects 0.000 description 4
- 229920002223 polystyrene Polymers 0.000 description 4
- 230000004481 post-translational protein modification Effects 0.000 description 4
- 238000012545 processing Methods 0.000 description 4
- 230000000644 propagated effect Effects 0.000 description 4
- 230000020978 protein processing Effects 0.000 description 4
- 238000010188 recombinant method Methods 0.000 description 4
- 230000001105 regulatory effect Effects 0.000 description 4
- 230000004044 response Effects 0.000 description 4
- 210000002966 serum Anatomy 0.000 description 4
- 239000001632 sodium acetate Substances 0.000 description 4
- 235000017281 sodium acetate Nutrition 0.000 description 4
- 239000002904 solvent Substances 0.000 description 4
- 239000012536 storage buffer Substances 0.000 description 4
- 238000001890 transfection Methods 0.000 description 4
- 230000003442 weekly effect Effects 0.000 description 4
- QKNYBSVHEMOAJP-UHFFFAOYSA-N 2-amino-2-(hydroxymethyl)propane-1,3-diol;hydron;chloride Chemical compound Cl.OCC(N)(CO)CO QKNYBSVHEMOAJP-UHFFFAOYSA-N 0.000 description 3
- 229930183010 Amphotericin Natural products 0.000 description 3
- QGGFZZLFKABGNL-UHFFFAOYSA-N Amphotericin A Natural products OC1C(N)C(O)C(C)OC1OC1C=CC=CC=CC=CCCC=CC=CC(C)C(O)C(C)C(C)OC(=O)CC(O)CC(O)CCC(O)C(O)CC(O)CC(O)(CC(O)C2C(O)=O)OC2C1 QGGFZZLFKABGNL-UHFFFAOYSA-N 0.000 description 3
- DCXYFEDJOCDNAF-UHFFFAOYSA-N Asparagine Natural products OC(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-N 0.000 description 3
- 241000894006 Bacteria Species 0.000 description 3
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 3
- CURLTUGMZLYLDI-UHFFFAOYSA-N Carbon dioxide Chemical compound O=C=O CURLTUGMZLYLDI-UHFFFAOYSA-N 0.000 description 3
- RYGMFSIKBFXOCR-UHFFFAOYSA-N Copper Chemical compound [Cu] RYGMFSIKBFXOCR-UHFFFAOYSA-N 0.000 description 3
- 241000699800 Cricetinae Species 0.000 description 3
- RTZKZFJDLAIYFH-UHFFFAOYSA-N Diethyl ether Chemical compound CCOCC RTZKZFJDLAIYFH-UHFFFAOYSA-N 0.000 description 3
- 239000012591 Dulbecco’s Phosphate Buffered Saline Substances 0.000 description 3
- CEAZRRDELHUEMR-URQXQFDESA-N Gentamicin Chemical compound O1[C@H](C(C)NC)CC[C@@H](N)[C@H]1O[C@H]1[C@H](O)[C@@H](O[C@@H]2[C@@H]([C@@H](NC)[C@@](C)(O)CO2)O)[C@H](N)C[C@@H]1N CEAZRRDELHUEMR-URQXQFDESA-N 0.000 description 3
- 229930182566 Gentamicin Natural products 0.000 description 3
- 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 3
- 241000282412 Homo Species 0.000 description 3
- 101001066305 Homo sapiens N-acetylgalactosamine-6-sulfatase Proteins 0.000 description 3
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 3
- BZLVMXJERCGZMT-UHFFFAOYSA-N Methyl tert-butyl ether Chemical compound COC(C)(C)C BZLVMXJERCGZMT-UHFFFAOYSA-N 0.000 description 3
- 102000003505 Myosin Human genes 0.000 description 3
- 108060008487 Myosin Proteins 0.000 description 3
- OVBPIULPVIDEAO-UHFFFAOYSA-N N-Pteroyl-L-glutaminsaeure Natural products C=1N=C2NC(N)=NC(=O)C2=NC=1CNC1=CC=C(C(=O)NC(CCC(O)=O)C(O)=O)C=C1 OVBPIULPVIDEAO-UHFFFAOYSA-N 0.000 description 3
- 108091034117 Oligonucleotide Proteins 0.000 description 3
- 241000283973 Oryctolagus cuniculus Species 0.000 description 3
- DNIAPMSPPWPWGF-UHFFFAOYSA-N Propylene glycol Chemical compound CC(O)CO DNIAPMSPPWPWGF-UHFFFAOYSA-N 0.000 description 3
- 241000700159 Rattus Species 0.000 description 3
- 230000009471 action Effects 0.000 description 3
- 238000013019 agitation Methods 0.000 description 3
- 150000001299 aldehydes Chemical class 0.000 description 3
- 229940009444 amphotericin Drugs 0.000 description 3
- APKFDSVGJQXUKY-INPOYWNPSA-N amphotericin B Chemical compound O[C@H]1[C@@H](N)[C@H](O)[C@@H](C)O[C@H]1O[C@H]1/C=C/C=C/C=C/C=C/C=C/C=C/C=C/[C@H](C)[C@@H](O)[C@@H](C)[C@H](C)OC(=O)C[C@H](O)C[C@H](O)CC[C@@H](O)[C@H](O)C[C@H](O)C[C@](O)(C[C@H](O)[C@H]2C(O)=O)O[C@H]2C1 APKFDSVGJQXUKY-INPOYWNPSA-N 0.000 description 3
- 238000010171 animal model Methods 0.000 description 3
- 239000003242 anti bacterial agent Substances 0.000 description 3
- 230000000844 anti-bacterial effect Effects 0.000 description 3
- 239000003429 antifungal agent Substances 0.000 description 3
- 229940121375 antifungal agent Drugs 0.000 description 3
- 239000007864 aqueous solution Substances 0.000 description 3
- 125000000637 arginyl group Chemical group N[C@@H](CCCNC(N)=N)C(=O)* 0.000 description 3
- 229960001230 asparagine Drugs 0.000 description 3
- 235000009582 asparagine Nutrition 0.000 description 3
- 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 3
- 230000000903 blocking effect Effects 0.000 description 3
- 239000001045 blue dye Substances 0.000 description 3
- 239000006227 byproduct Substances 0.000 description 3
- 239000002775 capsule Substances 0.000 description 3
- 239000012539 chromatography resin Substances 0.000 description 3
- 230000004186 co-expression Effects 0.000 description 3
- 238000000576 coating method Methods 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 239000003636 conditioned culture medium Substances 0.000 description 3
- 239000000356 contaminant Substances 0.000 description 3
- 229910052802 copper Inorganic materials 0.000 description 3
- 239000010949 copper Substances 0.000 description 3
- 238000009295 crossflow filtration Methods 0.000 description 3
- 230000003247 decreasing effect Effects 0.000 description 3
- 230000001627 detrimental effect Effects 0.000 description 3
- 230000018109 developmental process Effects 0.000 description 3
- 239000012153 distilled water Substances 0.000 description 3
- 239000003623 enhancer Substances 0.000 description 3
- 239000012091 fetal bovine serum Substances 0.000 description 3
- 229960000304 folic acid Drugs 0.000 description 3
- 235000019152 folic acid Nutrition 0.000 description 3
- 239000011724 folic acid Substances 0.000 description 3
- 239000012537 formulation buffer Substances 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 239000000499 gel Substances 0.000 description 3
- 230000002068 genetic effect Effects 0.000 description 3
- 239000008103 glucose Substances 0.000 description 3
- ZDXPYRJPNDTMRX-UHFFFAOYSA-N glutamine Natural products OC(=O)C(N)CCC(N)=O ZDXPYRJPNDTMRX-UHFFFAOYSA-N 0.000 description 3
- 230000007062 hydrolysis Effects 0.000 description 3
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 3
- 230000006872 improvement Effects 0.000 description 3
- 230000001939 inductive effect Effects 0.000 description 3
- 239000004615 ingredient Substances 0.000 description 3
- 230000003993 interaction Effects 0.000 description 3
- 230000003834 intracellular effect Effects 0.000 description 3
- 239000007951 isotonicity adjuster Substances 0.000 description 3
- 210000000265 leukocyte Anatomy 0.000 description 3
- 230000000670 limiting effect Effects 0.000 description 3
- 210000003712 lysosome Anatomy 0.000 description 3
- 230000001868 lysosomic effect Effects 0.000 description 3
- 210000004962 mammalian cell Anatomy 0.000 description 3
- 238000013507 mapping Methods 0.000 description 3
- 238000005259 measurement Methods 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 208000005340 mucopolysaccharidosis III Diseases 0.000 description 3
- 208000011045 mucopolysaccharidosis type 3 Diseases 0.000 description 3
- 239000002245 particle Substances 0.000 description 3
- 230000037361 pathway Effects 0.000 description 3
- 239000008188 pellet Substances 0.000 description 3
- 239000008363 phosphate buffer Substances 0.000 description 3
- 229920001223 polyethylene glycol Polymers 0.000 description 3
- 239000012460 protein solution Substances 0.000 description 3
- 102000005962 receptors Human genes 0.000 description 3
- 108020003175 receptors Proteins 0.000 description 3
- 238000004007 reversed phase HPLC Methods 0.000 description 3
- 239000012475 sodium chloride buffer Substances 0.000 description 3
- 239000007787 solid Substances 0.000 description 3
- 150000003431 steroids Chemical class 0.000 description 3
- 230000002103 transcriptional effect Effects 0.000 description 3
- PIEPQKCYPFFYMG-UHFFFAOYSA-N tris acetate Chemical compound CC(O)=O.OCC(N)(CO)CO PIEPQKCYPFFYMG-UHFFFAOYSA-N 0.000 description 3
- 210000002700 urine Anatomy 0.000 description 3
- 230000035899 viability Effects 0.000 description 3
- 239000011534 wash buffer Substances 0.000 description 3
- YBJHBAHKTGYVGT-ZKWXMUAHSA-N (+)-Biotin Chemical compound N1C(=O)N[C@@H]2[C@H](CCCCC(=O)O)SC[C@@H]21 YBJHBAHKTGYVGT-ZKWXMUAHSA-N 0.000 description 2
- QGZKDVFQNNGYKY-UHFFFAOYSA-O Ammonium Chemical compound [NH4+] QGZKDVFQNNGYKY-UHFFFAOYSA-O 0.000 description 2
- 241000272517 Anseriformes Species 0.000 description 2
- 108010053481 Antifreeze Proteins Proteins 0.000 description 2
- DWRXFEITVBNRMK-UHFFFAOYSA-N Beta-D-1-Arabinofuranosylthymine Natural products O=C1NC(=O)C(C)=CN1C1C(O)C(O)C(CO)O1 DWRXFEITVBNRMK-UHFFFAOYSA-N 0.000 description 2
- 241000283690 Bos taurus Species 0.000 description 2
- 238000009010 Bradford assay Methods 0.000 description 2
- BHPQYMZQTOCNFJ-UHFFFAOYSA-N Calcium cation Chemical compound [Ca+2] BHPQYMZQTOCNFJ-UHFFFAOYSA-N 0.000 description 2
- UXVMQQNJUSDDNG-UHFFFAOYSA-L Calcium chloride Chemical compound [Cl-].[Cl-].[Ca+2] UXVMQQNJUSDDNG-UHFFFAOYSA-L 0.000 description 2
- 241000283707 Capra Species 0.000 description 2
- 108010035563 Chloramphenicol O-acetyltransferase Proteins 0.000 description 2
- 108020004705 Codon Proteins 0.000 description 2
- 102220548356 Cyclic pyranopterin monophosphate synthase_C84S_mutation Human genes 0.000 description 2
- 102100024746 Dihydrofolate reductase Human genes 0.000 description 2
- YQYJSBFKSSDGFO-UHFFFAOYSA-N Epihygromycin Natural products OC1C(O)C(C(=O)C)OC1OC(C(=C1)O)=CC=C1C=C(C)C(=O)NC1C(O)C(O)C2OCOC2C1O YQYJSBFKSSDGFO-UHFFFAOYSA-N 0.000 description 2
- 241000588724 Escherichia coli Species 0.000 description 2
- 241000233866 Fungi Species 0.000 description 2
- 241000287828 Gallus gallus Species 0.000 description 2
- 102000005720 Glutathione transferase Human genes 0.000 description 2
- 108010070675 Glutathione transferase Proteins 0.000 description 2
- DHMQDGOQFOQNFH-UHFFFAOYSA-N Glycine Chemical compound NCC(O)=O DHMQDGOQFOQNFH-UHFFFAOYSA-N 0.000 description 2
- 108091027305 Heteroduplex Proteins 0.000 description 2
- 206010021033 Hypomenorrhoea Diseases 0.000 description 2
- UGQMRVRMYYASKQ-UHFFFAOYSA-N Hypoxanthine nucleoside Natural products OC1C(O)C(CO)OC1N1C(NC=NC2=O)=C2N=C1 UGQMRVRMYYASKQ-UHFFFAOYSA-N 0.000 description 2
- SIKJAQJRHWYJAI-UHFFFAOYSA-N Indole Chemical compound C1=CC=C2NC=CC2=C1 SIKJAQJRHWYJAI-UHFFFAOYSA-N 0.000 description 2
- ONIBWKKTOPOVIA-BYPYZUCNSA-N L-Proline Chemical compound OC(=O)[C@@H]1CCCN1 ONIBWKKTOPOVIA-BYPYZUCNSA-N 0.000 description 2
- 108010015372 Low Density Lipoprotein Receptor-Related Protein-2 Proteins 0.000 description 2
- 102100021922 Low-density lipoprotein receptor-related protein 2 Human genes 0.000 description 2
- 108060001084 Luciferase Proteins 0.000 description 2
- 239000005089 Luciferase Substances 0.000 description 2
- 241000124008 Mammalia Species 0.000 description 2
- 206010028095 Mucopolysaccharidosis IV Diseases 0.000 description 2
- 125000001429 N-terminal alpha-amino-acid group Chemical group 0.000 description 2
- 241001494479 Pecora Species 0.000 description 2
- BELBBZDIHDAJOR-UHFFFAOYSA-N Phenolsulfonephthalein Chemical compound C1=CC(O)=CC=C1C1(C=2C=CC(O)=CC=2)C2=CC=CC=C2S(=O)(=O)O1 BELBBZDIHDAJOR-UHFFFAOYSA-N 0.000 description 2
- 239000004743 Polypropylene Substances 0.000 description 2
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 2
- ONIBWKKTOPOVIA-UHFFFAOYSA-N Proline Natural products OC(=O)C1CCCN1 ONIBWKKTOPOVIA-UHFFFAOYSA-N 0.000 description 2
- 108010076504 Protein Sorting Signals Proteins 0.000 description 2
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 2
- CDBYLPFSWZWCQE-UHFFFAOYSA-L Sodium Carbonate Chemical compound [Na+].[Na+].[O-]C([O-])=O CDBYLPFSWZWCQE-UHFFFAOYSA-L 0.000 description 2
- 102000006601 Thymidine Kinase Human genes 0.000 description 2
- 108020004440 Thymidine kinase Proteins 0.000 description 2
- 241000723873 Tobacco mosaic virus Species 0.000 description 2
- XSQUKJJJFZCRTK-UHFFFAOYSA-N Urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 description 2
- 239000003070 absorption delaying agent Substances 0.000 description 2
- 238000010521 absorption reaction Methods 0.000 description 2
- 238000009825 accumulation Methods 0.000 description 2
- 239000002253 acid Substances 0.000 description 2
- 239000013543 active substance Substances 0.000 description 2
- 230000002411 adverse Effects 0.000 description 2
- 230000002528 anti-freeze Effects 0.000 description 2
- 239000000739 antihistaminic agent Substances 0.000 description 2
- 239000012131 assay buffer Substances 0.000 description 2
- 230000001580 bacterial effect Effects 0.000 description 2
- IQFYYKKMVGJFEH-UHFFFAOYSA-N beta-L-thymidine Natural products O=C1NC(=O)C(C)=CN1C1OC(CO)C(O)C1 IQFYYKKMVGJFEH-UHFFFAOYSA-N 0.000 description 2
- 230000008033 biological extinction Effects 0.000 description 2
- 230000008499 blood brain barrier function Effects 0.000 description 2
- 210000000988 bone and bone Anatomy 0.000 description 2
- 229910001424 calcium ion Inorganic materials 0.000 description 2
- 235000013877 carbamide Nutrition 0.000 description 2
- 229910002092 carbon dioxide Inorganic materials 0.000 description 2
- 238000006555 catalytic reaction Methods 0.000 description 2
- 238000005119 centrifugation Methods 0.000 description 2
- 210000001175 cerebrospinal fluid Anatomy 0.000 description 2
- 230000008859 change Effects 0.000 description 2
- 235000013330 chicken meat Nutrition 0.000 description 2
- OSASVXMJTNOKOY-UHFFFAOYSA-N chlorobutanol Chemical compound CC(C)(O)C(Cl)(Cl)Cl OSASVXMJTNOKOY-UHFFFAOYSA-N 0.000 description 2
- 230000024203 complement activation Effects 0.000 description 2
- 238000011109 contamination Methods 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
- DDRJAANPRJIHGJ-UHFFFAOYSA-N creatinine Chemical compound CN1CC(=O)NC1=N DDRJAANPRJIHGJ-UHFFFAOYSA-N 0.000 description 2
- 125000004122 cyclic group Chemical group 0.000 description 2
- 230000006378 damage Effects 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 238000003745 diagnosis Methods 0.000 description 2
- 108020001096 dihydrofolate reductase Proteins 0.000 description 2
- ZZVUWRFHKOJYTH-UHFFFAOYSA-N diphenhydramine Chemical compound C=1C=CC=CC=1C(OCCN(C)C)C1=CC=CC=C1 ZZVUWRFHKOJYTH-UHFFFAOYSA-N 0.000 description 2
- 238000009826 distribution Methods 0.000 description 2
- GNTDGMZSJNCJKK-UHFFFAOYSA-N divanadium pentaoxide Chemical compound O=[V](=O)O[V](=O)=O GNTDGMZSJNCJKK-UHFFFAOYSA-N 0.000 description 2
- 239000002552 dosage form Substances 0.000 description 2
- 238000012377 drug delivery Methods 0.000 description 2
- 229940126534 drug product Drugs 0.000 description 2
- 238000010828 elution Methods 0.000 description 2
- 238000005837 enolization reaction Methods 0.000 description 2
- 239000006167 equilibration buffer Substances 0.000 description 2
- 230000005284 excitation Effects 0.000 description 2
- 210000002950 fibroblast Anatomy 0.000 description 2
- 238000005194 fractionation Methods 0.000 description 2
- 230000009931 harmful effect Effects 0.000 description 2
- HNDVDQJCIGZPNO-UHFFFAOYSA-N histidine Natural products OC(=O)C(N)CC1=CN=CN1 HNDVDQJCIGZPNO-UHFFFAOYSA-N 0.000 description 2
- 210000005260 human cell Anatomy 0.000 description 2
- 238000007654 immersion Methods 0.000 description 2
- 238000002513 implantation Methods 0.000 description 2
- 238000000099 in vitro assay Methods 0.000 description 2
- 230000002779 inactivation Effects 0.000 description 2
- 230000000977 initiatory effect Effects 0.000 description 2
- NOESYZHRGYRDHS-UHFFFAOYSA-N insulin Chemical compound N1C(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(NC(=O)CN)C(C)CC)CSSCC(C(NC(CO)C(=O)NC(CC(C)C)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CCC(N)=O)C(=O)NC(CC(C)C)C(=O)NC(CCC(O)=O)C(=O)NC(CC(N)=O)C(=O)NC(CC=2C=CC(O)=CC=2)C(=O)NC(CSSCC(NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2C=CC(O)=CC=2)NC(=O)C(CC(C)C)NC(=O)C(C)NC(=O)C(CCC(O)=O)NC(=O)C(C(C)C)NC(=O)C(CC(C)C)NC(=O)C(CC=2NC=NC=2)NC(=O)C(CO)NC(=O)CNC2=O)C(=O)NCC(=O)NC(CCC(O)=O)C(=O)NC(CCCNC(N)=N)C(=O)NCC(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC=CC=3)C(=O)NC(CC=3C=CC(O)=CC=3)C(=O)NC(C(C)O)C(=O)N3C(CCC3)C(=O)NC(CCCCN)C(=O)NC(C)C(O)=O)C(=O)NC(CC(N)=O)C(O)=O)=O)NC(=O)C(C(C)CC)NC(=O)C(CO)NC(=O)C(C(C)O)NC(=O)C1CSSCC2NC(=O)C(CC(C)C)NC(=O)C(NC(=O)C(CCC(N)=O)NC(=O)C(CC(N)=O)NC(=O)C(NC(=O)C(N)CC=1C=CC=CC=1)C(C)C)CC1=CN=CN1 NOESYZHRGYRDHS-UHFFFAOYSA-N 0.000 description 2
- 238000007912 intraperitoneal administration Methods 0.000 description 2
- 238000004255 ion exchange chromatography Methods 0.000 description 2
- 238000001155 isoelectric focusing Methods 0.000 description 2
- 210000003734 kidney Anatomy 0.000 description 2
- 239000003446 ligand Substances 0.000 description 2
- 238000011068 loading method Methods 0.000 description 2
- 238000004949 mass spectrometry Methods 0.000 description 2
- BDAGIHXWWSANSR-UHFFFAOYSA-N methanoic acid Natural products OC=O BDAGIHXWWSANSR-UHFFFAOYSA-N 0.000 description 2
- 230000037230 mobility Effects 0.000 description 2
- 201000002273 mucopolysaccharidosis II Diseases 0.000 description 2
- 208000022018 mucopolysaccharidosis type 2 Diseases 0.000 description 2
- 208000010978 mucopolysaccharidosis type 4 Diseases 0.000 description 2
- 238000002703 mutagenesis Methods 0.000 description 2
- 231100000350 mutagenesis Toxicity 0.000 description 2
- 238000010606 normalization Methods 0.000 description 2
- 210000000056 organ Anatomy 0.000 description 2
- 238000006213 oxygenation reaction Methods 0.000 description 2
- 238000007911 parenteral administration Methods 0.000 description 2
- 239000000825 pharmaceutical preparation Substances 0.000 description 2
- 229960003531 phenolsulfonphthalein Drugs 0.000 description 2
- 238000007539 photo-oxidation reaction Methods 0.000 description 2
- 238000006552 photochemical reaction Methods 0.000 description 2
- 238000006303 photolysis reaction Methods 0.000 description 2
- 230000015843 photosynthesis, light reaction Effects 0.000 description 2
- 108091033319 polynucleotide Proteins 0.000 description 2
- 102000040430 polynucleotide Human genes 0.000 description 2
- 239000002157 polynucleotide Substances 0.000 description 2
- 229920001155 polypropylene Polymers 0.000 description 2
- 230000001323 posttranslational effect Effects 0.000 description 2
- 239000011591 potassium Substances 0.000 description 2
- 229910052700 potassium Inorganic materials 0.000 description 2
- 230000004952 protein activity Effects 0.000 description 2
- 230000009325 pulmonary function Effects 0.000 description 2
- RXWNCPJZOCPEPQ-NVWDDTSBSA-N puromycin Chemical compound C1=CC(OC)=CC=C1C[C@H](N)C(=O)N[C@H]1[C@@H](O)[C@H](N2C3=NC=NC(=C3N=C2)N(C)C)O[C@@H]1CO RXWNCPJZOCPEPQ-NVWDDTSBSA-N 0.000 description 2
- 238000003908 quality control method Methods 0.000 description 2
- 230000003134 recirculating effect Effects 0.000 description 2
- 238000011084 recovery Methods 0.000 description 2
- 230000022532 regulation of transcription, DNA-dependent Effects 0.000 description 2
- 238000009877 rendering Methods 0.000 description 2
- 230000000717 retained effect Effects 0.000 description 2
- 230000007958 sleep Effects 0.000 description 2
- 239000001488 sodium phosphate Substances 0.000 description 2
- 229910000162 sodium phosphate Inorganic materials 0.000 description 2
- 239000012064 sodium phosphate buffer Substances 0.000 description 2
- 210000000952 spleen Anatomy 0.000 description 2
- 230000001954 sterilising effect Effects 0.000 description 2
- 239000011550 stock solution Substances 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 238000006467 substitution reaction Methods 0.000 description 2
- QAOWNCQODCNURD-UHFFFAOYSA-L sulfate group Chemical group S(=O)(=O)([O-])[O-] QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 2
- 239000004094 surface-active agent Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 229940104230 thymidine Drugs 0.000 description 2
- 230000031998 transcytosis Effects 0.000 description 2
- GETQZCLCWQTVFV-UHFFFAOYSA-N trimethylamine Chemical compound CN(C)C GETQZCLCWQTVFV-UHFFFAOYSA-N 0.000 description 2
- 239000003656 tris buffered saline Substances 0.000 description 2
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 description 2
- 229910001456 vanadium ion Inorganic materials 0.000 description 2
- 239000003981 vehicle Substances 0.000 description 2
- LINZYZMEBMKKIT-UHFFFAOYSA-N (2,5-dioxopyrrolidin-1-yl) n-quinolin-6-ylcarbamate Chemical compound C=1C=C2N=CC=CC2=CC=1NC(=O)ON1C(=O)CCC1=O LINZYZMEBMKKIT-UHFFFAOYSA-N 0.000 description 1
- 125000003088 (fluoren-9-ylmethoxy)carbonyl group Chemical group 0.000 description 1
- 108091032973 (ribonucleotides)n+m Proteins 0.000 description 1
- FSJSYDFBTIVUFD-SUKNRPLKSA-N (z)-4-hydroxypent-3-en-2-one;oxovanadium Chemical compound [V]=O.C\C(O)=C\C(C)=O.C\C(O)=C\C(C)=O FSJSYDFBTIVUFD-SUKNRPLKSA-N 0.000 description 1
- DHBXNPKRAUYBTH-UHFFFAOYSA-N 1,1-ethanedithiol Chemical compound CC(S)S DHBXNPKRAUYBTH-UHFFFAOYSA-N 0.000 description 1
- IXPNQXFRVYWDDI-UHFFFAOYSA-N 1-methyl-2,4-dioxo-1,3-diazinane-5-carboximidamide Chemical compound CN1CC(C(N)=N)C(=O)NC1=O IXPNQXFRVYWDDI-UHFFFAOYSA-N 0.000 description 1
- IIZPXYDJLKNOIY-JXPKJXOSSA-N 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine Chemical compound CCCCCCCCCCCCCCCC(=O)OC[C@H](COP([O-])(=O)OCC[N+](C)(C)C)OC(=O)CCC\C=C/C\C=C/C\C=C/C\C=C/CCCCC IIZPXYDJLKNOIY-JXPKJXOSSA-N 0.000 description 1
- PAJMKGZZBBTTOY-UHFFFAOYSA-N 2-[[2-hydroxy-1-(3-hydroxyoctyl)-2,3,3a,4,9,9a-hexahydro-1h-cyclopenta[g]naphthalen-5-yl]oxy]acetic acid Chemical compound C1=CC=C(OCC(O)=O)C2=C1CC1C(CCC(O)CCCCC)C(O)CC1C2 PAJMKGZZBBTTOY-UHFFFAOYSA-N 0.000 description 1
- IVLXQGJVBGMLRR-UHFFFAOYSA-N 2-aminoacetic acid;hydron;chloride Chemical compound Cl.NCC(O)=O IVLXQGJVBGMLRR-UHFFFAOYSA-N 0.000 description 1
- 102220541122 3 beta-hydroxysteroid dehydrogenase/Delta 5->4-isomerase type 1_C87S_mutation Human genes 0.000 description 1
- 102220541131 3 beta-hydroxysteroid dehydrogenase/Delta 5->4-isomerase type 1_C88S_mutation Human genes 0.000 description 1
- ZOOGRGPOEVQQDX-UUOKFMHZSA-N 3',5'-cyclic GMP Chemical compound C([C@H]1O2)OP(O)(=O)O[C@H]1[C@@H](O)[C@@H]2N1C(N=C(NC2=O)N)=C2N=C1 ZOOGRGPOEVQQDX-UUOKFMHZSA-N 0.000 description 1
- OSWFIVFLDKOXQC-UHFFFAOYSA-N 4-(3-methoxyphenyl)aniline Chemical compound COC1=CC=CC(C=2C=CC(N)=CC=2)=C1 OSWFIVFLDKOXQC-UHFFFAOYSA-N 0.000 description 1
- FWMNVWWHGCHHJJ-SKKKGAJSSA-N 4-amino-1-[(2r)-6-amino-2-[[(2r)-2-[[(2r)-2-[[(2r)-2-amino-3-phenylpropanoyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]hexanoyl]piperidine-4-carboxylic acid Chemical compound C([C@H](C(=O)N[C@H](CC(C)C)C(=O)N[C@H](CCCCN)C(=O)N1CCC(N)(CC1)C(O)=O)NC(=O)[C@H](N)CC=1C=CC=CC=1)C1=CC=CC=C1 FWMNVWWHGCHHJJ-SKKKGAJSSA-N 0.000 description 1
- HSHNITRMYYLLCV-UHFFFAOYSA-N 4-methylumbelliferone Chemical compound C1=C(O)C=CC2=C1OC(=O)C=C2C HSHNITRMYYLLCV-UHFFFAOYSA-N 0.000 description 1
- 208000004998 Abdominal Pain Diseases 0.000 description 1
- 102000007469 Actins Human genes 0.000 description 1
- 108010085238 Actins Proteins 0.000 description 1
- 108010024223 Adenine phosphoribosyltransferase Proteins 0.000 description 1
- 206010067484 Adverse reaction Diseases 0.000 description 1
- 102000002260 Alkaline Phosphatase Human genes 0.000 description 1
- 108020004774 Alkaline Phosphatase Proteins 0.000 description 1
- 108010039224 Amidophosphoribosyltransferase Proteins 0.000 description 1
- 241001244729 Apalis Species 0.000 description 1
- 125000001433 C-terminal amino-acid group Chemical group 0.000 description 1
- 241000282465 Canis Species 0.000 description 1
- 241000282472 Canis lupus familiaris Species 0.000 description 1
- 241000701489 Cauliflower mosaic virus Species 0.000 description 1
- 241000700198 Cavia Species 0.000 description 1
- 229920001661 Chitosan Polymers 0.000 description 1
- 239000005496 Chlorsulfuron Substances 0.000 description 1
- 208000006069 Corneal Opacity Diseases 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- 208000028399 Critical Illness Diseases 0.000 description 1
- 102220530361 Cysteine protease ATG4B_C78S_mutation Human genes 0.000 description 1
- 241000701022 Cytomegalovirus Species 0.000 description 1
- IGXWBGJHJZYPQS-SSDOTTSWSA-N D-Luciferin Chemical compound OC(=O)[C@H]1CSC(C=2SC3=CC=C(O)C=C3N=2)=N1 IGXWBGJHJZYPQS-SSDOTTSWSA-N 0.000 description 1
- 101150074155 DHFR gene Proteins 0.000 description 1
- 102000053602 DNA Human genes 0.000 description 1
- 102000004594 DNA Polymerase I Human genes 0.000 description 1
- 108010017826 DNA Polymerase I Proteins 0.000 description 1
- 102000004163 DNA-directed RNA polymerases Human genes 0.000 description 1
- 108090000626 DNA-directed RNA polymerases Proteins 0.000 description 1
- CYCGRDQQIOGCKX-UHFFFAOYSA-N Dehydro-luciferin Natural products OC(=O)C1=CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 CYCGRDQQIOGCKX-UHFFFAOYSA-N 0.000 description 1
- BWGNESOTFCXPMA-UHFFFAOYSA-N Dihydrogen disulfide Chemical compound SS BWGNESOTFCXPMA-UHFFFAOYSA-N 0.000 description 1
- MYMOFIZGZYHOMD-UHFFFAOYSA-N Dioxygen Chemical compound O=O MYMOFIZGZYHOMD-UHFFFAOYSA-N 0.000 description 1
- 206010058314 Dysplasia Diseases 0.000 description 1
- 208000000059 Dyspnea Diseases 0.000 description 1
- 206010013975 Dyspnoeas Diseases 0.000 description 1
- KCXVZYZYPLLWCC-UHFFFAOYSA-N EDTA Chemical compound OC(=O)CN(CC(O)=O)CCN(CC(O)=O)CC(O)=O KCXVZYZYPLLWCC-UHFFFAOYSA-N 0.000 description 1
- 241000196324 Embryophyta Species 0.000 description 1
- 241000283086 Equidae Species 0.000 description 1
- 241000206602 Eukaryota Species 0.000 description 1
- 241000282324 Felis Species 0.000 description 1
- BJGNCJDXODQBOB-UHFFFAOYSA-N Fivefly Luciferin Natural products OC(=O)C1CSC(C=2SC3=CC(O)=CC=C3N=2)=N1 BJGNCJDXODQBOB-UHFFFAOYSA-N 0.000 description 1
- 208000002325 Funnel Chest Diseases 0.000 description 1
- 230000005526 G1 to G0 transition Effects 0.000 description 1
- 108010010803 Gelatin Proteins 0.000 description 1
- 102000053187 Glucuronidase Human genes 0.000 description 1
- 108010060309 Glucuronidase Proteins 0.000 description 1
- 239000004471 Glycine Substances 0.000 description 1
- 239000012981 Hank's balanced salt solution Substances 0.000 description 1
- 206010019842 Hepatomegaly Diseases 0.000 description 1
- 241000238631 Hexapoda Species 0.000 description 1
- 108010000521 Human Growth Hormone Proteins 0.000 description 1
- 108091006905 Human Serum Albumin Proteins 0.000 description 1
- 102000008100 Human Serum Albumin Human genes 0.000 description 1
- 241000701024 Human betaherpesvirus 5 Species 0.000 description 1
- 229920002153 Hydroxypropyl cellulose Polymers 0.000 description 1
- 206010020751 Hypersensitivity Diseases 0.000 description 1
- 206010021079 Hypopnoea Diseases 0.000 description 1
- 208000001953 Hypotension Diseases 0.000 description 1
- 108010091358 Hypoxanthine Phosphoribosyltransferase Proteins 0.000 description 1
- 102100029098 Hypoxanthine-guanine phosphoribosyltransferase Human genes 0.000 description 1
- 206010021143 Hypoxia Diseases 0.000 description 1
- 108010031792 IGF Type 2 Receptor Proteins 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 108700002232 Immediate-Early Genes Proteins 0.000 description 1
- 208000024781 Immune Complex disease Diseases 0.000 description 1
- 108700001097 Insect Genes Proteins 0.000 description 1
- 102000004877 Insulin Human genes 0.000 description 1
- 108090001061 Insulin Proteins 0.000 description 1
- 206010023204 Joint dislocation Diseases 0.000 description 1
- 108010025815 Kanamycin Kinase Proteins 0.000 description 1
- XUJNEKJLAYXESH-REOHCLBHSA-N L-Cysteine Chemical compound SC[C@H](N)C(O)=O XUJNEKJLAYXESH-REOHCLBHSA-N 0.000 description 1
- ZQISRDCJNBUVMM-UHFFFAOYSA-N L-Histidinol Natural products OCC(N)CC1=CN=CN1 ZQISRDCJNBUVMM-UHFFFAOYSA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical compound OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- ZQISRDCJNBUVMM-YFKPBYRVSA-N L-histidinol Chemical compound OC[C@@H](N)CC1=CNC=N1 ZQISRDCJNBUVMM-YFKPBYRVSA-N 0.000 description 1
- AEMOLEFTQBMNLQ-HNFCZKTMSA-N L-idopyranuronic acid Chemical compound OC1O[C@@H](C(O)=O)[C@@H](O)[C@H](O)[C@H]1O AEMOLEFTQBMNLQ-HNFCZKTMSA-N 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- QIVBCDIJIAJPQS-VIFPVBQESA-N L-tryptophane Chemical compound C1=CC=C2C(C[C@H](N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-VIFPVBQESA-N 0.000 description 1
- WHXSMMKQMYFTQS-UHFFFAOYSA-N Lithium Chemical compound [Li] WHXSMMKQMYFTQS-UHFFFAOYSA-N 0.000 description 1
- DDWFXDSYGUXRAY-UHFFFAOYSA-N Luciferin Natural products CCc1c(C)c(CC2NC(=O)C(=C2C=C)C)[nH]c1Cc3[nH]c4C(=C5/NC(CC(=O)O)C(C)C5CC(=O)O)CC(=O)c4c3C DDWFXDSYGUXRAY-UHFFFAOYSA-N 0.000 description 1
- 101710175625 Maltose/maltodextrin-binding periplasmic protein Proteins 0.000 description 1
- 102000019218 Mannose-6-phosphate receptors Human genes 0.000 description 1
- 102000051089 Melanotransferrin Human genes 0.000 description 1
- 108700038051 Melanotransferrin Proteins 0.000 description 1
- 208000024556 Mendelian disease Diseases 0.000 description 1
- 208000007650 Meningeal Carcinomatosis Diseases 0.000 description 1
- 201000011442 Metachromatic leukodystrophy Diseases 0.000 description 1
- 206010051696 Metastases to meninges Diseases 0.000 description 1
- 101100261636 Methanothermobacter marburgensis (strain ATCC BAA-927 / DSM 2133 / JCM 14651 / NBRC 100331 / OCM 82 / Marburg) trpB2 gene Proteins 0.000 description 1
- 108700005443 Microbial Genes Proteins 0.000 description 1
- 208000002678 Mucopolysaccharidoses Diseases 0.000 description 1
- 208000025797 Mucopolysaccharidosis type 4A Diseases 0.000 description 1
- 208000025915 Mucopolysaccharidosis type 6 Diseases 0.000 description 1
- 241001529936 Murinae Species 0.000 description 1
- 241000699670 Mus sp. Species 0.000 description 1
- 241000204031 Mycoplasma Species 0.000 description 1
- 101710099863 N-acetylgalactosamine-6-sulfatase Proteins 0.000 description 1
- 108010023320 N-acetylglucosamine-6-sulfatase Proteins 0.000 description 1
- UCWMWNHWCYYDTD-UHFFFAOYSA-N N-carboxyglycine Chemical compound OC(=O)CNC(O)=O UCWMWNHWCYYDTD-UHFFFAOYSA-N 0.000 description 1
- 206010028813 Nausea Diseases 0.000 description 1
- 229930193140 Neomycin Natural products 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 239000002033 PVDF binder Substances 0.000 description 1
- 206010034204 Pectus excavatum Diseases 0.000 description 1
- 108010033276 Peptide Fragments Proteins 0.000 description 1
- 102000007079 Peptide Fragments Human genes 0.000 description 1
- 102000006335 Phosphate-Binding Proteins Human genes 0.000 description 1
- 108010058514 Phosphate-Binding Proteins Proteins 0.000 description 1
- OAICVXFJPJFONN-UHFFFAOYSA-N Phosphorus Chemical compound [P] OAICVXFJPJFONN-UHFFFAOYSA-N 0.000 description 1
- 101100124346 Photorhabdus laumondii subsp. laumondii (strain DSM 15139 / CIP 105565 / TT01) hisCD gene Proteins 0.000 description 1
- 239000002202 Polyethylene glycol Substances 0.000 description 1
- 229920001213 Polysorbate 20 Polymers 0.000 description 1
- 102220512239 Prostaglandin E synthase_C89S_mutation Human genes 0.000 description 1
- 208000008425 Protein deficiency Diseases 0.000 description 1
- 206010037660 Pyrexia Diseases 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- 206010067171 Regurgitation Diseases 0.000 description 1
- 241000714474 Rous sarcoma virus Species 0.000 description 1
- 208000025816 Sanfilippo syndrome type A Diseases 0.000 description 1
- 208000032023 Signs and Symptoms Diseases 0.000 description 1
- 241000700584 Simplexvirus Species 0.000 description 1
- 108020004682 Single-Stranded DNA Proteins 0.000 description 1
- 241000282887 Suidae Species 0.000 description 1
- NINIDFKCEFEMDL-UHFFFAOYSA-N Sulfur Chemical compound [S] NINIDFKCEFEMDL-UHFFFAOYSA-N 0.000 description 1
- 208000001871 Tachycardia Diseases 0.000 description 1
- GUGOEEXESWIERI-UHFFFAOYSA-N Terfenadine Chemical compound C1=CC(C(C)(C)C)=CC=C1C(O)CCCN1CCC(C(O)(C=2C=CC=CC=2)C=2C=CC=CC=2)CC1 GUGOEEXESWIERI-UHFFFAOYSA-N 0.000 description 1
- 101710120037 Toxin CcdB Proteins 0.000 description 1
- 201000001943 Tricuspid Valve Insufficiency Diseases 0.000 description 1
- 206010044640 Tricuspid valve incompetence Diseases 0.000 description 1
- 239000007983 Tris buffer Substances 0.000 description 1
- GLNADSQYFUSGOU-GPTZEZBUSA-J Trypan blue Chemical compound [Na+].[Na+].[Na+].[Na+].C1=C(S([O-])(=O)=O)C=C2C=C(S([O-])(=O)=O)C(/N=N/C3=CC=C(C=C3C)C=3C=C(C(=CC=3)\N=N\C=3C(=CC4=CC(=CC(N)=C4C=3O)S([O-])(=O)=O)S([O-])(=O)=O)C)=C(O)C2=C1N GLNADSQYFUSGOU-GPTZEZBUSA-J 0.000 description 1
- 102000004142 Trypsin Human genes 0.000 description 1
- 108090000631 Trypsin Proteins 0.000 description 1
- QIVBCDIJIAJPQS-UHFFFAOYSA-N Tryptophan Natural products C1=CC=C2C(CC(N)C(O)=O)=CNC2=C1 QIVBCDIJIAJPQS-UHFFFAOYSA-N 0.000 description 1
- 229910021551 Vanadium(III) chloride Inorganic materials 0.000 description 1
- 108700005077 Viral Genes Proteins 0.000 description 1
- 238000002441 X-ray diffraction Methods 0.000 description 1
- 101000909800 Xenopus laevis Probable N-acetyltransferase camello Proteins 0.000 description 1
- 108010084455 Zeocin Proteins 0.000 description 1
- VQRCPOPQRYDVRE-KEWYIRBNSA-N [(3R,4R,5S,6R)-2-acetyl-3-amino-4,5-dihydroxy-6-(hydroxymethyl)oxan-2-yl] dihydrogen phosphate Chemical compound P(=O)(O)(O)OC1([C@H](N)[C@@H](O)[C@H](O)[C@H](O1)CO)C(C)=O VQRCPOPQRYDVRE-KEWYIRBNSA-N 0.000 description 1
- 230000005856 abnormality Effects 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 238000000862 absorption spectrum Methods 0.000 description 1
- 150000007960 acetonitrile Chemical class 0.000 description 1
- 125000002777 acetyl group Chemical group [H]C([H])([H])C(*)=O 0.000 description 1
- 230000021736 acetylation Effects 0.000 description 1
- 238000006640 acetylation reaction Methods 0.000 description 1
- 230000010933 acylation Effects 0.000 description 1
- 238000005917 acylation reaction Methods 0.000 description 1
- 230000006978 adaptation Effects 0.000 description 1
- 230000006838 adverse reaction Effects 0.000 description 1
- 239000000443 aerosol Substances 0.000 description 1
- 238000001042 affinity chromatography Methods 0.000 description 1
- 230000000172 allergic effect Effects 0.000 description 1
- 230000004075 alteration Effects 0.000 description 1
- 150000001408 amides Chemical class 0.000 description 1
- 125000000539 amino acid group Chemical group 0.000 description 1
- 125000003277 amino group Chemical group 0.000 description 1
- 229940126575 aminoglycoside Drugs 0.000 description 1
- BFNBIHQBYMNNAN-UHFFFAOYSA-N ammonium sulfate Chemical compound N.N.OS(O)(=O)=O BFNBIHQBYMNNAN-UHFFFAOYSA-N 0.000 description 1
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 1
- 239000001166 ammonium sulphate Substances 0.000 description 1
- 235000011130 ammonium sulphate Nutrition 0.000 description 1
- 230000003321 amplification Effects 0.000 description 1
- 238000000137 annealing Methods 0.000 description 1
- 229930002877 anthocyanin Natural products 0.000 description 1
- 235000010208 anthocyanin Nutrition 0.000 description 1
- 239000004410 anthocyanin Substances 0.000 description 1
- 150000004636 anthocyanins Chemical class 0.000 description 1
- 230000003466 anti-cipated effect Effects 0.000 description 1
- 230000001387 anti-histamine Effects 0.000 description 1
- 230000000340 anti-metabolite Effects 0.000 description 1
- 239000000427 antigen Substances 0.000 description 1
- 108091007433 antigens Proteins 0.000 description 1
- 102000036639 antigens Human genes 0.000 description 1
- 229940125715 antihistaminic agent Drugs 0.000 description 1
- 229940100197 antimetabolite Drugs 0.000 description 1
- 239000002256 antimetabolite Substances 0.000 description 1
- 208000008784 apnea Diseases 0.000 description 1
- 239000012736 aqueous medium Substances 0.000 description 1
- 238000003491 array Methods 0.000 description 1
- 208000010668 atopic eczema Diseases 0.000 description 1
- 210000002199 attachment cell Anatomy 0.000 description 1
- UNTBPXHCXVWYOI-UHFFFAOYSA-O azanium;oxido(dioxo)vanadium Chemical compound [NH4+].[O-][V](=O)=O UNTBPXHCXVWYOI-UHFFFAOYSA-O 0.000 description 1
- 239000013602 bacteriophage vector Substances 0.000 description 1
- 230000006399 behavior Effects 0.000 description 1
- 230000009286 beneficial effect Effects 0.000 description 1
- 230000003115 biocidal effect Effects 0.000 description 1
- 230000004071 biological effect Effects 0.000 description 1
- 229960000074 biopharmaceutical Drugs 0.000 description 1
- 229920001222 biopolymer Polymers 0.000 description 1
- 230000001851 biosynthetic effect Effects 0.000 description 1
- 229960002685 biotin Drugs 0.000 description 1
- 235000020958 biotin Nutrition 0.000 description 1
- 239000011616 biotin Substances 0.000 description 1
- 230000017531 blood circulation Effects 0.000 description 1
- 230000036765 blood level Effects 0.000 description 1
- 210000001218 blood-brain barrier Anatomy 0.000 description 1
- 238000009835 boiling Methods 0.000 description 1
- 238000010322 bone marrow transplantation Methods 0.000 description 1
- 108010006025 bovine growth hormone Proteins 0.000 description 1
- 229940098773 bovine serum albumin Drugs 0.000 description 1
- 210000004556 brain Anatomy 0.000 description 1
- 239000012512 bulk drug substance Substances 0.000 description 1
- 239000001110 calcium chloride Substances 0.000 description 1
- 229910001628 calcium chloride Inorganic materials 0.000 description 1
- 239000003560 cancer drug Substances 0.000 description 1
- 239000004202 carbamide Substances 0.000 description 1
- 239000001569 carbon dioxide Substances 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 230000021523 carboxylation Effects 0.000 description 1
- 238000006473 carboxylation reaction Methods 0.000 description 1
- 230000002802 cardiorespiratory effect Effects 0.000 description 1
- 238000013216 cat model Methods 0.000 description 1
- 230000003197 catalytic effect Effects 0.000 description 1
- 239000013592 cell lysate Substances 0.000 description 1
- 239000006285 cell suspension Substances 0.000 description 1
- 210000001638 cerebellum Anatomy 0.000 description 1
- 230000002490 cerebral effect Effects 0.000 description 1
- 210000004289 cerebral ventricle Anatomy 0.000 description 1
- 239000002738 chelating agent Substances 0.000 description 1
- 239000007795 chemical reaction product Substances 0.000 description 1
- 229960004926 chlorobutanol Drugs 0.000 description 1
- VJYIFXVZLXQVHO-UHFFFAOYSA-N chlorsulfuron Chemical compound COC1=NC(C)=NC(NC(=O)NS(=O)(=O)C=2C(=CC=CC=2)Cl)=N1 VJYIFXVZLXQVHO-UHFFFAOYSA-N 0.000 description 1
- 230000004087 circulation Effects 0.000 description 1
- 210000003703 cisterna magna Anatomy 0.000 description 1
- 238000010367 cloning Methods 0.000 description 1
- 238000012761 co-transfection Methods 0.000 description 1
- 239000011248 coating agent Substances 0.000 description 1
- 238000004440 column chromatography Methods 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 238000002591 computed tomography Methods 0.000 description 1
- 230000001143 conditioned effect Effects 0.000 description 1
- 108091036078 conserved sequence Proteins 0.000 description 1
- 238000010924 continuous production Methods 0.000 description 1
- 238000007796 conventional method Methods 0.000 description 1
- 239000002826 coolant Substances 0.000 description 1
- 238000001816 cooling Methods 0.000 description 1
- 229910000365 copper sulfate Inorganic materials 0.000 description 1
- ARUVKPQLZAKDPS-UHFFFAOYSA-L copper(II) sulfate Chemical compound [Cu+2].[O-][S+2]([O-])([O-])[O-] ARUVKPQLZAKDPS-UHFFFAOYSA-L 0.000 description 1
- 229940109239 creatinine Drugs 0.000 description 1
- 239000013078 crystal Substances 0.000 description 1
- 239000012531 culture fluid Substances 0.000 description 1
- WZHCOOQXZCIUNC-UHFFFAOYSA-N cyclandelate Chemical compound C1C(C)(C)CC(C)CC1OC(=O)C(O)C1=CC=CC=C1 WZHCOOQXZCIUNC-UHFFFAOYSA-N 0.000 description 1
- AIMMVWOEOZMVMS-UHFFFAOYSA-N cyclopropanecarboxamide Chemical compound NC(=O)C1CC1 AIMMVWOEOZMVMS-UHFFFAOYSA-N 0.000 description 1
- 210000000805 cytoplasm Anatomy 0.000 description 1
- 230000000254 damaging effect Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 238000006356 dehydrogenation reaction Methods 0.000 description 1
- 230000002939 deleterious effect Effects 0.000 description 1
- 238000012217 deletion Methods 0.000 description 1
- 230000037430 deletion Effects 0.000 description 1
- 230000005595 deprotonation Effects 0.000 description 1
- 238000010537 deprotonation reaction Methods 0.000 description 1
- 230000001066 destructive effect Effects 0.000 description 1
- 238000000502 dialysis Methods 0.000 description 1
- 239000010432 diamond Substances 0.000 description 1
- 229940061607 dibasic sodium phosphate Drugs 0.000 description 1
- 235000005911 diet Nutrition 0.000 description 1
- 230000037213 diet Effects 0.000 description 1
- 238000009792 diffusion process Methods 0.000 description 1
- 230000029087 digestion Effects 0.000 description 1
- UGMCXQCYOVCMTB-UHFFFAOYSA-K dihydroxy(stearato)aluminium Chemical compound CCCCCCCCCCCCCCCCCC(=O)O[Al](O)O UGMCXQCYOVCMTB-UHFFFAOYSA-K 0.000 description 1
- 230000003292 diminished effect Effects 0.000 description 1
- 229910001882 dioxygen Inorganic materials 0.000 description 1
- BNIILDVGGAEEIG-UHFFFAOYSA-L disodium hydrogen phosphate Chemical compound [Na+].[Na+].OP([O-])([O-])=O BNIILDVGGAEEIG-UHFFFAOYSA-L 0.000 description 1
- 238000011833 dog model Methods 0.000 description 1
- 231100000673 dose–response relationship Toxicity 0.000 description 1
- 229940000406 drug candidate Drugs 0.000 description 1
- 239000003480 eluent Substances 0.000 description 1
- 210000002472 endoplasmic reticulum Anatomy 0.000 description 1
- 230000003511 endothelial effect Effects 0.000 description 1
- 230000002708 enhancing effect Effects 0.000 description 1
- BEFDCLMNVWHSGT-UHFFFAOYSA-N ethenylcyclopentane Chemical compound C=CC1CCCC1 BEFDCLMNVWHSGT-UHFFFAOYSA-N 0.000 description 1
- 230000007717 exclusion Effects 0.000 description 1
- 238000002270 exclusion chromatography Methods 0.000 description 1
- 230000029142 excretion Effects 0.000 description 1
- 230000001815 facial effect Effects 0.000 description 1
- 238000011049 filling Methods 0.000 description 1
- 239000006260 foam Substances 0.000 description 1
- 235000019253 formic acid Nutrition 0.000 description 1
- 239000012458 free base Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 239000012737 fresh medium Substances 0.000 description 1
- 230000004927 fusion Effects 0.000 description 1
- 230000005021 gait Effects 0.000 description 1
- 229930182830 galactose Natural products 0.000 description 1
- 239000007789 gas Substances 0.000 description 1
- 238000001502 gel electrophoresis Methods 0.000 description 1
- 238000001641 gel filtration chromatography Methods 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
- 239000011491 glass wool Substances 0.000 description 1
- 102000006602 glyceraldehyde-3-phosphate dehydrogenase Human genes 0.000 description 1
- 108020004445 glyceraldehyde-3-phosphate dehydrogenase Proteins 0.000 description 1
- 230000013595 glycosylation Effects 0.000 description 1
- 238000006206 glycosylation reaction Methods 0.000 description 1
- 239000008187 granular material Substances 0.000 description 1
- 239000012561 harvest cell culture fluid Substances 0.000 description 1
- 210000003128 head Anatomy 0.000 description 1
- 230000004217 heart function Effects 0.000 description 1
- 238000003505 heat denaturation Methods 0.000 description 1
- 108010089932 heparan sulfate sulfatase Proteins 0.000 description 1
- 208000010726 hind limb paralysis Diseases 0.000 description 1
- 101150113423 hisD gene Proteins 0.000 description 1
- 239000012510 hollow fiber Substances 0.000 description 1
- 238000009396 hybridization Methods 0.000 description 1
- 150000002429 hydrazines Chemical class 0.000 description 1
- 235000011167 hydrochloric acid Nutrition 0.000 description 1
- 239000000017 hydrogel Substances 0.000 description 1
- 230000002209 hydrophobic effect Effects 0.000 description 1
- 150000004679 hydroxides Chemical class 0.000 description 1
- 150000002443 hydroxylamines Chemical class 0.000 description 1
- 229910052588 hydroxylapatite Inorganic materials 0.000 description 1
- 238000012872 hydroxylapatite chromatography Methods 0.000 description 1
- 125000004029 hydroxymethyl group Chemical group [H]OC([H])([H])* 0.000 description 1
- 239000001863 hydroxypropyl cellulose Substances 0.000 description 1
- 235000010977 hydroxypropyl cellulose Nutrition 0.000 description 1
- 208000024326 hypersensitivity reaction type III disease Diseases 0.000 description 1
- 230000036543 hypotension Effects 0.000 description 1
- 208000018875 hypoxemia Diseases 0.000 description 1
- 230000001900 immune effect Effects 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 239000012535 impurity Substances 0.000 description 1
- PZOUSPYUWWUPPK-UHFFFAOYSA-N indole Natural products CC1=CC=CC2=C1C=CN2 PZOUSPYUWWUPPK-UHFFFAOYSA-N 0.000 description 1
- RKJUIXBNRJVNHR-UHFFFAOYSA-N indolenine Natural products C1=CC=C2CC=NC2=C1 RKJUIXBNRJVNHR-UHFFFAOYSA-N 0.000 description 1
- 208000015181 infectious disease Diseases 0.000 description 1
- 230000036512 infertility Effects 0.000 description 1
- 239000007972 injectable composition Substances 0.000 description 1
- 239000002054 inoculum Substances 0.000 description 1
- 150000007529 inorganic bases Chemical class 0.000 description 1
- 238000003780 insertion Methods 0.000 description 1
- 230000037431 insertion Effects 0.000 description 1
- 229940125396 insulin Drugs 0.000 description 1
- 239000000543 intermediate Substances 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 150000002500 ions Chemical class 0.000 description 1
- JJWLVOIRVHMVIS-UHFFFAOYSA-N isopropylamine Chemical compound CC(C)N JJWLVOIRVHMVIS-UHFFFAOYSA-N 0.000 description 1
- 125000000468 ketone group Chemical group 0.000 description 1
- 230000003907 kidney function Effects 0.000 description 1
- 210000003127 knee Anatomy 0.000 description 1
- 238000009533 lab test Methods 0.000 description 1
- 238000012007 large scale cell culture Methods 0.000 description 1
- 238000011031 large-scale manufacturing process Methods 0.000 description 1
- 239000000787 lecithin Substances 0.000 description 1
- 235000010445 lecithin Nutrition 0.000 description 1
- 229940067606 lecithin Drugs 0.000 description 1
- 230000029226 lipidation Effects 0.000 description 1
- 238000004811 liquid chromatography Methods 0.000 description 1
- 239000006193 liquid solution Substances 0.000 description 1
- 229910052744 lithium Inorganic materials 0.000 description 1
- 230000003908 liver function Effects 0.000 description 1
- 230000007774 longterm Effects 0.000 description 1
- 210000003141 lower extremity Anatomy 0.000 description 1
- 238000009593 lumbar puncture Methods 0.000 description 1
- 210000004705 lumbosacral region Anatomy 0.000 description 1
- 210000004072 lung Anatomy 0.000 description 1
- 210000001165 lymph node Anatomy 0.000 description 1
- 238000002595 magnetic resonance imaging Methods 0.000 description 1
- 238000012423 maintenance Methods 0.000 description 1
- 206010025482 malaise Diseases 0.000 description 1
- 150000002704 mannoses Chemical class 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 230000010534 mechanism of action Effects 0.000 description 1
- 230000001404 mediated effect Effects 0.000 description 1
- 238000002844 melting Methods 0.000 description 1
- 230000008018 melting Effects 0.000 description 1
- 230000002503 metabolic effect Effects 0.000 description 1
- 230000004060 metabolic process Effects 0.000 description 1
- 239000002207 metabolite Substances 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- HPNSFSBZBAHARI-UHFFFAOYSA-N micophenolic acid Natural products OC1=C(CC=C(C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-UHFFFAOYSA-N 0.000 description 1
- 230000000813 microbial effect Effects 0.000 description 1
- 235000013336 milk Nutrition 0.000 description 1
- 239000008267 milk Substances 0.000 description 1
- 210000004080 milk Anatomy 0.000 description 1
- 230000003278 mimic effect Effects 0.000 description 1
- 150000007522 mineralic acids Chemical class 0.000 description 1
- 238000006011 modification reaction Methods 0.000 description 1
- 229940045641 monobasic sodium phosphate Drugs 0.000 description 1
- 230000000877 morphologic effect Effects 0.000 description 1
- 238000013425 morphometry Methods 0.000 description 1
- 206010028093 mucopolysaccharidosis Diseases 0.000 description 1
- 208000000690 mucopolysaccharidosis VI Diseases 0.000 description 1
- 208000012226 mucopolysaccharidosis type IIIA Diseases 0.000 description 1
- 230000021332 multicellular organism growth Effects 0.000 description 1
- 231100000219 mutagenic Toxicity 0.000 description 1
- 230000003505 mutagenic effect Effects 0.000 description 1
- 229960000951 mycophenolic acid Drugs 0.000 description 1
- HPNSFSBZBAHARI-RUDMXATFSA-N mycophenolic acid Chemical compound OC1=C(C\C=C(/C)CCC(O)=O)C(OC)=C(C)C2=C1C(=O)OC2 HPNSFSBZBAHARI-RUDMXATFSA-N 0.000 description 1
- 230000008693 nausea Effects 0.000 description 1
- 229960004927 neomycin Drugs 0.000 description 1
- 230000007935 neutral effect Effects 0.000 description 1
- 239000002547 new drug Substances 0.000 description 1
- 231100000252 nontoxic Toxicity 0.000 description 1
- 230000003000 nontoxic effect Effects 0.000 description 1
- 239000004745 nonwoven fabric Substances 0.000 description 1
- 238000010899 nucleation Methods 0.000 description 1
- 238000003199 nucleic acid amplification method Methods 0.000 description 1
- 230000000414 obstructive effect Effects 0.000 description 1
- QIQXTHQIDYTFRH-UHFFFAOYSA-N octadecanoic acid Chemical compound CCCCCCCCCCCCCCCCCC(O)=O QIQXTHQIDYTFRH-UHFFFAOYSA-N 0.000 description 1
- 239000003921 oil Substances 0.000 description 1
- 235000019198 oils Nutrition 0.000 description 1
- 150000007524 organic acids Chemical class 0.000 description 1
- 235000005985 organic acids Nutrition 0.000 description 1
- 150000007530 organic bases Chemical class 0.000 description 1
- 210000001672 ovary Anatomy 0.000 description 1
- 239000007800 oxidant agent Substances 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- XYJRXVWERLGGKC-UHFFFAOYSA-D pentacalcium;hydroxide;triphosphate Chemical compound [OH-].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[Ca+2].[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O.[O-]P([O-])([O-])=O XYJRXVWERLGGKC-UHFFFAOYSA-D 0.000 description 1
- 230000000737 periodic effect Effects 0.000 description 1
- 230000002093 peripheral effect Effects 0.000 description 1
- 229960003742 phenol Drugs 0.000 description 1
- CWCMIVBLVUHDHK-ZSNHEYEWSA-N phleomycin D1 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC[C@@H](N=1)C=1SC=C(N=1)C(=O)NCCCCNC(N)=N)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C CWCMIVBLVUHDHK-ZSNHEYEWSA-N 0.000 description 1
- NBIIXXVUZAFLBC-UHFFFAOYSA-K phosphate Chemical compound [O-]P([O-])([O-])=O NBIIXXVUZAFLBC-UHFFFAOYSA-K 0.000 description 1
- 239000010452 phosphate Substances 0.000 description 1
- 108010079892 phosphoglycerol kinase Proteins 0.000 description 1
- 235000011007 phosphoric acid Nutrition 0.000 description 1
- 150000003016 phosphoric acids Chemical class 0.000 description 1
- 229910052698 phosphorus Inorganic materials 0.000 description 1
- 239000011574 phosphorus Substances 0.000 description 1
- 230000026731 phosphorylation Effects 0.000 description 1
- 238000006366 phosphorylation reaction Methods 0.000 description 1
- 230000002186 photoactivation Effects 0.000 description 1
- 239000004033 plastic Substances 0.000 description 1
- 229920003023 plastic Polymers 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 230000000379 polymerizing effect Effects 0.000 description 1
- 229920005862 polyol Polymers 0.000 description 1
- 150000003077 polyols Chemical class 0.000 description 1
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 description 1
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 description 1
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 1
- BQFYGYJPBUKISI-UHFFFAOYSA-N potassium;oxido(dioxo)vanadium Chemical compound [K+].[O-][V](=O)=O BQFYGYJPBUKISI-UHFFFAOYSA-N 0.000 description 1
- 244000144977 poultry Species 0.000 description 1
- 235000013594 poultry meat Nutrition 0.000 description 1
- 238000001556 precipitation Methods 0.000 description 1
- 239000002243 precursor Substances 0.000 description 1
- 238000009597 pregnancy test Methods 0.000 description 1
- 239000003755 preservative agent Substances 0.000 description 1
- 230000002335 preservative effect Effects 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- MFDFERRIHVXMIY-UHFFFAOYSA-N procaine Chemical compound CCN(CC)CCOC(=O)C1=CC=C(N)C=C1 MFDFERRIHVXMIY-UHFFFAOYSA-N 0.000 description 1
- 229960004919 procaine Drugs 0.000 description 1
- 238000003672 processing method Methods 0.000 description 1
- 230000002035 prolonged effect Effects 0.000 description 1
- 125000001500 prolyl group Chemical group [H]N1C([H])(C(=O)[*])C([H])([H])C([H])([H])C1([H])[H] 0.000 description 1
- 230000007065 protein hydrolysis Effects 0.000 description 1
- 238000001742 protein purification Methods 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- 238000009613 pulmonary function test Methods 0.000 description 1
- 239000013014 purified material Substances 0.000 description 1
- 229950010131 puromycin Drugs 0.000 description 1
- 239000002510 pyrogen Substances 0.000 description 1
- 238000000275 quality assurance Methods 0.000 description 1
- 238000011002 quantification Methods 0.000 description 1
- 239000010453 quartz Substances 0.000 description 1
- 230000005855 radiation Effects 0.000 description 1
- 230000002285 radioactive effect Effects 0.000 description 1
- 238000003259 recombinant expression Methods 0.000 description 1
- 231100000272 reduced body weight Toxicity 0.000 description 1
- 239000012557 regeneration buffer Substances 0.000 description 1
- 230000010076 replication Effects 0.000 description 1
- 230000029058 respiratory gaseous exchange Effects 0.000 description 1
- 230000004043 responsiveness Effects 0.000 description 1
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 1
- 230000002441 reversible effect Effects 0.000 description 1
- 102200132744 rs137853222 Human genes 0.000 description 1
- 102200105259 rs587777638 Human genes 0.000 description 1
- 102200083964 rs74315317 Human genes 0.000 description 1
- 239000012266 salt solution Substances 0.000 description 1
- 238000005070 sampling Methods 0.000 description 1
- 238000003118 sandwich ELISA Methods 0.000 description 1
- 229920006395 saturated elastomer Polymers 0.000 description 1
- 210000004761 scalp Anatomy 0.000 description 1
- 239000006152 selective media Substances 0.000 description 1
- 238000012163 sequencing technique Methods 0.000 description 1
- 208000013220 shortness of breath Diseases 0.000 description 1
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicon dioxide Inorganic materials O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 1
- 210000003625 skull Anatomy 0.000 description 1
- 201000002859 sleep apnea Diseases 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000007974 sodium acetate buffer Substances 0.000 description 1
- 239000000661 sodium alginate Substances 0.000 description 1
- 235000010413 sodium alginate Nutrition 0.000 description 1
- 229940005550 sodium alginate Drugs 0.000 description 1
- 229910000029 sodium carbonate Inorganic materials 0.000 description 1
- AJPJDKMHJJGVTQ-UHFFFAOYSA-M sodium dihydrogen phosphate Chemical compound [Na+].OP(O)([O-])=O AJPJDKMHJJGVTQ-UHFFFAOYSA-M 0.000 description 1
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 description 1
- UPDATVKGFTVGQJ-UHFFFAOYSA-N sodium;azane Chemical compound N.[Na+] UPDATVKGFTVGQJ-UHFFFAOYSA-N 0.000 description 1
- 238000010532 solid phase synthesis reaction Methods 0.000 description 1
- 239000004334 sorbic acid Substances 0.000 description 1
- 235000010199 sorbic acid Nutrition 0.000 description 1
- 229940075582 sorbic acid Drugs 0.000 description 1
- 241000894007 species Species 0.000 description 1
- 238000002798 spectrophotometry method Methods 0.000 description 1
- 238000001228 spectrum Methods 0.000 description 1
- 210000000278 spinal cord Anatomy 0.000 description 1
- 230000010473 stable expression Effects 0.000 description 1
- 238000004659 sterilization and disinfection Methods 0.000 description 1
- 150000005845 steroid sulfates Chemical class 0.000 description 1
- 239000000126 substance Substances 0.000 description 1
- 235000000346 sugar Nutrition 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- 125000000020 sulfo group Chemical group O=S(=O)([*])O[H] 0.000 description 1
- 229910052717 sulfur Inorganic materials 0.000 description 1
- 239000011593 sulfur Substances 0.000 description 1
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 1
- 238000004114 suspension culture Methods 0.000 description 1
- 230000006794 tachycardia Effects 0.000 description 1
- 239000008399 tap water Substances 0.000 description 1
- 235000020679 tap water Nutrition 0.000 description 1
- 230000002123 temporal effect Effects 0.000 description 1
- 238000010257 thawing Methods 0.000 description 1
- 238000011285 therapeutic regimen Methods 0.000 description 1
- 230000004797 therapeutic response Effects 0.000 description 1
- RTKIYNMVFMVABJ-UHFFFAOYSA-L thimerosal Chemical compound [Na+].CC[Hg]SC1=CC=CC=C1C([O-])=O RTKIYNMVFMVABJ-UHFFFAOYSA-L 0.000 description 1
- 229940033663 thimerosal Drugs 0.000 description 1
- 238000004809 thin layer chromatography Methods 0.000 description 1
- HNKJADCVZUBCPG-UHFFFAOYSA-N thioanisole Chemical compound CSC1=CC=CC=C1 HNKJADCVZUBCPG-UHFFFAOYSA-N 0.000 description 1
- 238000012090 tissue culture technique Methods 0.000 description 1
- 238000004448 titration Methods 0.000 description 1
- 230000037317 transdermal delivery Effects 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 230000009261 transgenic effect Effects 0.000 description 1
- 230000001052 transient effect Effects 0.000 description 1
- 238000013519 translation Methods 0.000 description 1
- 230000005945 translocation Effects 0.000 description 1
- 230000032258 transport Effects 0.000 description 1
- WUUHFRRPHJEEKV-UHFFFAOYSA-N tripotassium borate Chemical compound [K+].[K+].[K+].[O-]B([O-])[O-] WUUHFRRPHJEEKV-UHFFFAOYSA-N 0.000 description 1
- LENZDBCJOHFCAS-UHFFFAOYSA-N tris Chemical compound OCC(N)(CO)CO LENZDBCJOHFCAS-UHFFFAOYSA-N 0.000 description 1
- 101150081616 trpB gene Proteins 0.000 description 1
- 101150111232 trpB-1 gene Proteins 0.000 description 1
- 239000012588 trypsin Substances 0.000 description 1
- 208000025883 type III hypersensitivity disease Diseases 0.000 description 1
- 238000013060 ultrafiltration and diafiltration Methods 0.000 description 1
- 108020005087 unfolded proteins Proteins 0.000 description 1
- 241000701447 unidentified baculovirus Species 0.000 description 1
- 241001515965 unidentified phage Species 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 238000004148 unit process Methods 0.000 description 1
- 150000003672 ureas Chemical class 0.000 description 1
- 238000002562 urinalysis Methods 0.000 description 1
- 238000001291 vacuum drying Methods 0.000 description 1
- 230000009677 vaginal delivery Effects 0.000 description 1
- IBYSTTGVDIFUAY-UHFFFAOYSA-N vanadium monoxide Chemical compound [V]=O IBYSTTGVDIFUAY-UHFFFAOYSA-N 0.000 description 1
- HQYCOEXWFMFWLR-UHFFFAOYSA-K vanadium(iii) chloride Chemical compound [Cl-].[Cl-].[Cl-].[V+3] HQYCOEXWFMFWLR-UHFFFAOYSA-K 0.000 description 1
- UUUGYDOQQLOJQA-UHFFFAOYSA-L vanadyl sulfate Chemical compound [V+2]=O.[O-]S([O-])(=O)=O UUUGYDOQQLOJQA-UHFFFAOYSA-L 0.000 description 1
- 235000015112 vegetable and seed oil Nutrition 0.000 description 1
- 239000008158 vegetable oil Substances 0.000 description 1
- 210000003462 vein Anatomy 0.000 description 1
- 230000002861 ventricular Effects 0.000 description 1
- 210000003501 vero cell Anatomy 0.000 description 1
- 229960004854 viral vaccine Drugs 0.000 description 1
- 230000004304 visual acuity Effects 0.000 description 1
- 230000000007 visual effect Effects 0.000 description 1
- 239000002699 waste material Substances 0.000 description 1
- 239000003643 water by type Substances 0.000 description 1
Classifications
-
- 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
- C12N9/00—Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
- C12N9/14—Hydrolases (3)
- C12N9/16—Hydrolases (3) acting on ester bonds (3.1)
Definitions
- the present invention is generally directed to methods and compositions for producing activated enzymes for enzyme replacement therapy in lysosomal storage disease.
- Enzyme replacement therapy relies on recombinant protein production for the preparation of therapeutically effective protein compositions for the treatment of a variety of disorders.
- Enzyme replacement therapy has been employed for the treatment of such disorders (Kakkis et al., N Engl J Med 344, 182-8, 2001).
- a key problem in the manufacture of lysosomal sulfatases is low yield.
- the Chinese hamster ovary cell line CHO-Kl is a common cell line used in biopharmaceutical manufacturing and it previously has been employed to establish clones expressing a number of the sulfatases.
- DHFR amplification cell lines that produce a high yield of the recombinant protein have been particularly elusive and have not been obtained (Bielicki et al., Biochem J 311 (Pt 1), 333-9, 1995; Bielicki et al., Biochem J 289 (Pt 1), 241-6, 1993; Bielicki et al., Biochem J 329 (Pt 1), 145-50, 1998; Litjens et al., Biochem J 327 (Pt 1), 89-94, 1997).
- arylsulfatase B (ASB) from recombinant retrovirus in deficient human fibroblasts has been reported to occur only at the expense of endogenous sulfatase expression levels (Anson et al., Biochem J 294 (Pt 3), 657-62, 1993).
- ASA arylsulfatase A
- Overexpression of arylsulfatase A (ASA) also causes measurable decreases in endogenous sulfatases (Ohashi et al., Gene Ther 2, 363-8, 1995). Therefore, recombinant production of enzymes is hindered by endogenous limitation on the total sulfatase output of the mammalian cell line being used for such production.
- Cysteine conversion to formylglycine proceeds by sequential oxidation and hydrolysis reactions.
- the oxidation step is enzyme catalyzed, and hence is saturable (Schmidt et al., Cell 82, 271-8, 1995).
- the hydrolysis step releases H 2 S, which is toxic.
- the cell normally expresses sulfatase enzymes at relatively low levels, during recombinant enzyme production where large quantities of the enzyme are produced, the conversion step is overwhelmed by large amounts of recombinant enzyme. Saturation of the sulfatase activation process also is thought to have detrimental effects on enzyme yield and on the health/viability of the overexpressing line.
- the degree of misfolding likely depends on the particular sulfatase in question.
- I2S iduronate 2-sulfatase
- GALNS N- acetylgalactosamine 6-sulfatase
- co-expression of SUMFl with sulfatase enzymes promises to remove the constraint that low sulfatase yield has had on efforts to treat sulfatase deficiencies by ERT (Baenziger, Cell 113:421-422, 2003).
- co-expression of sulfatase processing enzymes with the sulfatases may have other attendant problems relating to toxicity associated with the conversion of cysteine to formylglycine.
- limits on the capacity of the sulfatase modification machinery may be an adaptation to H 2 S toxicity.
- H 2 S toxicity Even at a modest sulfatase expression rate of lpg/cell/day, the cell will be burdened with a build-up of H 2 S that would reach intracellular concentrations of at least 30 ⁇ M over a 24-hour interval.
- 1 pg of sulfatase is equivalent to 3.0 x 10 "17 moles of sulfatase expressed per cell per day. The same number of moles of H 2 S will be generated if all of the sulfatase is activated.
- the present application is based in part on the discovery that serine mutants of sulfatases that comprise a serine residue instead of a cysteine residue at the active site can be activated using a vanadate-dependent photoreaction which converts the active site serine of these mutant sulfatases to a formylglycine residue thereby rendering the mutants catalytically active.
- the photoreaction involves conversion of the active site serine to a formylglycine.
- particular embodiments of the present invention are directed to methods of converting recombinant sulfatase protein to a catalytically active sulfatase enzyme by contacting a preparation that comprises the protein with a photoactivatable redox agent, such as for example, vanadate and irradiating the preparation with light in the range of between 300 nm and 400 nm wavelength.
- the sulfatase may be any sulfatase that comprises a serine residue instead of an active site cysteine residue.
- the sulfatase may be a eukaryotic sulfatase protein, or a prokaryotic sulfatase protein.
- the protein is a mammalian sulfatase, and more particularly, it is a human sulfatase protein.
- the recombinant sulfatase protein is a mutant mammalian arylsulfatase protein in which the active site cysteine residue has been replaced by a serine residue. More particularly, the active-site serine in the mutant mammalian arylsulfatase is converted to formylglycine by incubating the mammalian arylsulfatase with vanadate in the presence of an energy source of 300 nm to 400 nm wavelength or a laser energy source.
- the preparation is contacted with the redox agent prior to the irradiating step, however, the preparation may be irradiated prior to contacting with the vanadate as long as the contacting with the vanadate ions is sufficient to produce an activation of the sulfatase enzyme.
- the preparation is subjected to a cyclic irradiation wherein the sample is subjected to a period of irradiation followed by a period of incubation in a dark environment, and preferably the cyclic irradiation is repeated, 2, 3, 4, 5, or more times to achieve an optimal photoactivation of the sulfatase.
- the time for the period of irradiation and the time period for incubation in the dark is not limiting and may be selected from any convenient time that is sufficient to activate at least 10% of the mutant sulfatase as compared to the absence of the activation by irradiation and vanadate.
- An exemplary time for irradiating is a 30-minute period of irradiation ("light period of irradiation cycle") followed by a 20 minute period of incubation in the dark ("dark period of irradiation cycle").
- the light period of the irradiation cycle may be 10 minutes, 15 minutes, 20 minutes, 30 minutes, 40 minutes, 50 minutes, 60 minutes, 90 minutes, 120 minutes or longer.
- the dark period of the irradiation cycle may be 5 minutes, 10 minutes, 15 minutes, 20 minutes 30 minutes, 40 minutes, 50 minutes, 60 minutes, 90 minutes, 120 minutes or longer. Any combination of light and dark period may be used
- the sulfatase may be any sulfatase that comprises an active site serine residue instead of an active site cysteine residue. It is particularly contemplated that the arylsulfatase is selected from the group consisting of arylsulfatase A, arylsulfatase B, Arylsulfatase C, arylsulfatase D, arylsulfatase E, arylsulfatase F, arylsulfatase G, Sulfatase 1, Sulfatase 2, galactosamine (N-acetyl)-6-sulfate sulfatase, iduronate 2 sulfatase, N-sulfoglucosamine sulfohydrolase, glucosamine (N-acetyl)-6-sulfatase and arylsulfatase A cerebroside.
- the arylsulfatase is an arylsulfatase B comprising a C ⁇ S mutation at a position corresponding to amino acid position number 91 of SEQ ID NO:4.
- the arylsulfatase is an arylsulfatase A comprising a C ⁇ S mutation at a position corresponding to amino acid position number 91 of SEQ ID NO.2.
- the sulfatase is a protein sequence comprising an amino acid sequence selected from the group consisting of SEQ E) NO:27, SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO.37, SEQ ID NO:38, and SEQ ID NO:39 or variants thereof that retain sulfatase activity.
- the method uses an arylsulfatase protein that is a mutant human arylsulfatase B protein obtained from a CHO cell line according to a method comprising the steps of transfecting CHO cells with an expression construct comprising a nucleic acid encoding an arylsulfatase an arylsulfatase B comprising a C ⁇ S mutation at a position corresponding to amino acid position number 91 of SEQ ID NO:4; selecting cells transfected with the vector; culturing the selected cells under conditions to permit the secretion of the mutant human arylsulfatase B protein (ASB) protein into the culture medium of the cells; and isolating mutant human ASB protein from the culture medium.
- the CHO cell line is a CHO-Kl cell line.
- the CHO cell line is a CHO-S cell line.
- the method uses an arylsulfatase protein that is a mutant human arylsulfatase A protein obtained from a CHO cell line according to a method comprising transfecting CHO cells with an expression construct comprising a nucleic acid encoding an arylsulfatase A (ASA) comprising a C ⁇ S mutation at a position corresponding to amino acid position number 91 of SEQ ID NO:2; selecting cells transfected with the vector; culturing the selected cells under conditions to permit the secretion of the mutant ASA protein into the culture medium of the cells; and isolating the mutant ASA protein from the culture medium.
- the CHO cell line is a CHO-Kl cell line.
- the CHO cell line is a CHO-S cell line.
- Recombinant CHO-S cell lines that are transfected with expression constructs that encode mutant ASA and ASB proteins form another aspect of the invention.
- Also contemplated herein are methods of preparing a pharmaceutically acceptable composition comprising a catalytically active arylsulfatase comprising contacting a preparation comprising recombinant arylsulfatase protein with a photoactivatable redox agent composition, irradiating the preparation with light in the range of between 300 nm and 400 nm wavelength, and formulating the preparation in a pharmaceutically acceptable carrier, diluent or excipient.
- Additional aspects of the invention describe methods of preparing a pharmaceutically acceptable composition comprising a catalytically active arylsulfatase comprising contacting a preparation comprising recombinant arylsulfatase protein with a vanadate composition, irradiating the preparation with light in the range of between 300 nm and 400 nm wavelength, and formulating the preparation in a pharmaceutically acceptable carrier, diluent or excipient.
- compositions comprising catalytically active arylsulfatase proteins prepared according to the method discussed above and described in further detail herein below, and a pharmaceutically acceptable carrier, diluent or excipient.
- Figure 2A Specific productivity of stably transfected pools of cells expressing either ASB or ASB C91 S at 450 ⁇ g/mL G418 and 900 ⁇ g/mL G418.
- Cells were grown in duplicate T-25 flasks. Medium was conditioned for 48 hours prior to harvest. Levels of secreted sulfatase in the conditioned medium were assayed by ELISA. Cell numbers were measured with a Coulter counter.
- FIG. 2B Western blots of secreted and intracellular protein preparations from pools of stably transfected CHO cells expressing ASA or ASA
- Figure 2C shows normalized NPT and sulfatase transcript ratios obtained from pools of stably transfected cells selected at either 450 ⁇ g/mL G418 or 900 ⁇ g/mL G418 and expressing different sulfatase proteins.
- RNA was isolated and transcribed as described in Example 1. Sulfatase and NPT transcript levels were normalized to the ribosomal S14 internal standard.
- Figure 2D shows the ratio of secreted protein per transcript for pools of stably transfected cells expressing either ASB or ASB C91S. Protein levels were measured by ELISA and normalized to cell number measured with a Coulter counter; transcript levels were measured by RT-QPCR and normalized to the ribosomal S 14 internal standard.
- Figure 3 A shows the dependence of the photo-conversion of ASB C91S on vanadate concentration and time-of-irradiation.
- ASB C91S 10 ⁇ L, 0.5 mg/mL
- 50 mM NaOAc pH 6.5 100 mM NaCl
- Vanadate from a 100 mM stock in water at pH 10 was added to each well in order to achieve final concentrations of 10, 40 and 400 ⁇ M. maintained at 20°C as described in Example 1.
- the plate was then irradiated for 120 minutes as described. Sample aliquots (2 ⁇ L) were removed at 60, 90 and 120 minutes and assayed as described.
- Figure 3B shows photo-conversion of ASB C91S concomitant with photo-inactivation of native ASB by vanadate and light.
- ASB C91S 0.5 mg/mL
- native ASB 0.3 mg/mL
- Protein samples were irradiated in 1.5 mL polypropylene microcentrifuge tubes at 20°C as described in the Example 1.
- a 2 mM sodium metavanadate stock in 20 mM Tris pH 9.6 was diluted into the protein solutions to a final concentration of 8 ⁇ M (2 uL to X).
- UVA intensity measurements were made at 1, 15, 25 and 40 minutes during the course of the experiment with a probe fixed at the same distance from the source as the samples. Irradiation intensity had a mean and standard deviation of 90 ⁇ 5 ⁇ W/cm.
- Figure 3C shows activation of ASBC91S with vanadate and light.
- ASB C91S was converted to active enzyme using the conditions specified in Example 1, including 100 mM Tris-acetate pH 8 and 10-500 molar equivalents of vanadate.
- activity values for ASB C91S obtained using different activation conditions were divided by activity values for the same amount of wild-type ASB obtained using the same conditions, but without irradiation (irradiation of wild-type ASB in the absence of vanadate has no effect on the activity of the enzyme, data not shown).
- Closed circles and dotted line represents activation at 500 ⁇ M vanadate. Closed diamonds and solid line represents activation at 10 ⁇ M vanadate.
- Figure 3D shows the LC/MS profile of peptides derived from ASB and from ASB treated with vanadate and light.
- Mammalian sulfatases are involved in the degradation of sulfated substrate such as mucopolysaccharides, cerebroside sulfates and sulfated steroids.
- sulfated substrate such as mucopolysaccharides, cerebroside sulfates and sulfated steroids.
- Key to the catalytic activity of all of sulfatases is the presence of a C ⁇ -formylglycine residue , which is post-translationally generated by the sequential oxidation and hydrolysis of cysteine in the endoplasmic reticulum.
- the sulfatases are inactive and cause multiple sulfatase deficiency (MSD), a rare, yet fatal lysosomal storage disease.
- MSD multiple sulfatase deficiency
- a serine mutant protein might be expected in some cases to be expressed more efficiently than native enzyme since it will not be prone to misfolding as a result of inappropriate disulfide formation that may result in a related enzyme that contains the native cysteine residues. For the same reason, overexpression of the serine mutant would be less likely to be detrimental to the overexpressing cell because the conversion of the serine to formylglycine would not produce H 2 S.
- serine mutants of ASB and ASA have been shown to be virtually indistinguishable from their respective native counterparts by epitope analysis and X-ray diffraction (Brooks et al., Biochem J 307 ( Pt 2), 457-63, 1995, 27, 28). The present application meets the challenge of identifying an in vitro process that converts serine to formylglycine in a secreted serine-mutant protein.
- serine mutants of sulfatases may be prepared and are expressed in recombinant cell lines at much higher levels that the native than the native enzymes under identical conditions.
- data and details presented below demonstrate an in vitro sulfatase processing method in which vanadate and light can be used to convert the serine mutant of sulfatases to catalytically active sulfatases as demonstrated by the conversion of ASB (C91S) to catalytically active ASB.
- Vanadate-dependent photo-conversion of sulfatase mutants represents a novel method of sulfatase production. Methods and compositions for exploiting this finding in the production of members of the sulfatase family for enzyme replacement therapy are provided.
- Vanadate is a mimic of phosphate and sulfate, but vanadium has redox chemistry that phosphorus and sulfur do not have.
- vanadate (V +5 ) can undergo reduction to vanadyl (V +4 ) in the presence of suitable electron sources. This reaction is facilitated by light.
- the electron sources for vanadate reduction are amino acid side-chains.
- the present application for the first time demonstrates that serine mutants of mammalian sulfatases can be activated using a vanadate-dependent photoreaction, which converts the active site serine of these sulfatases to a formylglycine residue thereby rendering the mutants catalytically active.
- the photoreaction involves conversion of the active site serine to a formylglycine.
- the sulfatase preparation is contacted with the a photoactivatable redox agent and ultraviolet light after the sulfatase preparation has been purified from a recombinant cell line, however, it is contemplated that the vanadate-dependent photoreaction may take place while the sulfatase preparation is still in the culture medium.
- photoactivatable redox agent is used to mean an agent that catalyzes a serine to formylglycine change when contacted with a sulfatase in the presence of an energy source from light in the wavelength of 300 nm to about 400 nm or a laser source. In preferred embodiments this agent is a vanadate composition.
- the CHO cell culture from which the recombinant protein is to be isolated is irradiated with U.V. light source of greater than 300 nm and supplied with a source of vanadium ions.
- Vanadate compositions that may be supplied to the sulfatase include but are not limited to ammonium polyvanadate, sodium ammonium vanadate, potassium orthovanadate, potassium metavanadate, potassium vanadate/borate- solution, lithium vanadate, sodium metavanadate, ammonium metavanadate, vanadium tetroxide, vanadium pentoxide, hexavanadium tridekaoxide vanadylacetylacetonate, vanadyl sulphate, vanadium triacetylacetonate, vanadyl oxalate-solution, vanadium trichloride or any other readily available source of vanadium.
- the concentration of vanadium used in the methods of the invention may vary.
- the concentration of vanadium in a solution of sulfatase being activated may range from about 0.01 mM vanadium to about 10 mM.
- the vanadate is present at concentration of less than O.OlmM.
- the vanadate is present at concentrations of 0.01 ⁇ M to O.OlmM.
- the sulfatase mutant and the vanadate are present in equimolar quantities.
- the reaction is a catalytic reaction and as such, it is contemplated that the vanadate may be present at concentration that is less than the concentration of the sulfatase.
- the vanadyl reaction product is re-oxidized to vanadate in the presence of atmospheric oxygen or other oxidizing agent.
- the sulfatase mutant may be contacted with an excess concentration of vanadate and excess vanadate maybe removed from the sulfatase mutant-vanadate complex.
- the vanadium ions may be present at a concentration of 0.01 ⁇ M, 0.05 ⁇ M, 0.10 ⁇ M, 0.2 ⁇ M, 0.4 ⁇ M, 0.5 ⁇ M, 0.6 ⁇ M, 0.7 ⁇ M, 0.8 ⁇ M, 0.9 ⁇ M, 1.0 ⁇ M, 2 ⁇ M, 4 ⁇ M, 5 ⁇ M, 6 ⁇ M, 7, 8 ⁇ M, 9 ⁇ M, 0.01 mM; 0.02 mM; 0.04 mM; 0.05 mM; 0.10 mM; 0.15 mM, 0.2 mM, 0.25 mM, 0.30 mM, 0.35 mM, 0.40 mM, 0.45 mM, 0.50 mM, 0.55 mM, 0.60 mM, 0.65 mM, 0.70 mM, 0.75 mM, 0.80 mM, 0.85 mM, 0.90 mM, 1.0 mM, 1.25 mM,
- Sources of UV light are well known to those of skill in the art as are methods of irradiating compositions.
- a UV light source having a wavelength of from about 300nm to about 400 nm is used to irradiate sulfatases.
- the irradiation may be by way of a constant source of light source.
- the compositions may be irradiated periodically using intermittent periods of irradiation interspersed with periods of full spectrum light or periods of darkness.
- Light of less than 300nm may be filtered with materials that strongly absorb light of less than 300nm.
- An alternative irradiation source is laser light treatment.
- a protocol for pulsed laser photolysis could be very useful in efficiently inducing the photo-dependent activation former and then allowing the light-independent activation to occur while the laser is off, followed by another burst of laser irradiation etc.
- the problem of destructive oxidation of the already activated sulfatase might be addressed if the complex between activated sulfatase and vanadate (which probably results in inactivation upon photolysis) and the complex of sulfatase mutant and vanadate (which is required for activation) may be partly or wholly differentiable by their absorption spectra.
- the interval irradiation and darkness may be optimized such that it is sufficient to activate at least 10%, more preferably 20%, even more preferably 30%, even more preferably 40%, 50%, 60%, 75%, 80%, 85%, 95%, or more of the mutant sulfatase as compared to the absence of the activation by irradiation and vanadate.
- Such an interval will preferably be optimized to determine the time at which cleavage or degradation of the protein . occurs so as to produce an upper limit to the time period of irradiation as that period at which measurable protein degradation or degradation of enzyme activity is observed.
- An exemplary time period for irradiation is 30 minutes, followed by a dark period of 20 minutes.
- This irradiation cylce may be repeated 2, 3, 4, 5, or more times.
- Another exemplary time period of irradiation is 120 minutes. It should be understood that any time period of 10, 20, 30, 45, 60, 75, 90, 120 minutes (of light, optionally followed by a similar period of incubation in the dark) or any multiple of any such time period may be used and optimized for a given sulfatase. Thus, it is contemplated that the in optimization protocols, the skill artisan will subject the mutant sulfatase to varying concentrations of vanadate and varying wavelengths of light and varying time intervals of irradiation.
- each of these parameters will be varied separately such that initially, a first parameter, e.g., vanadate concentration is optimized, followed by optimization of a second parameter e.g., optimization of the wavelength of energy source of irradiation, followed by optimization of interval of time of irradiation.
- a first parameter e.g., vanadate concentration
- a second parameter e.g., optimization of the wavelength of energy source of irradiation
- mutant sulfatases will be prepared and used for a variety of methods and formulations. Methods of preparing such enzymes may involve the production of the proteins using automated peptide synthesis methods or by recombinant expression. Automated peptide synthesis methods are useful for producing short peptide fragments e.g., fragment of 50 amino acids in length whereas recombinant techniques are preferred where a longer protein sequence is to be generated. General principles for making proteins are well known to those of skill in the art. Methods and compositions for the preparation and purification of sulfatases are further described below.
- recombinant sulfatases may be processed in vitro to produce an active enzyme containing a formylglycine residue at the active site.
- the formylglycine residue is generated from the sulfatase having a serine residue at the active site.
- seventeen sulfatases have been identified in humans (Ferrante et al., 2002; Morimoto-Tomita et al., 2002; http ://www.mad- cow.org/00/annotation_frames/tools/genbrow/sulfatases/sulfatases.html).
- arylsulfatases e.g., arylsulfatase A, B, C, D, E, F, G, sulfatase 1, sulfatase 2, iduronate 2 sulfatase, galactosamine (N-acetyl)- ⁇ -sulfate sulfatase, N- sulfoglucosamine sulfohydrolase, and glucosamine (N-acetyl)-6-sulfatase. Homologs of these proteins from various mammalian sources are known to those of skill in the art. A ClustalW alignment of mammalian sulfatases showing the alignment of the active site cysteinyl residues is presented in Figure 1.
- the following table provides the active site cysteine residue and the GenBank Ace. No. of various exemplary mammalian sulfatases. Given that the nucleic acid sequences of these sulfatases are known to those of skill in the art, and that the active site cysteine residue (identified in table below) may be mutated to serine residue using site-directed mutagenesis, those of skill in the art will readily be able to produce recombinant such sulfatases that comprise an active site serine rather than an active site cysteine.
- Such recombinant proteins will serve as substrates for the photoreaction methods of the present invention in which a combination of vanadate and a light source such as a source of light of from about 300 nm to about 400 nm or a pulsating laser light source is employed to render the sulfatases active by conversion of the active site serine residue to a formylgycine residue.
- a light source such as a source of light of from about 300 nm to about 400 nm or a pulsating laser light source is employed to render the sulfatases active by conversion of the active site serine residue to a formylgycine residue.
- arylsulfatase F NM_004042 SEQ ID NO: 12 C78S SEQ ID NO: 32 arylsulfatase G BC012375 SEQ ID NO: 14 C84S SEQ ID NO: 33
- the active sulfatases may be further treated to ensure that the formylglycine is stabilized.
- Formylglycine is an aldehyde.
- chemical reagents e.g., hydrazines, hydroxylamines, bis-alcohols and others
- the active site also contains a metal ion (Ca 2+ or Mg 2+ ) also in the active site of sulfatases. Therefore, different metal ions may also be employed to stabilize the formylglycine by making the formylglycine less reactive. These metal ions could then be substituted with Ca or Mg after the activation reaction is finished (by dialysis for example) to reconstitute the active enzyme.
- sequences of other sulfatases also may be similarly modified.
- those of skill in the art may desire to modify the active sites of sulfatases from other mammalian sources (see e.g., clustal alignment in Figure 1 showing sequences from other sources including rat; bovine; feline; chicken and the like).
- Automated syntheses are typically performed in solution or on a solid support in accordance with conventional techniques.
- Various automatic synthesizers are commercially available and are used in accordance with known protocols. See, for example, Stewart and Young, Solid Phase Peptide Synthesis, 2d. ed., Pierce Chemical Co., (1984);Tam et al., J. Am. Chem. Soc, 105:6442, (1983); Merrifield, Science, 232: 341-347, (1986); and Barany and Merrifield, The Peptides, Gross and Meienhofer, eds, Academic Press, New York, 1-284, (1979); Fields, (1997) Solid- Phase Peptide Synthesis.
- the peptides are synthesized by solid-phase technology employing an exemplary peptide synthesizer such as a Model 433 A from Applied Biosystems Inc.
- an exemplary peptide synthesizer such as a Model 433 A from Applied Biosystems Inc. This instrument combines the FMOC chemistry with the HBTU activation to perform solid-phase peptide synthesis. Synthesis starts with the C- terminal amino acid. Amino acids are then added one at a time till the N-terminus is reached. Three steps are repeated each time an amino acid is added. Initially, there is deprotection of the N-terminal amino acid of the peptide bound to the resin. The second step involves activation and addition of the next amino acid and the third step involves deprotection of the new N-terminal amino acid. In between each step there are washing steps. This type of synthesizer is capable of monitoring the deprotection and coupling steps.
- the protected peptide and the resin are collected, the peptide is then cleaved from the resin and the side-chain protection groups are removed from the peptide. Both the cleavage and deprotection reactions are typically carried out in the presence of 90% TFA, 5% thioanisole and 2.5% ethanedithiol.
- the peptide is precipitated in the presence of MTBE (methyl t-butyl ether). Diethyl ether is used in the case of very hydrophobic peptides.
- the peptide is then washed a plurality of times with MTBE in order to remove the protection groups and to neutralize any residual acidity. The purity of the peptide is further monitored by mass spectrometry and in some case by amino acid analysis and sequencing.
- the peptides also may be modified, and such modifications may be carried out on the synthesizer with very minor interventions.
- An amide could be added at the C-terminus of the peptide.
- An acetyl group could be added to the N- terminus.
- Biotin, stearate and other modifications could also be added to the N- terminus.
- the purity of any given peptide, generated through automated peptide synthesis or through recombinant methods, is typically determined using reverse phase HPLC analysis. Chemical authenticity of each peptide is established by any method well known to those of skill in the art. In certain embodiments, the authenticity is established by mass spectrometry. Additionally, the peptides also are quantified using amino acid analysis in which microwave hydrolyses are conducted. In one aspect, such analyses use a microwave oven such as the CEM Corporation's MDS 2000 microwave oven.
- the peptide (approximately 2 ⁇ g protein) is contacted with e.g., 6 N HCl (Pierce Constant Boiling e.g., about 4 ml) with approximately 0.5% (volume to volume) phenol (Mallinckrodt).
- 6 N HCl Pulierce Constant Boiling e.g., about 4 ml
- phenol Melinckrodt
- the samples Prior to the hydrolysis, the samples are alternately evacuated and flushed with N2.
- the protein hydrolysis is conducted using a two-stage process. During the first stage, the peptides are subjected to a reaction temperature of about 100°C and held that temperature for 1 minute. Immediately after this step, the temperature is increased to 150°C and held at that temperature for about 25 minutes.
- the samples are dried and amino acid from the hydrolysed peptides samples are derivatized using 6-aminoquinolyl-N- hydroxysuccinimidyl carbamate to yield stable ureas that fluoresce at 395 nm (Waters AccQ Tag Chemistry Package).
- the samples are analyzed by reverse phase HPLC and quantification is achieved using an enhanced integrator. Such conditions may readily be adapted for large scale production and/or for purification of other peptides.
- recombinant DNA technology may be employed wherein a nucleotide sequence which encodes a peptide of the invention is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression as described herein below. Recombinant methods are especially preferred for producing longer polypeptides.
- the arylsulfatases of any of sequences of SEQ ID NO:27-SEQ ID NO:39 are prepared using recombinant techniques.
- a variety of expression vector/host systems may be utilized to contain and express the arylsulfatase proteins. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with bacterial expression vectors (e.g., Ti or pBR322 plasmid); or animal cell systems.
- microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transfected with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic
- Mammalian cells that are useful in recombinant protein productions include but are not limited to VERO cells, HeLa cells, Chinese hamster ovary (CHO) cell lines, COS cells (such as COS- 7), W138, BHK, HepG2, 3T3, RIN, MDCK, A549, PC12, K562 and 293 cells.
- the arylsulfatases are prepared in a CHO cell line such as a CHO Kl cell line.
- the recombinant sulfatases may be produced from a stable transfected CHO-Kl cell line prepared according to methods outlined for the preparation of the CSL4S-342 cell line (Crawley, J.Clin.Invest. 99:651-662, 1997; Peters, et al. J. Biol. Chem. 265:3374-3381).
- CSL4S-342 stable transfected CHO-Kl cell line
- other mammalian host systems for the expression of recombinant proteins also are well known to those of skill in the art.
- Host cell strains may be chosen for a particular ability to process the expressed protein or produce certain post-translation modifications that will be useful in providing protein activity. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation and acylation. Post-translational processing, which cleaves a "prepro" form of the protein, may also be important for correct insertion, folding and/or function. Different host cells such as CHO, HeLa, MDCK, 293, WI38, and the like have specific cellular machinery and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the introduced, foreign protein.
- the transformed cells are used for long-term, high- yield protein production and as such stable expression is desirable.
- the cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
- the selectable marker is designed to confer resistance to selection and its presence allows growth and recovery of cells, which successfully express the introduced sequences. Resistant clumps of stably transformed cells can be proliferated using tissue culture techniques appropriate to the cell. A number of selection systems may be used to recover the cells that have been transformed for recombinant protein production.
- Such selection systems include, but are not limited to, HSV thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase and adenine phosphoribosyltransferase genes, in tk-, hgprt- or aprt- cells, respectively.
- anti-metabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate; gpt, which confers resistance to mycophenolic acid; neo, which confers resistance to the aminoglycoside G418; also which confers resistance to chlorsulfuron; and hygro, which confers resistance to hygromycin.
- Additional selectable genes that may be useful include trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine.
- Markers that give a visual indication for identification of transformants include anthocyanins, ⁇ -glucuronidase and its substrate, GUS, and luciferase and its substrate, luciferin.
- expression vectors comprising polynucleotide molecules that encode those arylsulfatases.
- the wild-type proteins of SEQ ID NO:2, SEQ ID NO:4; SEQ H) NO:6; SEQ ID NO:8; SEQ ID NC-.10; SEQ ID NO:12; SEQ ID NO.14; SEQ ID NO:16; SEQ ID NO:18; SEQ ID NO:20; SEQ ID NO:22; SEQ ID NO:24; and SEQ ID NO:26 are encoded by nucleic acid sequence of SEQ ID NO:1; SEQ ID NO:3; SEQ H) NO:5; SEQ ID NO:7; SEQ ID NO:9; SEQ ID NO:11; SEQ ID NO.13; SEQ JD NO:15; SEQ ID NO:17; SEQ ID NO:19; SEQ ID NO:21; SEQ ID NO:23; SEQ and ID NO:25, respectively.
- the expression vectors include DNA encoding the given protein being operably linked to suitable transcriptional or translational regulatory sequences, such as those derived from a mammalian, microbial, viral, or insect gene.
- suitable transcriptional or translational regulatory sequences such as those derived from a mammalian, microbial, viral, or insect gene. Examples of regulatory sequences include transcriptional promoters, operators, or enhancers, rnRNA ribosomal binding sites, and appropriate sequences, which control transcription and translation.
- an expression vector is prepared to transfer a cDNA that encodes an arylsulfatase into a suitable cell or cell line for expression thereof.
- the cDNA is transfected into a Chinese hamster ovary cell, such as the CHO-Kl cell line.
- the production procedure comprises the following steps: (a) growing cells transfected with a DNA encoding all or a biologically active fragment or mutant of any of the sulfatases of SEQ ID NO:27; SEQ ID NO:28; SEQ ID NO:28; SEQ ID NO:29; SEQ ID NO:30; SEQ ID NO:31; SEQ ID NO:32; SEQ ID NO:33; SEQ ID NO:34; SEQ ID NO:35; SEQ E) NO:36; SEQ ID NO:37; SEQ ID NO:38 or SEQ ID NO:39 in a suitable growth medium to an appropriate density, (b) introducing the transfected cells into a bioreactor, (c) supplying a suitable growth medium to the bioreactor, (d) harvesting said medium containing the recombinant enzyme, and (e) substantially removing the transfected cells from the harvest medium.
- expression vector expression construct
- expression cassette any type of genetic construct containing a nucleic acid coding for a gene product in which part or all of the nucleic acid encoding sequence is capable of being transcribed.
- the expression construct may further comprise a selectable marker that allows for the detection of the expression of a peptide or polypeptide.
- a drug selection marker aids in cloning and in the selection of transformants, for example, neomycin, puromycin, hygromycin, DHFR, zeocin and histidinol.
- enzymes such as herpes simplex virus thymidine kinase (tk) (eukaryotic), b-galactosidase, luciferase, or chloramphenicol acetyltransferase (CAT) (prokaryotic) may be employed.
- Immunologic markers also can be employed.
- epitope tags such as the FLAG system (IBI, New Haven, CT), HA and the 6xHis system (Qiagen, Chatsworth, CA) may be employed.
- GST glutathione S-transferase
- NEB maltose binding protein system
- selectable marker employed is not believed to be important, so long as it is capable of being expressed simultaneously with the nucleic acid encoding a gene product. Further examples of selectable markers are well known to one of skill in the art.
- promoter refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the DNA encoding the peptide substrate or the fusion polypeptide. Thus, a promoter nucleotide sequence is operably linked to a given DNA sequence if the promoter nucleotide sequence directs the transcription of the sequence.
- phrase 1 "under transcriptional control" means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression of the gene.
- any promoter that will drive the expression of the nucleic acid may be used.
- the particular promoter employed to control the expression of a nucleic acid sequence of interest is not believed to be important, so long as it is capable of directing the expression of the nucleic acid in the targeted cell.
- a human cell it is preferable to position the nucleic acid coding region adjacent to and under the control of a promoter that is capable of being expressed in a human cell.
- a promoter might include either a human or viral promoter.
- Common promoters include, e.g., the human cytomegalovirus (CMV) immediate early gene promoter, the S V40 early promoter, the Rous sarcoma virus long terminal repeat, ⁇ -actin, rat insulin promoter, the phosphoglycerol kinase promoter and glyceraldehyde-3 -phosphate dehydrogenase promoter, all of which are promoters well known and. readily available to those of skill in the art, can be used to obtain high- level expression of the coding sequence of interest.
- CMV human cytomegalovirus
- Another regulatory element that is used in protein expression is an enhancer. These are genetic elements that increase transcription from a promoter located at a distant position on the same molecule of DNA. Where an expression construct employs a cDNA insert, one will typically desire to include a polyadenylation signal sequence to effect proper polyadenylation of the gene transcript. Any polyadenylation signal sequence recognized by cells of the selected transgenic animal species is suitable for the practice of the invention, such as human or bovine growth hormone and SV40 polyadenylation signals.
- a terminator Also contemplated as an element of the expression cassette is a terminator. These elements can serve to enhance message levels and to minimize read through from the cassette into other sequences.
- the termination region which is employed primarily will be one selected for convenience, since termination regions for the applications such as those contemplated by the present invention appear to be relatively interchangeable.
- the termination region may be native with the transcriptional initiation, may be native to the DNA. sequence of interest, or may be derived for another source. '
- Site-specific mutagenesis will be useful in the present invention to create the C ⁇ S mutants that are described herein above in Table 1.
- This technique employs specific mutagenesis of , the underlying DNA (that encodes the amino acid sequence that is targeted for modification).
- Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences that encode the DNA sequence of the desired mutation, as well as a sufficient number of. . adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed.
- a primer of about 17 to 25 nucleotides in length is preferred, with about 5 to 10 residues on both sides of the junction of the sequence being altered.
- Site-directed mutagenesis typically employs a bacteriophage vector that exists in both a single stranded and double stranded form.
- Typical vectors useful in site-directed mutagenesis include vectors such as the Ml 3 phage. These phage vectors are commercially available and their use is generally well known to those skilled in the art.
- Double stranded plasmids also are routinely employed in site directed mutagenesis, which eliminates the step of transferring the gene of interest from a phage to a plasmid.
- site-directed mutagenesis is performed by first obtaining a single-stranded vector, or melting of two strands of a double stranded vector, which includes within its sequence a DNA sequence encoding the desired protein.
- An oligonucleotide primer bearing the desired mutated sequence is synthetically prepared.
- the codons that encode the active site mutations shown in Table 1 are mutated into serine codons by PCT methods using non-coding mutagenesis primers.
- Such a primer is then annealed with the single-stranded DNA preparation, taking into account the degree of mismatch when selecting hybridization (annealing) conditions, and subjected to DNA polymerizing enzymes such as E.
- heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation.
- This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected that include recombinant vectors bearing the mutated sequence arrangement.
- the present invention is directed to the production of serine mutant recombinant sulfatase proteins and the subsequent in vitro activation of these mutants.
- the proteins are prepared from CHO cells transfected with expression constructs by growing the transfected cells in a suitable growth medium to an appropriate density, introducing the transfected cells into a bioreactor, supplying a suitable growth medium to the bioreactor, harvesting the medium containing the recombinant enzyme, and substantially removing the transfected cells from the harvest medium.
- Exemplary such methods for the production of recombinant arylsulfatase B are provided in U.S. Patent Application No. 10/704,365, filed November 7, 2003 published as U.S. patent publication number (specifically incorporated herein by reference in its entirety).
- an exemplary suitable medium for the growth of the transfected cells is a JRH Excell 302 medium supplemented with L-glutamine, glucose and hypoxanthine/thymidine, optionally with or without G418.
- the JRH Excell 302 medium is further supplemented with folic acid, serine, and asparagine, and there is no G418 present in the medium.
- This medium has been shown to provide a higher purity precursor recombinant arylsulfatase B as compared medium supplemented with G418 but not with folic acid, serine, and asparagine (see U.S. Patent Application No. 10/704,365).
- the CHO cells expressing any of the recombinant sulfatases described herein my be grown in such a medium.
- the cells are grown to a cell density of about 1 x 10 7 cells/ml resulting in 10-40 mg/ml of active enzyme.
- the cells are grown in a bioreactor.
- the transfected cells are grown in a bioreactor for about 5 to 15 days. More preferably, it is about 9 days.
- the transfected cells are grown in a bioreactor using a perfusion-based process with collections continuing up to 35 days. More preferably, the transfected cells are grown in a bioreactor using a perfusion- based process with collections continuing up to 45 days. Even more preferably, the transfected cells are grown in a bioreactor using a perfusion-based process with collections continuing up to 60 days. Even much more preferably, the transfected cells are grown in a bioreactor using a perfusion-based process with collections continuing up to 90 days.
- WO International Application No. PCT US03/26831 WO 2004/020971, incorporated herein by reference.
- the transfected cells may be grown as either a suspension culture or an anchorage-dependent culture in a suitable bioreactors.
- the bioreactors should preferably have high volume-specific culture surface area in order to achieve high cell density and high yield of recombinant product.
- Microcarrier cell culture in stirred tank bioreactor provides very high volume-specific culture surface area and has been used for the production of viral vaccines (Griffiths, J. B., In “Animal Cell Biotechnology", vol. 3, pl79-220, (Eds. Spier, R. E. and Griffiths, J. B.), Academic Press, London. (1986)).
- the multiplate CellcubeTM cell culture system manufactured by Corning-Costar also offers a very high volume-specific culture surface area.
- This motion ensures cell suspension, bulk mixing, and oxygen transfer from the liquid surface to the respiring cells without damaging shear forces or foam generation.
- Air is passed through the bag to provide oxygen, and sweep out evolved carbon dioxide.
- the specially designed bags used in this system are optimized to induce wave motion in the culture media.
- the wave motion provides nutrient mixing and oxygenation to support more than IxIO 7 cells/ml. Therefore, cells can grow to a much higher density than that obtainable with rollers or spinners.
- This type of bioreactor is described in further detail in U.S. Patent No. 6,190,913. Such bioreactors are used for perfusion and the scale up to culture volumes over 500 liters.
- a batch process is a closed system in which a typical growth profile is seen. A lag phase is followed by exponential, stationary and decline phases. In such a system, the environment is continuously changing as nutrients are depleted and metabolites accumulate. This makes analysis of factors influencing cell growth and productivity, and hence optimization of the process, a complex task. Productivity of a batch process may be increased by controlled feeding of key nutrients to prolong the growth cycle. Such a fed-batch process is still a closed system because cells, products and waste products are not removed.
- perfusion of fresh medium through the culture can be achieved by retaining the cells with a variety of devices (e.g., fine mesh spin filter, hollow fiber or flat plate membrane filters, settling tubes).
- Spin filter cultures can produce cell densities of approximately 5 x 10 7 cells/ml.
- a true open system and the simplest perfusion process is the chemostat in which there is an inflow of medium and an outflow of cells and products.
- Culture medium is fed to the reactor at a predetermined and constant rate, which maintains the dilution rate of the culture ⁇ at a value less than the maximum specific growth rate of the cells (to prevent washout of the cell mass from the reactor).
- Culture fluid containing cells and cell products and byproducts is removed at the same rate.
- Roller-bottles may be used to anchorage-dependent non-perfused attachment cell culture.
- Fully automated robots are available that can handle thousands of roller bottles per day, thus eliminating the risk of contamination and inconsistency associated with the otherwise required intense human handling.
- roller bottle cultures can achieve cell densities of close to 0.5 x 10 6 cells/cm 2 (corresponding to approximately 10 9 cells/bottle or almost 10 7 cells/ml of culture media).
- Microcarriers also may be used in large scale anchorage-dependent • culture processes.
- the transfected cells are propagated on the surface of small solid particles suspended in the growth medium by slow agitation. Cells attach to the microcarriers and grow gradually to confluency on the microcarrier surface.
- Microcarrier cultures offer a high surface-to-volume ratio (variable by changing the carrier concentration) which leads to high cell density yields and a potential for obtaining highly concentrated cell products. Cell yields are up to 1-2 x 10 7 cells/ml when cultures are propagated in a perfused reactor mode.
- the cells can be propagated in one unit process vessels instead of using many small low-productivity vessels (i.e., flasks or dishes), thereby resulting in a more efficient use of nutrient and culture medium.
- a microcarrier system is discussed hi WO 2004/020971.
- the CHO cells may be cultured using microencapsulation.
- the cells are retained inside a semipermeable hydrogel membrane.
- a porous membrane is formed around the cells permitting the exchange of nutrients, gases, and metabolic products with the bulk medium surrounding the capsule.
- Microencapsulated cells are easily propagated in stirred tank reactors and, with beads sizes in the range of 150-1500 ⁇ m in diameter, are easily retained in a perfused reactor using a fine-meshed screen.
- the ratio of capsule volume to total media volume can be maintained from as dense as 1 :2 to 1 :10.
- intracapsular cell densities of up to 10 8 the effective cell density in the culture is 1-5 x 10 7 .
- microencapsulation over other processes include the protection from the deleterious effects of shear stresses which occur from sparging and agitation, the ability to easily retain beads for the purpose of using perfused systems, scale up is relatively straightforward and the ability to use the beads for implantation.
- the cells are initially inoculated into T-75 flask with 25mL of JRH Exell 302 medium supplemented with 4 niM L-glutamine, 4.5 g/L glucose and 10mg/L hypoxanthine/thymidine; further supplemented with folic acid, serine and asparagine (no G418). These cells are cultured in this medium for approximately three days or until cell density of approximately 1 x 10 10 cell is achieved.
- the cells may be transferred to a 250 niL spinner flask and cultured in a volume of 175 mL of supplemented medium (no G418), this cell culture is allowed to grow to an exponential phase (approx. 3 days).
- the cells are then transferred to a 1 L flask and grown in a 800 mL medium supplemented (no G418).
- the cells are transferred to an 8 L spinner flask and grown in a volume of 4L medium for 1 to 2 days.
- the 4 liters of cells are then divided into two 8 L flasks and grown for a further 1-2 days in 5.5 L volumes of medium in each 8 L flask.
- the cells from the two 8 L flasks are harvested, resuspended in 7 mL medium and used to inoculate a 110 L bioreactor.
- the cells are grown in 100 L of medium for a period of approx. 9 days.
- the supernatant from the bioreactor is pumped into 100 L bags and refrigerated overnight.
- the cells are then removed from the harvested medium by filtration through a lO ⁇ m membrane cartridge followed by l ⁇ m and 0.2 ⁇ m cartridges. Since the cells have been allowed to settle overnight the final 5 to 10% of the harvest medium is discarded prior to filtration.
- the CHO cells are grown in a perfused attachment system in which there is a continuous flow of medium at a steady rate, through or over a population of cells (of a physiological nutrient solution). More particularly, the transfected cells are grown in a cell culture process that is a perfusion-based process with collections continuing for up to 35 or more days with a collection rate of approximately 400 L per day from one 110 L bioreactor. Preferably, the collection rate is approximately 800 L per day from one 110 L bioreactor.
- the following table provides a comparison between an exemplary batch-fed process and a perfusion process for the production of recombinant sulfatase proteins from the transfected CHO cells.
- The. inoculum preparation for scale-up process is the same for the fed batch and perfusion processes.
- the recombinant cell culture is initiated by thawing a single vial from the Working Cell Bank and transferring its contents (approximately 1 mL) to approximately 25 mL of EX-CELL 302 Medium (Modified w/ L-Glutamine, No Phenol Red) in a T75 cm2 cell culture flask.
- EX-CELL 302 Medium Modified w/ L-Glutamine, No Phenol Red
- the cell culture is incubated until a viable cell count of approximately 0.8 x 10 6 cells/mL is achieved.
- Each cell expansion step is monitored for cell growth (cell density) and viability (via trypan blue exclusion).
- EX-CELL 302 Medium Modified w/ L-Glutamine, No Phenol Red
- cell transfers are preformed aseptically in a laminar flow hood.
- the cell culture is expanded sequentially from the T75 cm2 flask to a 250 mL spinner flask, to two 250 mL spinner flasks, to two 3 L spinner flasks, and finally to two 8 L spinner flasks.
- the entire scale-up process lasts approximately 14 days.
- the two 8 L spinner flasks are at a density of at least 1.0 x 10 6 cells/mL, the flasks are used to seed one 110 L bioreactor.
- the bioreactor operations for recombinant sulfatase expression or production or manufacture utilize a perfusion- based cell culture process.
- the bioreactor, using the perfusion process can control cell densities up to as high as 37 million cells per mL; compared to 4-5 million cells per mL using the fed batch process.
- the perfusion-based process runs longer (35 days) than the fed batch process (11 to 12 days) and produces a greater .
- volume of harvested cell culture fluid approximately 400 L/day at a perfusion rate of 5 vessel volumes per day
- the fed batch process 190 L/run.
- harvesting is performed up to 35 days for a total collection of approximately 8400 L of supernatant.
- cell counts and viability determinations may be made. After the final removal of the cells from the harvested medium in either procedure, the cells are assessed for quality control purposes. The end of production cells are evaluated for genetic stability, identity, sterility and adventitious agent contamination per ICH guideline. Preferably, EPC results, obtained from a 35-day long bioreactor run, AC60108, produced under cGMP conditions, show no growth or negative results or no detection of the presence of bacteria and fungi, mycoplasma, adventitious viral contaminants, murine viruses, or like contaminants or particles.
- EPC results obtained from a 35-day long bioreactor run, AC60108, produced under cGMP conditions, show no growth or negative results or no detection of the presence of bacteria and fungi, mycoplasma, adventitious viral contaminants, murine viruses, or like contaminants or particles.
- arylsulfatase proteins may be purified from the supernatant of the cell culture.
- purification may employ purification techniques that are well known to those of skill in the art. These techniques involve, at one level, the crude fractionation of the cellular milieu to polypeptide and non- polypeptide fractions. Having separated the recombinant proteins from other CHO and other culture derived proteins, the sulfatase proteins of interest may be further purified using chromatographic and electrophoretic techniques to achieve partial or complete purification (or purification to homogeneity).
- Analytical methods particularly suited to the preparation of a pure peptide are ion-exchange chromatography, exclusion chromatography; polyacrylamide gel electrophoresis; isoelectric focusing.
- Particularly efficient methods of purifying peptides include fast protein liquid chromatography (FPLC) and high performance liquid chromatography (HPLC).
- FPLC fast protein liquid chromatography
- HPLC high performance liquid chromatography
- Certain aspects of the present invention concern the purification, and in particular embodiments, the substantial purification, of an encoded polypeptide, protein or peptide.
- the term "purified polypeptide, protein or peptide" as used herein, is intended to refer to a composition, isolated from other components, wherein the polypeptide, protein or peptide is purified to any degree relative to its naturally- obtainable state.
- a purified polypeptide, protein or peptide therefore also refers to a polypeptide, protein or peptide, free from the environment in which it may naturally occur.
- purified will refer to a polypeptide, protein or peptide composition that has been subjected to fractionation to remove various other components, and which composition substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation will refer to a composition in which the polypeptide, protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the proteins in the composition.
- the quality of the recombinant enzyme is key to patients. In preferred embodiments, the recombinant enzyme is substantially pure to a purity level greater than 90%, preferably greater than 91%, greater than 92%, greater than 93%, greater than 94%.
- the recombinant protein is at least 95% and does not contain contaminants from the CHO, such as CHO proteins.
- the recombinant protein is at least 96%, at least 97%, at least 98%, at least 99% pure.
- the protein is 99.2% pure, 99.4% pure, 99.5% pure, 99.6% pure, 99.8% pure or 100% pure.
- Various techniques suitable for use in protein purification will be well known to those of skill in the art.
- the cellular milieu is separated from the supernatant to prevent fouling of the columns.
- the growth medium containing the recombinant enzyme is passed through an ultrafiltration and diafiltration step.
- the filtered solution is passed through a DEAE Sepharose chromatography column, then a Blue Sepharose chromatography column, then a Cu++ Chelating Sepharose chromatography column, and then a Phenyl Sepharc%e chromatography column.
- Such a four step column chromatography including using a DEAE Sepharose, a Blue Sepharose, a Cu++ Chelating Sepharose and a Phenyl Sepharose chromatography column sequentially results in especially highly purified recombinant enzyme.
- chromatography steps may be omitted or substituted or the order of the steps altered within the scope of the present invention.
- the eluent from the final chromatography column is ultrafiltered/diafiltered, and an appropriate step is performed to remove any remaining viruses.
- appropriate sterilizing steps may be performed as desired.
- the ultrafiltration/diafiltration step is performed with a sodium phosphate solution of about 10 mM and with a sodium chloride solution of about 100 mM at a pH of about 7.3.
- the DEAE Sepharose chromatography step is performed at a pH of about 7.3 wherein the elute solution is adjusted with an appropriate buffer, preferably a sodium chloride and sodium phosphate buffer.
- the Blue Sepharose chromatography step is performed at a pH of about 5.5 wherein the elute solution is adjusted with an appropriate buffer, preferably a sodium chloride and sodium acetate buffer.
- the Cu++ Chelating Sepharose chromatography step is performed with an elution buffer including sodium chloride and sodium acetate.
- a second ultrafiltration/diafiltration step is performed on the eluate from the chromatography runs wherein the recombinant enzyme is concentrated to a concentration of about 1 mg/ml in a formulation buffer such as a sodium chloride and sodium phosphate buffer to a pH of about 5.5 to 6.0, most preferably to a pH of 5.8.
- Phosphate buffer is a preferred buffer used in the process because phosphate buffer prevents critical degradation and improves the stability of the enzyme.
- the initial purification of the recombinant sulfatases comprises an ultrafiltration/diafiltration step in which filtered harvest fluid (e.g., the filtered supernatant from the bulk cell culture described above) is concentrated ten fold and then diafiltered with 5 volumes of 10 mM Sodium Phosphate, 100 mM NaCl, pH 7.3 using a tangential flow filtration (TFF) system.
- filtered harvest fluid e.g., the filtered supernatant from the bulk cell culture described above
- diafiltered e.g., the filtered supernatant from the bulk cell culture described above
- step 2 the diafiltrate is then subjected to a first chromatographic separation on a
- DEAE Sepharose FF flow through column.
- the DEAE sepharose column Prior to loading, the DEAE sepharose column is prewashed and equilibrated: it is prewashed with buffer 1 (0.1 N NaOH) and then buffer 2 (100 mM NaPO4 pH 7.3). The column is then equilibrated with an equilibration buffer (100 mM NaCl, 10 mM NaPO4, pH 7.3). Once the column has been equilibrated, the product from the ultrafiltration/diafiltration step is loaded onto the column and washed (wash buffe ⁇ lOO mM NaCl, 1OmM NaPO4, pH 7.3).
- the protein is eluted from the column with a strip buffer (I M NaCl, 10 mM NaPO4, pH 7.3).
- the column is then sanitized (sanitization buffer:0.5 N NaOH) and stored (storage buffer: 0.1 N NaOH).
- the protein eluted from the DEAE Sepharose FF column in step 2 is applied to a second chromatographic separation on a Blue Sepharose FF.
- This column is prewashed with 0.1 N NaOH, followed by a washing step with distilled water and a third prewash with 1 M NaAc, pH 5.5.
- This washed column is equilibrated with 150 niM NaCl, 20 mM NaAc, pH 5.5, before the DEAE Sepharose FF column flow through is loaded. After the protein has been loaded onto the column, the loaded column is washed (wash buffer: 150 mM NaCl, 20 mM NaAc, pH 5.5) and the protein is eluted (elution buffer: 500 mM NaCl, 20 mM NaAc, pH 5.5).
- the Blue Sepharose column is regenerated (regeneration buffer: 1 M NaCl, 20 mM NaAc, pH 5.5); sanitized .(sanitization buffer: 0.1 N NaOH, 0.5-2 hours) and stored (storage buffer: 500 mM NaCl, 20 mM NaAc, pH 5.5, 20% ETOH).
- the eluted protein from the Blue Sepharose column in step 3 is subjected to a fourth chromatographic separation in which the eluate from step 3 is subjected to metal ion chelation chromatography on a Cu++ Chelating Sepharose FF chromatography column.
- the Cu++ column Prior to application of the eluate from step 3, the Cu++ column is sanitized with 0.1 N NaOH, and washed with water.
- the sepharose column is then charged with metal ions using a solution of 0.1 M Copper Sulfate and equilibrated in an appropriate buffer (20 mM NaAc, 0.5 M NaCl, 10% Glycerol, pH 6.0).
- the eluate from step 3 is loaded onto the column and washed with an initial wash buffer (20 mM NaAc, 0.5 M NaCl, 10%' Glycerol, pH 6.0), the loaded column is then washed with a second wash buffer (20 • mM NaAc, 1 M NaCl, 10% Glycerol, pH 4.0), followed by a third washing step (20 mM NaAc, 1 M NaCl, 10% Glycerol, pH 3.8). After washing, the protein is eluted (elution buffer: 20 mM NaAc, 1 M NaCl, 10% Glycerol, pH 3.6).
- the column is then stripped of any remaining metal ions using a strip buffer (50 mM EDTA, 1 M NaCl); sanitized (sanitization Buffer: 0.5 N NaOH, 0.5-2 hours) and stored (storage buffer: 0.1 N NaOH).
- strip buffer 50 mM EDTA, 1 M NaCl
- sanitized 0.5 N NaOH, 0.5-2 hours
- storage buffer 0.1 N NaOH
- a fifth step the eluted proteins from the Cu++ column are subjected to a fourth chromatographic separation in which the eluate is applied to a Phenyl Sepharose HP column.
- the phenyl sepharose HP column Prior to loading of the eluate from step 4, the phenyl sepharose HP column is prewashed with 0.1 N NaOH followed by distilled water and then equilibrated (equilibration buffer: 3 M NaCl, 20 mM NaAc, pH 4.5).
- the Cu++ Chelating Sepharose eluate is then loaded onto the column and washed initially with buffer 1 (3.0 M NaCl , 20 mM NaAc, pH 4.5) and then with buffer 2 (1.5 M NaCl, 20 mM NaAc, pH 4.5).
- the proteins are eluted (elution buffer: 1.0 M NaCl, 20 mM, NaAc, pH 4.5).
- the column is then stripped (strip buffer: 0 M NaCl, 20 mM NaAc, pH 4.5), sanitized (sanitization Buffer: 0.5 N NaOH) and stored (storage Buffer: 0.1 N NaOH).
- step 6 the recombinant protein is concentrated and diafiltered to a final concentration of 1.5 mg/ml in formulation buffer (150 mM NaCl, 10 mM NaPO4, pH 5.8) using a TFF system. If necessary, this preparation may be diluted in formulation buffer to a concentration of 1.0mg/ml. The preparation is then sterilized and the viral load reduced.
- the sterilized bulk drug substance can be sterilized through a 0.04 micron or preferably a 2 micron filter in a class 100 laminar flow hood into Type 1 glass vials. The vials may be filled to a final volume of about 5mL using a semi-automatic liquid filling machine.
- the vials may then be manually stoppered, sealed and labeled, hi specific embodiments, the vials contain lmg/mL of the sulfatase along with excipients sodium phosphate, monobasic, 1 H20 (9mM), sodium phosphate, dibasic, 7 H20 (ImM) and sodium chloride (15OmM).
- the vanadate-dependent photoreaction may be carried out at any stage during this purification process.
- the photoreaction is carried out immediately prior to placing the sulfatase drug product into the vials.
- Another preferred method of purifying recombinant sulfatase protein comprises: (a) obtaining a fluid containing the sulfatase; (b) contacting the fluid with a Cibracon blue dye interaction chromatography resin; (c) contacting the fluid with a copper chelation chromatography resin; (d) contacting the fluid with a phenyl hydrophobic interaction chromatography resin; and (e) recovering the sulfatase protein.
- steps (b), (c) and (d) are performed sequentially. This method requires no more than three chromatography steps or columns. In order to obtain highly purified sulfatase no further chromatography steps or columns are required. Thus, the method does not comprise the fluid contacting a DEAE Sepharose resin.
- the recovered recombinant protein preferably has a purity of at least equal to or greater than 95%.
- the overall recovery yield can be at least about 40-60%.
- step (b) comprises passing the fluid through a Cibracon blue dye interaction chromatography column. More preferably, the Cibracon blue dye interaction chromatography column is a Blue Sepharose 6 Fast Flow column.
- step (c) comprises passing the fluid through a copper chelation chromatography column. More preferably, the copper chelation chromatography column is a Chelating Sepharose Fast Flow column.
- step (d) comprises passing the fluid through a phenyl hydrophobic interaction chromatography column. More preferably, the phenyl hydrophobic interaction chromatography column is a Phenyl Sepharose 6 Fast Flow High Sub column.
- the temporal sequence of steps (b), (c) and (d) is step (b), step (c) and step (d).
- the recovering step can comprise ultrafiltration and/or diafiltration of the fluid.
- the recovering can comprise filtering the fluid to remove DNA and/or filtering the fluid to remove virus.
- the filtering, for removing virus can comprise passing said fluid through a 0.02 ⁇ m filter.
- RP- HPLC reverse-phase high performance liquid chromatography
- Proteins are detected as peaks on a chromatogram by ultraviolet absorbance at 210 nni. The areas of each peak are calculated, and the sample purity can be calculated as the ratio of the recombinant sulfatase protein peak area to the total area of all peaks in the chromatogram.
- RP- HPLC is a proven high-resolution, reproducible method of determining the purity of recombinant sulfatases such as recombinant human arylsulfatase B.
- ELISA also may be. used to determine the purity of the proteins.
- sulfatases produced and purified herein may be tested for their activity in vitro and efficacy in vivo.
- Example 1 Such assays typically incubate the protein composition in the presence of a substrate to be cleaved. The cleavage of the substrate is then monitored using spectrophotometry or fluorometery.
- the substrate may be one, which is radiolabeled, and as such the detection of the cleavage products may be achieved through detection of the radioactive label.
- the cleavage products may be separated using chromatography, e.g., thin layer chromatography, HPLC and the like.
- the assays may be varied depending on the particular protein and substrate being tested.
- the recombinant activated protein of the present invention will be tested in vivo in animal models and in a clinical setting.
- Canine and feline models for diseases such as MPS VI and MPS I have been developed.
- such models maybe treated with a dosage of a given enzyme ranging from about 0.1 to about lOmg/kg, more preferably between about 1 mg/kg and about 5 mg/kg and more particularly at about 2 mg/kg and clinical parameters of the disease are monitored. Studies also may be performed to compare enzyme distribution, clearance of tissue glycosaminoglycan storage, sulfatide storage and decrease of urinary glycosaminoglycans or sulfatide levels after bolus and slow (2 hour) infusion.
- a dosage of a given enzyme ranging from about 0.1 to about lOmg/kg, more preferably between about 1 mg/kg and about 5 mg/kg and more particularly at about 2 mg/kg and clinical parameters of the disease are monitored. Studies also may be performed to compare enzyme distribution, clearance of tissue glycosaminoglycan storage, sulfatide storage and decrease of urinary glycosaminoglycans or sulfatide levels after bolus and slow (2 hour) infusion.
- Other symptoms that may be monitored include facial dysmorphia, Diffuse corneal clouding, epiphyseal dysplasia, Subluxations, Pectus excavatum, reduced body weight, reduced spine flexibility, hind limb gait, hind limb paralysis, and the like.
- Other biochemical/morphological determinations indicate that by 35 days, organs of untreated cats have maximal storage of glycosaminoglycans in tissues (Crawley, et al., J. Clin. Invest. 99:651-662 (1997)).
- Urinary glycosaminoglycan levels are elevated at birth in both normal and MPS VI cats but after approximately 40 days, normal cats have decreased levels. Any alleviation of these symptoms will be useful in the present invention.
- the recombinant enzyme should produce an improvement in urinary sulfatides, body weight/growth, bone morphometry and clearance of stored material (i.e., the. material that is stored in the lysosomal disorder associated with the particular enzyme deficiency) from several tissues relative to these parameters in the absence of the treatment.
- stored material i.e., the. material that is stored in the lysosomal disorder associated with the particular enzyme deficiency
- the dosage ranges and efficacy parameters identified from such animal studies may be used for the development of clinical protocols in humans Moreover, many related enzymes are already in human clinical trials and as such, the protocols used in those clinical trials will be effective in testing the compositions of the present invention.
- the clinical development program for the recombinant sulfatase will comprise an initial open-label clinical trial that will provide an assessment of weekly infusions of the enzyme for safety, pharmacokinetics, and initial response of both surrogate and defined clinical eridpoints. Such evaluation will be conducted for a minimum of three months to collect sufficient safety information for 5 evaluable patients. At this time, should the initial dose of 1 mg/kg not produce a reasonable reduction in excess urinary glycosaminoglycans or produce a significant direct clinical benefit, the dose will be doubled and maintained for an additional three months to establish safety and to evaluate further efficacy.
- a timed walk test or stair climb (as a measure of exercise tolerance), full pulmonary function (PFT) evaluation, reduction in levels of urinary sulfatides or other storage products produced due to lack of specific enzyme involved and hepatomegaly (as measures of kidney, CNS and liver storage of sulfatides or other storage products produces due to the specific enzyme deficiency), growth velocity, joint range of motion, Children's Health Assessment Questionnaire (CHAQ), visual acuity, cardiac function, sleeping studies, and two different global assessments; one performed by the investigator, one performed by the patient/caregiver.
- PFT full pulmonary function
- CHAIQ Children's Health Assessment Questionnaire
- a second secondary objective is to determine pharmacokinetic parameters of infused drug in the circulation, and general distribution and half-life of intracellular enzyme using leukocytes and buccal tissue as sources of tissue. It is anticipated that these measures will help relate dose to clinical response based on the levels of enzyme delivered to the lysosomes of cells.
- the subjects will be admitted for a two week baseline evaluation that will include a medical history and physical exam, psychological testing, endurance testing (treadmill), a standard set of clinical laboratory tests (CBC, Panel 20, CH50, UA), a MRI or CAT scan of the body (liver and spleen volumetric determination, bone and bone marrow evaluation, and lymph node and tonsillar size), a cardiology evaluation (echocardiogram, EKG, CXR), an airway evaluation (pulmonary function tests), a sleep study to evaluate for obstructive events during sleep, a joint restriction analysis (range of motion will be measured at the elbows and interphalangeal joints), a LP with CNS pressure, and biochemical studies (buccal sulfatase activity on two occasions, leukocyte sulfatase activity on two occasions, urinary sulfatide on three occasions, serum generation for ELISA of anti-sulfatase antibodies and 24 hour urine for creatinine clearance).
- a medical history and physical exam that will include a
- each patient will be photographed and videotaped performing some physical movements such as attempting to raise their hands over their heads and walking. Patients will be titrated with antihistamines such that pretreatment with these agents could be effectively employed prior to infusion of enzyme.
- An exemplary proposed human dose of 1 mg/kg (50 U/kg) or 2mg/kg (100U/kg) will be administered weekly by i.v. infusion over 4 hours. The patient will remain in the hospital for the first two weeks, followed by short stays for the next four weeks. Treatment for the final six weeks will be conducted at a facility close to the patient's home. Patients will return to the hospital for a complete evaluation at three months.
- the patient may be male or female, aged five years or older with a documented diagnosis of the lysosomal storage disease to be treated confirmed by measurable clinical signs and symptoms of that disease, and supported by a diminished fibroblast or leukocyte enzyme activity level of the sulfatase enzyme in question.
- Female patients of childbearing potential must have a negative pregnancy test (urine ⁇ -hCG) just prior to each dosing and must be advised to use a medically accepted method of contraception throughout the study.
- a patient will be excluded from this study if the patient has previously undergone bone marrow transplantation; is pregnant or lactating; has received an investigational drug within 30 days prior to study enrollment; or has a medical condition, serious intercurrent illness, or other extenuating circumstance that may significantly decrease study compliance.
- Patients will receive the recombinant sulfatase enzyme at a dose of e.g., 1 mg/kg (-50 U/kg) for the first 3 months of the study. In the event that excess urine sulfatides are not decreased by a reasonable amount and no clinical benefit is observed, the dose will be doubled. Dose escalation will occur only after all 5 patients have undergone 3 months of therapy.
- This recombinant enzyme dosage form will be administered intravenously over approximately a four-hour period once weekly for a minimum of 12 consecutive weeks.
- a peripheral intravenous catheter will be placed in the cephalic or other appropriate vein and an infusion of saline begun at 30 cc/hr.
- the patient will be premedicated with up to 1.25 mg/kg of diphenylhydramine intravenously based on titration experiments completed prior to the trial.
- the recombinant enzyme is diluted into 100 cc of normal saline supplemented with 1 mg/ml human albumin.
- the diluted enzyme is then infused at 1 mg/kg (about 50 units per kg) over a 4 hour period with cardiorespiratory and pulse oximeter monitoring.
- the patients are monitored clinically as well as for any adverse reaction to the infusion. If any unusual symptoms are observed, including but not limited to malaise, shortness of breath, hypoxemia, hypotension, tachycardia, nausea, chills, fever, and abdominal pain, the infusion will be stopped immediately.
- an additional dose of diphenylhydramine, oxygen by mask, a bolus of intravenous fluids or other appropriate clinical interventions such as steroid treatment may be administered. If an acute reaction does occur, an assessment for the consumption of complement in the serum will be tested. A second i.v. site will be used for the sampling required for pharmacokinetic analysis.
- the enzyme therapy is determined to be safe if no significant acute reactions occur that cannot be prevented by altering the rate of administration of the enzyme, or acute antihistamine or steroid use.
- the longer-term administration of the enzyme will be determined to be safe if no significant abnormalities are observed in the clinical examinations, clinical labs, or other appropriate studies.
- the presence of antibodies or complement activation will not by themselves be considered unsafe, but such antibodies will require monitoring by ELISA, and by clinical assessments of possible immune complex disease.
- Improvements in the surrogate and clinical endpoints are expected as a result of delivery of enzyme and removal of glycosaininoglycan storage from the body. Dose escalation can be performed if mean excess levels urinary glycosaminoglycans, sulfatides and other products of the lysosomal storage disease are not reduced by a reasonable amount over three months and no significant clinical benefit is observed at 3 months. Improvements should include improved airway index or resolution of sleep apnea, improved j oint mobility, and increased endurance.
- the enzyme replacement therapy will advantageously result in normalization of liver volume and urinary glycosaminoglycan excretion, reduction in spleen size and apnea/hypopnea events, increase in height and growth velocity in prepubertal subjects, increase in shoulder flexion and elbow and knee extension, and reduction in tricuspid regurgitation or pulmonic regurgitation.
- those of skill in the art are specifically referred to Example 5 of U.S. Patent No. 6,585,971 for assessing- such effects.
- the therapeutic administering of the recombinant sulfatase involves administration of human recombinant enzyme, which reduces lysosomal storage in cells of the individual having the lysosomal storage disease.
- the composition being delivered comprises about 1 mg recombinant enzyme/20 kg of body weight of the mammal being treated for the disorder.
- the enzyme being administered to the subject is one, which comprises a moiety that may be readily taken up by a high affinity uptake receptor on the surface of a cell.
- a high affinity uptake receptor on the surface of a cell.
- such a receptor may be the mannose-6-phosphate receptor and the enzyme may be modified to comprise up to about an average of about at least 20% bis-phosphorylated oligosaccharides per enzyme.
- the enzyme may comprise 10%, 15%, 18%, 20%, 25%, 30%, 35%, 40%, 45% bis-phosphorylated oligosaccharides per enzyme.
- the lysosomal enzymes for use in the present invention may be conjugated to a RAP and RAP polypeptides, which selectively bind to LRP receptors such as megalin that may be present on cells.
- RAP or other megalin ligand molecules will serve to increase the transport of the lysosomal enzyme across the blood brain barrier and/or deliver agents to lysosomes of cells within the CNS.
- Methods and compositions for preparing enzyme compositions that comprise RAP moieties attached thereto are described in detail in U.S. Patent Application No. 10/206,448, filed on July 25, 2002 and in U.S. Patent Application No. 10/600,862, filed June 20, 2003, each incorporated herein by reference.
- those of skill in the art may employ a delivery of the enzyme conjugated to melanotransferrin (p97) as described in e.g.,
- compositions for administration according to the present invention can comprise the recombinant enzymes described herein either ' alone or e.g., conjugated to moiety that facilitates the transcytosis of the sulfatase enzyme.
- the pharmaceutical compositions also may include additional therapeutic agents for the treatment of the given disease being treated. Regardless of whether the active component of the pharmaceutical composition is an enzyme alone, an enzyme conjugated to an uptake moiety, or the combination of either or both of these former entities with yet another therapeutic composition, each of these preparations is in some aspects provided in a pharmaceutically acceptable form optionally combined: with a pharmaceutically acceptable carrier.
- compositions for the treatment of the given ⁇ disorder are determined readily by those with ordinary skill in the art using assays that are used for the diagnosis of the disorder and determining the level of effect a given therapeutic intervention produces.
- the suitable dose of a composition according to the present invention will depend upon the age, health and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
- the dosage is tailored to the individual subject, as is understood and determinable by one of skill in the art, without undue experimentation. This typically involves adjustment of a standard dose, e.g.-, reduction of the dose if the patient has a low body weight.
- the total dose of therapeutic recombinant sulfatase enzyme may be administered in multiple doses or in a single dose.
- the compositions are administered alone, in other embodiments the compositions are administered in conjunction with other therapeutics directed to the disease or directed to other symptoms thereof.
- compositions of the invention are formulated into suitable pharmaceutical compositions, i.e., in a form appropriate for in vivo applications in the therapeutic intervention of a given disease. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals. In some aspects, the compositions are prepared for administration directly to the lung. These formulations are for oral administration via an inhalant, however, other routes of administration are contemplated (e.g. injection and the like).
- An inhaler device is any device useful in the administration of the inhalable medicament.
- inhaler devices include nebulizers, metered dose inhalers, dry powder inhalers, intermittent positive pressure breathing apparatuses, humidifiers, bubble environments, oxygen chambers, oxygen masks and artificial respirators.
- the enzyme is administered by introduction into the central nervous system of the subject, e.g., into the cerebrospinal fluid of the subject.
- the enzyme is introduced intrathecally, e.g., into the lumbar area, or the cistema magna or intraventricularly into a cerebral . ventricle space.
- the therapy may be given using an Ommaya reservoir, which is in common use for intrathecally administering drugs for meningeal carcinomatosis (Lancet 2: 983-84, 1963). More specifically, in this method, a ventricular tube is inserted through a hole formed in the anterior horn and is connected to an Ommaya reservoir installed under the scalp, and the reservoir is subcutaneously punctured to intrathecally deliver the particular enzyme being replaced, which is injected into the reservoir.
- Other devices for intrathecal administration of therapeutic compositions to an individual are described in U.S. Patent No. 6,217,552, incorporated herein by reference.
- the drug maybe intrathecally given, for example, by a single injection, or continuous infusion. It should be understood that the dosage treatment may be in the form of a single dose administration or multiple doses.
- the term "intrathecal administration” is intended to include delivering a pharmaceutical composition directly into the cerebrospinal fluid of a subject, by techniques including lateral cerebroventricular injection through a burrhole or cisternal or lumbar puncture or the like (described in Lazorthes et al. Advances in Drug Delivery Systems and Applications in Neurosurgery, 143-192 and Omaya et al, Cancer Drug Delivery, 1 : 169-179, the contents of which are incorporated herein by reference).
- the term "lumbar region” is intended to include the area between the third and fourth lumbar (lower back) vertebrae and, more inclusively, the L2-S1 region of the spine.
- compositions in accordance with the present invention are intended to include access to the space around and below the cerebellum via the opening between the skull and the top of the spine.
- Cerebral ventricle is intended to include the cavities in the brain that are continuous with the central canal of the spinal cord.
- Administration of a pharmaceutical composition in accordance with the present invention to any of the above mentioned sites can be achieved by direct injection of the composition or by the use of infusion pumps.
- the composition of the invention can be formulated in liquid solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution or phosphate buffer.
- the enzyme may be formulated in solid form and re-dissolved or suspended immediately prior to use. Lyophilized forms are also included.
- the injection can be, for example, in the form of a bolus injection or continuous infusion (e.g., using infusion pumps) of the enzyme.
- the pharmaceutically acceptable formulation provides sustained delivery, e.g., "slow release" of the enzyme or other pharmaceutical composition used in the present invention, to a subject for at least one, two, three, four weeks or longer periods of time after the pharmaceutically acceptable formulation is administered to the subject.
- sustained delivery is intended to include > continual delivery of a pharmaceutical composition of the invention in vivo over a period of time following administration, preferably at least several days, a week or several weeks.
- Sustained delivery of the composition can be demonstrated by, for example, the continued therapeutic effect of the enzyme over time (e.g., sustained delivery of the enzyme can be demonstrated by continued reduced amount of storage granules in the subject). Alternatively, sustained delivery of the enzyme may be demonstrated by detecting the presence of the enzyme in vivo over time.
- the pharmaceutical formulation used in the method of the invention contains a therapeutically effective amount of an enzyme for use in enzyme replacement therapy of a lysosome storage disease. Such a therapeutically effective amount is any amount effective, at dosages and for periods of time necessary, to achieve the desired result.
- the compositions comprises a therapeutically effective amount of a sulphatase such as arylsulfatase B.
- a therapeutically effective amount of a sulfatase may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the enzyme (alone or in combination with one or more other agents) to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.
- a non-limiting range for a therapeutically effective concentration of enzyme is O.OOl ⁇ g enzyme/ml to about 150 ⁇ g enzyme/ml. It is to be noted that dosage values may vary with the severity of the condition to be alleviated.
- compositions of the invention are provided in , . lyophilized form to be reconstituted prior to 1 administration.
- the pharmaceutical compositions may be formulated into tablet form. Buffers and solutions for the reconstitution of the pharmaceutical compositions may be provided along with the pharmaceutical formulation to produce aqueous compositions of the present invention for administration.
- aqueous compositions will comprise an effective amount of each of the therapeutic agents being used, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium.
- Such compositions also are referred to as inocula.
- phrases "pharmaceutically or pharmacologically acceptable” refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the therapeutic compositions, its use in therapeutic compositions is contemplated. Supplementary active ingredients also are incorporated into the compositions.
- compositions according to the present invention will be via any common route so long as the target tissue is available via that route.
- these compositions are formulated for oral administration, such as by an inhalant.
- other conventional routes of administration e.g., by subcutaneous, intravenous, intradermal, intramusclar, intramarnmary, intraperitoneal, intrathecal, intraocular, retrobulbar, intrapulmonary (e.g., term release), aerosol, sublingual, nasal, anal, vaginal, or transdermal delivery, or by surgical implantation at a particular site also is used particularly when oral administration is problematic.
- the treatment may consist of a single dose or a plurality of doses over a period of time.
- the active compounds are prepared for administration as solutions of free base or pharmacologically acceptable salts in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
- Dispersions also are prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
- the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid to the extent that easy syringability exists.
- the carrier is a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
- a coating such as lecithin
- surfactants for example, water, alcohol, glycol, and the like
- microorganisms The prevention of the action of microorganisms is brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions is brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
- agents delaying absorption for example, aluminum monostearate and gelatin.
- Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
- dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
- the methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
- pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
- the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients also are incorporated into the compositions.
- compositions of the present invention are formulated in a neutral or salt form.
- Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups also are derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
- solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
- the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like.
- parenteral administration in an aqueous solution for example, the solution is suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
- aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
- Unit dose is defined as a discrete amount of a therapeutic composition dispersed in a suitable carrier.
- parenteral administration of the therapeutic compounds is carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product.
- the frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration.
- the optimal pharmaceutical formulation will be determined by one of skill in the art depending on the route of administration and the desired dosage. Such formulations may influence the physical state, stability, rate of in vivo release and rate of in vivo clearance of the administered agents.
- a suitable dose is calculated according to body weight, body surface areas or organ size. The availability of animal models is particularly useful in facilitating a determination of appropriate dosages of a given therapeutic. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein as well as the pharmacokinetic data observed in animals or human clinical trials.
- appropriate dosages are ascertained through the use of established assays for determining blood levels in conjunction with relevant dose response data.
- the final dosage regimen will be determined by the attending physician, considering factors which modify the action of drugs, e.g., the drug's specific activity, severity of the damage and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. As studies are conducted, further information will emerge regarding appropriate dosage levels and duration of treatment for specific diseases and conditions. It will be appreciated that the pharmaceutical compositions and treatment methods of the invention are useful in fields of human medicine and veterinary medicine. Thus the subject to be treated is a mammal, such as a human or other mammalian animal.
- subjects include for example, farm animals including cows, sheep, pigs, horses and goats, companion animals such as dogs and cats, exotic and/or zoo animals, laboratory animals including mice rats, rabbits, guinea pigs and hamsters; and poultry such as chickens, turkey ducks and geese.
- kits for use in the treatment of various disorders include at least a first composition comprising the sulfatases described above in a pharmaceutically acceptable carrier. Another component is a second therapeutic agent for the treatment of the disorder along with suitable container and vehicles for administrations of the therapeutic compositions.
- the kits may additionally comprise solutions or buffers for effecting the delivery of the first and second compositions.
- the kits may further comprise catheters, syringes or other delivering devices for the delivery of one or more of the compositions used in the methods of the invention.
- the kits may further comprise instructions containing administration protocols for the therapeutic regimens.
- kits of the invention may comprise instructions for specific routes of administration e.g., intrathecal administration of the therapeutic compositions of the present invention, in addition to ' the therapeutic compositions.
- the kits of the invention may comprise catheters or other devices for the intrathecal administration of the enzyme replacement therapy that are preloaded with the therapeutic compositions of the present invention.
- catheters preloaded with 0.001 mg, 0.005 mg, 0.01 mg, 0.015 mg, 0.02 mg, 0.03 mg, 0.04 mg, 0.05 mg, 0.06 mg, 0.07 mg, 0.08 mg, 0.09 mg, 0.1 mg, 0.2 mg, 0.3 mg, 0.4 mg, 0.5 mg, 0.6 mg, 0.7 mg, 0.8 mg, 0.9 mg, or 1.0 mg or more of a given recombinant sulfatase of the present invention e.g., a sulfatase having a sequence of SEQ ID NO:27; SEQ E) NO:28; SEQ ID NO:28; SEQ ID NO:29; SEQ ID NO:30; SEQ ID NO:31; SEQ ID NO:32; SEQ ID NO:33; SEQ ID NO:34; SEQ ID NO:35; SEQ ID NO:36; SEQ TD NO:37; SEQ ID NO:38 or SEQ ID NO:39, or biologically active fragments or mutants thereof that have
- the preloaded catheters may be refillable and presented in kits that have appropriate amounts of the enzyme for refilling such catheters.
- Example 1 Materials and Methods ' Sulfatases, such as e.g., arylsulfatase B (ASB) and arylsulfatase A
- ASB arylsulfatase B
- A arylsulfatase A
- ASA ASA are subject to a unique post-translational modification that is obligate for their function.
- the modification reaction conversion of a cysteine to a formylglycine, becomes saturated when these enzymes are over-expressed.
- the present application • shows that the modification the mutants of ASB and ASA in which the modified cysteine is substituted with a serine are expressed much more efficiently than the native enzymes under identical conditions.
- the purified ASB mutant can then be converted to catalytically active ASB using vanadate and light, thereby obviating the need for the preparation of expression constructs and transfected cells that are transfected with both the sulfatase and a sulfatase processing enzyme, such as SUMFl (Cosma et al., Cell 113:445-456, 2003).
- SUMFl sulfatase processing enzyme
- the BP 15 rabbit polyclonal anti-ASB antibody and Gl 92 sheep polyclonal anti-ASB antibody were raised against native recombinant human enzymes.
- Rabbit polyclonal anti-ASA were obtained from UCLA. Antibodies were purified over protein G columns prior to use. PCR primers were synthesized by Qiagen. SybrGreen QPCR reagents were purchased from Roche Biochemicals. Highly purified recombinant human ASB was manufactured at BioMarin Pharmaceutical, Inc.
- Samples were diluted 1000-fold in assay buffer (50 mM NaOAc pH 5.6). Aliquots of the dilutions (5 ⁇ L) were combined with 150 ⁇ l of 5 mM 4-MU- sulfate substrate (4-MUS, Sigma) in assay buffer and incubated at 37°C for 20 minutes in black Costar flat-bottom plates. Reactions were terminated with 150 ⁇ l of 350 mM glycine, 440 mM sodium carbonate pH 10.7. Fluorescence was measured with a micro-plate fluorimeter (Molecular Devices) using an excitation wavelength of 366 nm and an emission wavelength of 446 nm. All samples were run in duplicate.
- assay buffer 50 mM NaOAc pH 5.6
- One unit of enzyme activity is defined as 1 ⁇ mole of 4-methylumbelliferone (4-MU) produced per minute at 37 0 C.
- 4-MU 4-methylumbelliferone
- a specific activity for ASB of 67 U/mg was used. Total protein was measured by Bradford assay (BioRad) using bovine serum albumin as the standard. .
- ASB native and mutant proteins were quantified by sandwich ELISA with the polyclonal BP15 antibody (ASB). Detecting antibody was conjugated to HRP. A standard curve was constructed with dilutions of purified ASB.
- ASA Western Blotting For Western blotting, proteins were separated by reducing SDS-PAGE, electroblotted to PVDF membranes, and blocked for 1 hour at room temperature with blocking buffer containing 2% (w/v) dry milk powder in Tris-buffered saline (TBS, 20 mM Tris-HCl, 150 mM NaCl, pH 7.5) with 0.05% (v/v) NP-40.
- TBS Tris-buffered saline
- ASA antigen was detected with anti-rhASA antiserum diluted 25, 000-fold into blocking buffer. Bound primary antibodies were then detected with affinity-purified alkaline phosphatase- conjugated goat secondary antibody (Promega) diluted 5, 000-fold in blocking buffer. Blots were developed with Western Blue colorimetric substrate (Promega), rinsed with distilled water and scanned.
- Protein concentrations for purified ASB and ASB C91S were calculated from A 28O values and theoretical extinction coefficients based on amino acid composition. The latter were obtained using ProtParam software. By this method of determination, both ASB and ASB C91 S had extinctions coefficients of 2 mLmg "1 cm "1 .
- PvNA was extracted using Qiagen RNeasyTM reagents and reverse transcribed with Superscript 11+ (Invitrogen) and an oligo (T)25VN primer using manufacturer's protocols. cDNA was assayed using a.Roche LightCyclerTM with SybrGreenTM reagents.
- a human ASB cDNA was amplified from human liver cDNA (BD Biosciences) by high-stringency PCR using Stratagene PfuTurboTM polymerase and the primers ASBFl: 5'-G CGATAGGTACCGCCATGGGTCCGCG CGGCGCGGCGAGC-3'(SEQIDNO:48)andASBR2: 5'-GCGATAC TCGAGCCCTGAAATCCTACATCCAAGGG -3' (SEQ ID NO:49).
- the cDNA was sequenced and found to be identical to that previously described.
- the cDNA was then reamplified with a different 5 '-primer to decrease the GC bias at the 5 '-end without changing the amino acid sequence (ASBatF: 5'-GCG ATAGGTACCGCCATGGGTCCTAGAGGAGCTGCTTCTT TGCCTCGAGGACCCGGACCTCGGCGGCTGCTCCTC-S') (SEQ ID NO: 50).
- the engineered cDNA was digested with Kpnl and Xhol and ligated into pcDNA3.1(+) (Invitrogen) to give pcDNA3.1-ASB.
- the 5 '-modified ASB was used to make both the native and C91S proteins.
- the pcDNA3.1-ASB plasmid was then cut with AatII and the vector portion recircularized in order to remove the 5' -182 base pairs of the ASB cDNA.
- the deleted vector was then cut with Pstl and the vector portion again recircularized in order to remove the 3'-760 base pairs of the cDNA.
- ASBmut3 5'- P G G CCGGCGGGGTGCTCCTGGACAACTACTACACGCAGCC GCTGAGCACGCCGTCGCGGAGCCAGCTGC (SEQ ID NO:51)
- ASBmut4 5'-P CAGCTG G CTCCG CG ACG G CG TG CTCAG CG G CTG CG TG TAG TAG TCCAG G AG CACCCCG C C -3 ') (SEQ ID NO:52).
- the mutation introduces an ApaLI site into the ASB coding sequence.
- the remaining, mutagenized, portion of ASB, flanked by AatII and Pstl sites was then used to reconstruct a full-length ASB C91S cDNA by ligation with Kpnl/Aatll and Pst/Xbal fragments from pcDNA3.1 (+)-ASB into pcDNA 3.1 (+).
- the reconstructed cDNA was sequenced and found to be identical to the published sequence except for the engineered changes described.
- a human ASA cDNA was amplified from human liver cDNA (BD Biosciences) by high-stringency PCR using Stratagene PfuTurboTM polymerase and the primers AryAFl: 5'-GCGATAGGTACCGCCATGGGGGCACC GCGGTCCCTCCTC -3' (SEQ TD NO:53) and AryAR: 5'-GCGATACT CGAGTCAGGCATGGGGATCTGGGCAATG-S' (SEQ ID NO: 54).
- the amplified cDNA was cut with Kpnl and Xhol for ligation into pcDNA3.1 (+).
- the cDNA was sequenced and found to be identical to that previously described.
- the ASA C69S was created in the same vector using the Promega Altered Sites IITM kit and a single mutagenic primer (ASAser 5' - P C G G CCGGTCAGGAGGGCGGCCCTAGAGGGTGTGCTCAGA GACACAGGCAC- 3') (SEQ ID NO:55).
- CHO-Kl cells were seeded at 2 x 10 5 cells/well in Corning Cell- WellTM 6-well polystyrene plates and allowed to grow overnight. Cells were transfected with Bio-Rad CytofecteneTM reagent using manufacturer protocols. Cells were selected in UltraCHO (Cambrex) medium supplemented with 2.5% dialyzed fetal bovine serum, 2 mM glutamine, 50 ⁇ g/mL gentamycin, 2.5 ⁇ g/mL amphotericin and either 450 or 900 ⁇ g/mL G418. Resistant colonies were evident within a week. All transfections yielded hundreds of colonies. Mock transfection dishes had no colonies. After 12 days, resistant pools of colonies for each of the construct types were pooled, expanded and frozen.
- Cells were trypsinized and counted with a Coulter counter. Cell pellets were washed with Dulbecco's phosphate-buffered saline (DPBS) and stored at -80°C. Cell lysates were prepared by subjecting DPBS-resuspended cell pellets to three cycles of freeze-thaw.
- DPBS Dulbecco's phosphate-buffered saline
- the ASB C91S sequence was cloned into a second vector, pBMPCINtl.
- the pBMPCINt 1-ASB C91S construct was linearized with Sail, gel purified and used to transfect CHO-S (Invitrogen) cells as described above. Cells were selected with 450 ⁇ g/mL G418. A total of 11 clones were picked and screened for expression of ASB by ELISA with the BP 15 antibody. Clone 7 had the highest volumetric productivity of ASB C91S (1 ⁇ g/mL) with an associated specific productivity under these conditions of 3 pg/cell/day. Clone 7 was used for production of ASB C91S.
- ASB C91S was purified on an anti-ASB (Gl 92) affinity column prepared by coupling the antibody to NHS-sepharose beads. Harvest medium was loaded onto the column. Unbound proteins were eluted with Dulbecco's phosphate- buffered saline. ASB C91S was eluted with 100 mM Glycine-HCl pH 2.25. Fractions were collected into tubes containing Tris-HCl, bringing the pH of the eluate immediately up to 6.5. The purified material from the affinity column was buffer exchanged and concentrated in 50 mM NaOAc pH 6.5 with a Millipore Centricon Plus-20 (8,000 NMWL). Purified protein was stored at 4 0 C. Photochemical reactions
- UVP Pen-Ray 5.5 Watt lamps 90- 0012-02 were used as light sources. The lamps were powered by a UVP
- Photochemical UV power supply 99-0055-01 and housed in an Ace Glass jacketed immersion well (7892-45). Lamp temperature was held at 20°C with a 1:1 mixture of FlagTM anti-freeze and tap water, which was pumped through the immersion well jacket at 16 liters per minute. Coolant temperature was maintained with a recirculating chiller. Samples in microfuge tubes were floated in a water bath in a 500 mL jacketed beaker. The beaker temperature was controlled with a second recirculating chiller. The lamp was positioned immediately below the samples. UVB radiation was removed with the anti-freeze mixture (which absorbs strongly at 280 nm) and with polystyrene plates.
- Irradiation dose was measured with an Oriel GoldiluxTM Smartmeter Photometer using calibrated UVA (365 nm) and UVB (280 nm) probes. All samples were irradiated in 1.5 mL polypropylene microfuge tubes (USA Scientific 1415-2600) or Corning clear polystyrene 96-well plates.
- a 10 ⁇ M solution of affinity-purified ASB C91S or ASB in 50 mM sodium acetate pH 5.6, 150 mM NaCl was diluted 10-fold into 100 mM Tris-acetate pH 8.0. Protein solutions were then supplemented with 10 ⁇ M or 500 ⁇ M sodium metavanadate from a 100 mM stock solution (prepared as described above).
- the ASB C91S solution was then placed in a 1 cm-path length quartz cuvette and irradiated at a light intensity of 3.7 mW/cm 2 measured at 350 nm. Excitation wavelengths were restricted to the 250- 450 nm range using appropriate cut-off filters. Irradiation was performed for intervals from 10 to 240 seconds after which samples were placed on ice.
- Wild-type ASB (0.2 ⁇ g/mL, 200 ⁇ L) was combined with 10 niM vanadate in 50 niM sodium acetate pH 5.6, 150 mM NaCl.
- the protein solution was irradiated for 2 minutes with 250-450 nm light at an intensity of 3.7 mW/cm 2 . Total inactivation was confirmed by activity assay as described above. Protein was precipitated by the addition of 470 ⁇ L of acetone and pelleted by centrifugation. The protein pellet was then washed with acetone and allowed to air-dry prior to resuspension in 10 ⁇ L of 8 M urea.
- the resolubilized protein (10 ⁇ L) was then combined with an equal volume of 0.5 M Tris-HCl pH 8.2, 1 ⁇ L of 0.1 M CaCl 2 , 74 ⁇ L of water, and 5 ⁇ L of trypsin (0.5 ⁇ g/ ⁇ L). Digestion was allowed to proceed at room temperature for 16 hours. Disulfide bonds were reduced by the addition of DTT to 2 mM and incubation for an additional 30 minutes at 37°C. All reactions were quenched by the addition of 2 ⁇ L formic acid.
- Digested peptides were resolved and analyzed with a Hewlett-Packard 1100 LC/MS with an Agilent 1100 series LC/electrospray MSD (Phenomenex column C18 RP; P/N 00G-4396-B0; 250 x 2mm).
- Example 2 Expression of Recombinant proteins in CHO cells
- the present Example provides details of CHO cell production, transcript levels, protein productivity and protein processing efficiency. The methods described in Example 1 were used to generate the results described herein.
- CHO cells expressing ASB 5 ASB C91 S, ASA and ASA C69S were created in order to ascertain the level to which sulfatase conversion affects expression.
- AU sequences were expressed in identical vectors. All vectors contained the neomycin phosphotransferase gene (NPT) in the same expression cassette to allow selection on G418. Complete native and mutant expression vectors differed by a single base pair.
- Cells in replicate 6-well plates were transfected at the same time with identical amounts of different plasmids using the same reagents. Transfected cells were selected at 450 and at 900 ⁇ g/mL G418 with the thought that the latter level would enforce the requirement for high expression of NPT and the immediately adjacent sulfatase.
- the data indicate at least three differences in the expression behavior of the native and serine mutant sulfatases: Serine mutant protein is expressed at higher levels than native enzyme on both cell and transcript normalized bases; overexpression of native enzyme appears to be toxic in a way that serine mutant expression is not; and, in a possibly related observation, serine mutant transcript appears to be converted to secreted protein more efficiently than native transcript. Protein productivity
- NPT transcript levels increased with increasing levels G418 selection. More surprising was the observation that native ASB and ASA transcript levels in the same pools did not increase (Figure 1, Panel C). In agreement with the levels of secreted protein, transcript levels for the native enzymes actually went down in cells surviving at 900 ⁇ g/mL G418. This result suggests that transcription of the native sulfatases and transcription of NPT have become uncoupled in cells surviving at higher G418 concentrations. That uncoupling would be required for survival is indicative, again, of some kind of toxicity associated with expression of native ASB and ASA. In contrast to the native transcripts, ASB C91S and ASA C69S transcript levels increased in parallel with NPT from the 450 to the 900 ⁇ g/mL G418 level. If it is toxicity that constrains increases in native enzyme expression with increasing G418 selection, then serine mutant expression does not seem to result in the same toxicity.
- the data from the pool experiments is consistent with enhanced expression of serine-mutant sulfatases relative to native sulfatases.
- Reduced productivity of the native enzymes may be a result of toxicity and decreased protein- processing efficiency. It is likely that the former is a result of the latter, whereby decreases in protein processing efficiency leads to stresses on the cell that compromise the ability to survive under strong antibiotic selection.
- Example 3 A vanadate-dependent photoreaction to produce activated sulfatases
- the present example demonstrates this phenomenon using ASB C91S.
- Affinity-purified ASB C91S was irradiated with near UV light in the presence of 2, 20, 40 and 400 ⁇ M sodium metavanadate in 50 mM NaOAc pH 6.5, 100 mM NaCl. Light below 300 nm was removed by placing appropriate absorptive materials between the light source and the samples (see Example 1). Samples were subjected to cycles of irradiation consisting of 30 minutes of light followed by 20 minutes in the dark on ice. Treated samples were then assayed for activity against the 4-MUS after dilution of the vanadate to concentrations below those necessary to inhibit the sulfatase.
- ASB C91 S was converted to catalytically active enzyme by the above- described procedure. Maximal activation was achieved at the highest vanadate concentration (Figure 3A). No activation was observed by irradiation in the absence of vanadate or with vanadate in the absence of irradiation. Increases in the amount of activated enzyme were observed through the last time point. Activation was reproducible over a number of experiments. Assuming that the entire mutant enzyme population in the sample was activatible, conversion efficiencies ranged from 20- 30%.
- the present invention for the first time demonstrates that there may be an association between vanadate and a serine side-chain that leads to light-catalyzed two-electron transfer from the serine C ⁇ to vanadium. This reaction would explain the observed serine- mutant sulfatase conversion.
- the oxovanadium (V +3 ) generated as a by-product would be expected to corn-proportionate in presence of vanadate (V +5 ) to form two moles of vanadyl (V +4 ). Vanadyl slowly oxidizes back to vanadate in the presence of oxygen.
- a second vanadate-dependent photo-oxidation step can then occur leading to addition of molecular oxygen to the backbone Ca and eventual cleavage of the myosin peptide chain.
- Enolization of the aldehyde in the context of the sulfatase active site is less likely since the protein is expected to strongly stabilize the hydrated keto form of the side-chain.
- the present example clearly demonstrates the vanadate dependent phtoreactive activation of ASB.
- the active site of sulfatase enzymes is highly conserved among both eukaryotic and prokaryotic sulfatases. Therefore, it is predicted that light-dependent oxidation of serine mutants will be generally applicable to all members of the sulfatase family and will therefore be useful in the activation of prokaryotic sulfatases as well as eukaryotic sulfatases.
- compositions and/or methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the compositions and methods of this invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the compositions and/or methods and in the steps or in the sequence of steps of the method described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.
Landscapes
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Zoology (AREA)
- Wood Science & Technology (AREA)
- Molecular Biology (AREA)
- Microbiology (AREA)
- Biotechnology (AREA)
- Biomedical Technology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Enzymes And Modification Thereof (AREA)
Abstract
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US56854404P | 2004-05-06 | 2004-05-06 | |
US60/568,544 | 2004-05-06 |
Publications (2)
Publication Number | Publication Date |
---|---|
WO2005113765A2 true WO2005113765A2 (fr) | 2005-12-01 |
WO2005113765A3 WO2005113765A3 (fr) | 2006-01-12 |
Family
ID=35428930
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2005/014097 WO2005113765A2 (fr) | 2004-05-06 | 2005-04-25 | Procedes d'activation de sulfatases, methodes et compositions d'utilisation associees |
Country Status (1)
Country | Link |
---|---|
WO (1) | WO2005113765A2 (fr) |
Cited By (14)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
EP2064334A4 (fr) * | 2006-09-21 | 2010-05-05 | Univ California | Marqueurs d'aldéhyde et utilisations de ceux-ci dans le cadre d'une modification de protéines en un site spécifique |
US7722865B2 (en) | 2008-01-18 | 2010-05-25 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US20120180148A1 (en) * | 2011-01-07 | 2012-07-12 | Jin Thong-Gyu | Transgenic mouse model for developing enzyme replacement therapy for iduronate-2-sulfatase deficiency syndrome |
WO2013148277A1 (fr) | 2012-03-30 | 2013-10-03 | Shire Human Genetic Therapies, Inc. | Administration sous-cutanée d'iduronate 2-sulfatase |
WO2013148323A1 (fr) * | 2012-03-30 | 2013-10-03 | Shire Human Genetic Therapies | Procédés d'analyse et de préparation de compositions protéiques |
WO2014005036A1 (fr) | 2012-06-29 | 2014-01-03 | Shire Human Genetic Therapies, Inc. | Méthode de production de l'iduronate-2-sulfatase recombinée |
WO2014005019A3 (fr) * | 2012-06-29 | 2014-02-27 | Shire Human Genetic Therapies, Inc. | Cellules pour l'obtention d'iduronate-2-sulfatase recombinante |
US8729232B2 (en) | 2008-03-27 | 2014-05-20 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
EP2723369A4 (fr) * | 2011-06-24 | 2014-12-24 | Green Cross Corp | Composition et formulation comprenant de l'iduronate-2-sulfatase humaine recombinée et procédé pour la préparer |
US9051556B2 (en) | 2012-06-29 | 2015-06-09 | Shire Human Genetic Therapies, Inc. | Purification of iduronate-2-sulfatase |
US9200264B2 (en) | 2008-01-18 | 2015-12-01 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US9896672B2 (en) | 2011-06-24 | 2018-02-20 | Green Cross Corporation | Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof |
US11208632B2 (en) | 2016-04-26 | 2021-12-28 | R.P. Scherer Technologies, Llc | Antibody conjugates and methods of making and using the same |
US12102689B2 (en) | 2015-11-09 | 2024-10-01 | R.P. Scherer Technologies, Llc | Anti-CD22 antibody-maytansine conjugates and methods of use thereof |
-
2005
- 2005-04-25 WO PCT/US2005/014097 patent/WO2005113765A2/fr active Application Filing
Non-Patent Citations (2)
Title |
---|
CHRISTIANSON T. ET AL.: 'Overexpression of inactive arysulphatase mutants and in vitro activation by light-dependent oxidation with vanadate.' BIOCHEM. J. vol. 382, September 2004, pages 581 - 587, XP002992392 * |
ZALK P. ET AL.: 'ATP-binding sites in brain p97/VCP (valosin-containing protein), a multifunctional AAA ATPase.' BIOCHEM J. vol. 374, May 2003, pages 473 - 480, XP002992393 * |
Cited By (48)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US10745464B2 (en) | 2006-09-21 | 2020-08-18 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
EP2064334A4 (fr) * | 2006-09-21 | 2010-05-05 | Univ California | Marqueurs d'aldéhyde et utilisations de ceux-ci dans le cadre d'une modification de protéines en un site spécifique |
US7985783B2 (en) | 2006-09-21 | 2011-07-26 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US8097701B2 (en) | 2006-09-21 | 2012-01-17 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US9447390B2 (en) | 2006-09-21 | 2016-09-20 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US8349910B2 (en) | 2006-09-21 | 2013-01-08 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
EP2548967A3 (fr) * | 2006-09-21 | 2013-03-06 | The Regents of The University of California | Marqueurs d'aldéhyde et leurs utilisations dans le cadre d'une modification de protéines en un site spécifique |
US8846866B2 (en) | 2006-09-21 | 2014-09-30 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US10150806B2 (en) | 2006-09-21 | 2018-12-11 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US9873867B2 (en) | 2008-01-18 | 2018-01-23 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US7722865B2 (en) | 2008-01-18 | 2010-05-25 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US10934534B2 (en) | 2008-01-18 | 2021-03-02 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US10563183B2 (en) | 2008-01-18 | 2020-02-18 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
EP4413994A3 (fr) * | 2008-01-18 | 2024-12-18 | BioMarin Pharmaceutical Inc. | Fabrication d'enzymes de sulfatase lysosomales humaines hautement phosphorylées actives et leurs utilisations |
US12188058B2 (en) | 2008-01-18 | 2025-01-07 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
EP3219795A1 (fr) * | 2008-01-18 | 2017-09-20 | BioMarin Pharmaceutical Inc. | Fabrication d'enzymes de sulfatase lysosomales humaines actives hautement phosphorylées et leurs utilisations |
EP2245145B1 (fr) * | 2008-01-18 | 2016-12-14 | BioMarin Pharmaceutical Inc. | Fabrication d'enzymes de sulfatase lysosomales humaines hautement phosphorylées actives et leurs utilisations |
US9200264B2 (en) | 2008-01-18 | 2015-12-01 | Biomarin Pharmaceutical Inc. | Manufacture of active highly phosphorylated human lysosomal sulfatase enzymes and uses thereof |
US8729232B2 (en) | 2008-03-27 | 2014-05-20 | The Regents Of The University Of California | Aldehyde tags, uses thereof in site-specific protein modification |
US20140351964A1 (en) * | 2011-01-07 | 2014-11-27 | Medigenebio Corporation | Transgenic mouse model for developing enzyme replacement therapy for iduronate-2-sulfatase deficiency syndrome |
US20120180148A1 (en) * | 2011-01-07 | 2012-07-12 | Jin Thong-Gyu | Transgenic mouse model for developing enzyme replacement therapy for iduronate-2-sulfatase deficiency syndrome |
EP2723369B1 (fr) | 2011-06-24 | 2016-04-13 | Green Cross Corporation | Composition et formulation comprenant de l'iduronate-2-sulfatase humaine recombinée et procédé pour la préparer |
US9896672B2 (en) | 2011-06-24 | 2018-02-20 | Green Cross Corporation | Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof |
US9249397B2 (en) | 2011-06-24 | 2016-02-02 | Green Cross Corporation | Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof |
EP2723369A4 (fr) * | 2011-06-24 | 2014-12-24 | Green Cross Corp | Composition et formulation comprenant de l'iduronate-2-sulfatase humaine recombinée et procédé pour la préparer |
US9206402B2 (en) | 2011-06-24 | 2015-12-08 | Green Cross Corporation | Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof |
US10174299B2 (en) | 2011-06-24 | 2019-01-08 | Green Cross Corporation | Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof |
EP2830642A4 (fr) * | 2012-03-30 | 2015-12-02 | Shire Human Genetic Therapies | Administration sous-cutanée d'iduronate 2-sulfatase |
WO2013148277A1 (fr) | 2012-03-30 | 2013-10-03 | Shire Human Genetic Therapies, Inc. | Administration sous-cutanée d'iduronate 2-sulfatase |
WO2013148323A1 (fr) * | 2012-03-30 | 2013-10-03 | Shire Human Genetic Therapies | Procédés d'analyse et de préparation de compositions protéiques |
US9603908B2 (en) | 2012-03-30 | 2017-03-28 | Shire Human Genetic Therapies, Inc. | Subcutaneous administration of iduronate-2-sulfatase |
KR101710487B1 (ko) | 2012-06-29 | 2017-02-27 | 샤이어 휴먼 지네틱 테라피즈 인크. | 재조합 이듀로네이트-2-설파타제를 생산하는 세포 |
WO2014005019A3 (fr) * | 2012-06-29 | 2014-02-27 | Shire Human Genetic Therapies, Inc. | Cellules pour l'obtention d'iduronate-2-sulfatase recombinante |
CN104540517A (zh) * | 2012-06-29 | 2015-04-22 | 夏尔人类遗传性治疗公司 | 用于产生重组艾杜糖-2-硫酸酯酶的细胞 |
CN104583414A (zh) * | 2012-06-29 | 2015-04-29 | 夏尔人类遗传性治疗公司 | 产生重组艾杜糖-2-硫酸酯酶的方法 |
KR20150030242A (ko) * | 2012-06-29 | 2015-03-19 | 샤이어 휴먼 지네틱 테라피즈 인크. | 재조합 이듀로네이트-2-설파타제를 생산하는 세포 |
US9719074B2 (en) | 2012-06-29 | 2017-08-01 | Shire Human Genetic Therapies, Inc. | Cells for producing recombinant iduronate-2-sulfatase |
US10344270B2 (en) | 2012-06-29 | 2019-07-09 | Shire Human Genetic Therapies, Inc. | Methods and compositions for treatment of Hunter syndrome |
US9051556B2 (en) | 2012-06-29 | 2015-06-09 | Shire Human Genetic Therapies, Inc. | Purification of iduronate-2-sulfatase |
EA035511B1 (ru) * | 2012-06-29 | 2020-06-26 | Шир Хьюман Дженетик Терапис, Инк. | Клетка и способ для получения рекомбинантной идуронат-2-сульфатазы |
CN107267460A (zh) * | 2012-06-29 | 2017-10-20 | 夏尔人类遗传性治疗公司 | 用于产生重组艾杜糖‑2‑硫酸酯酶的细胞 |
WO2014005036A1 (fr) | 2012-06-29 | 2014-01-03 | Shire Human Genetic Therapies, Inc. | Méthode de production de l'iduronate-2-sulfatase recombinée |
US9150841B2 (en) | 2012-06-29 | 2015-10-06 | Shire Human Genetic Therapies, Inc. | Cells for producing recombinant iduronate-2-sulfatase |
US11530393B2 (en) | 2012-06-29 | 2022-12-20 | Takeda Pharmaceutical Company Limited | Compositions comprising iduronate-2-sulfatase |
US9492511B2 (en) | 2012-06-29 | 2016-11-15 | Shire Human Genetic Therapies, Inc. | Methods and compositions for treatment of hunter syndrome |
US12102689B2 (en) | 2015-11-09 | 2024-10-01 | R.P. Scherer Technologies, Llc | Anti-CD22 antibody-maytansine conjugates and methods of use thereof |
US11788066B2 (en) | 2016-04-26 | 2023-10-17 | R.P. Scherer Technologies, Llc | Antibody conjugates and methods of making and using the same |
US11208632B2 (en) | 2016-04-26 | 2021-12-28 | R.P. Scherer Technologies, Llc | Antibody conjugates and methods of making and using the same |
Also Published As
Publication number | Publication date |
---|---|
WO2005113765A3 (fr) | 2006-01-12 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US7041487B2 (en) | Recombinant α-L-iduronidase, methods for producing and purifying the same and methods for treating diseases caused by deficiencies thereof | |
RU2179034C2 (ru) | ЛЕЧЕНИЕ В СЛУЧАЕ ДЕФИЦИТА α-ГАЛАКТОЗИДАЗЫ А | |
US7122354B2 (en) | Nucleic acid encoding a chimeric polypeptide | |
CN103635203B (zh) | 包含重组人艾杜糖醛酸-2-硫酸酯酶的组合物和制剂及其制备方法 | |
WO2005113765A2 (fr) | Procedes d'activation de sulfatases, methodes et compositions d'utilisation associees | |
KR101535791B1 (ko) | 개량형 이듀로네이트-2-설파타제 및 이의 용도 | |
CN104711243B (zh) | 重组的弹性蛋白酶蛋白质及其制备方法和用途 | |
CN107949638A (zh) | 新型人血清白蛋白突变体 | |
CN1300323A (zh) | 重组α-L-艾杜糖苷酸酶,其生产和纯化的方法以及治疗其缺乏导致的疾病的方法 | |
EA022220B1 (ru) | Способ получения и очистки рекомбинантной лизосомальной альфа-маннозидазы | |
CN104955946A (zh) | 靶向的艾杜糖醛酸-2-硫酸酯酶化合物 | |
ES2445028T3 (es) | N-acetilgalactosamina-4-sulfatasa precursora, métodos de tratamiento usando dicha enzima y métodos para producir y purificar dicha enzima | |
JP2023519934A (ja) | ゴーシェ病の処置において使用するためのベータ-グルコセレブロシダーゼのバリアント | |
CA2443555C (fr) | Enzymes therapeutiques, et methodes de traitement du mps-vi et de lignees de cellules en vue de l'obtention de ces enzymes par recombinaison | |
AU2001255771A1 (en) | Enzymes useful for treating and methods for treating MPS-VI and cell lines for producing such enzymes recombinantly | |
AU2003220717B2 (en) | Therapy for alpha-galactosidase a deficiency | |
ES2824623T3 (es) | Proceso para la producción y purificación de alfa-manosidasa lisosómica recombinante | |
AU2008200265B2 (en) | Therapy for alpha-galactosidase A deficiency | |
TW202122409A (zh) | 人類重組精胺酸酶1之產生方法及其用途 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
AK | Designated states |
Kind code of ref document: A2 Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW |
|
AL | Designated countries for regional patents |
Kind code of ref document: A2 Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG |
|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
NENP | Non-entry into the national phase in: |
Ref country code: DE |
|
WWW | Wipo information: withdrawn in national office |
Country of ref document: DE |
|
122 | Ep: pct application non-entry in european phase |