NZ731196B2 - A novel purification process for isolation and commercial production of recombinant tnk-tpa (tenecteplase) - Google Patents
A novel purification process for isolation and commercial production of recombinant tnk-tpa (tenecteplase) Download PDFInfo
- Publication number
- NZ731196B2 NZ731196B2 NZ731196A NZ73119615A NZ731196B2 NZ 731196 B2 NZ731196 B2 NZ 731196B2 NZ 731196 A NZ731196 A NZ 731196A NZ 73119615 A NZ73119615 A NZ 73119615A NZ 731196 B2 NZ731196 B2 NZ 731196B2
- Authority
- NZ
- New Zealand
- Prior art keywords
- range
- tpa
- tnk
- chromatography
- present
- Prior art date
Links
- 238000000746 purification Methods 0.000 title claims abstract description 41
- 238000004519 manufacturing process Methods 0.000 title description 5
- 108010039185 Tenecteplase Proteins 0.000 title description 4
- 229960000216 Tenecteplase Drugs 0.000 title description 4
- 238000002955 isolation Methods 0.000 title description 4
- 238000000034 method Methods 0.000 claims abstract description 56
- 238000001914 filtration Methods 0.000 claims abstract description 13
- 125000002485 formyl group Chemical group [H]C(*)=O 0.000 claims abstract 2
- 230000003612 virological Effects 0.000 claims description 46
- FAPWRFPIFSIZLT-UHFFFAOYSA-M sodium chloride Chemical compound [Na+].[Cl-] FAPWRFPIFSIZLT-UHFFFAOYSA-M 0.000 claims description 44
- XSQUKJJJFZCRTK-UHFFFAOYSA-N urea Chemical compound NC(N)=O XSQUKJJJFZCRTK-UHFFFAOYSA-N 0.000 claims description 44
- UIIMBOGNXHQVGW-UHFFFAOYSA-M buffer Substances [Na+].OC([O-])=O UIIMBOGNXHQVGW-UHFFFAOYSA-M 0.000 claims description 31
- 238000004587 chromatography analysis Methods 0.000 claims description 31
- 239000000203 mixture Substances 0.000 claims description 28
- 239000011780 sodium chloride Substances 0.000 claims description 28
- 230000002779 inactivation Effects 0.000 claims description 27
- 241000700605 Viruses Species 0.000 claims description 23
- 239000004202 carbamide Substances 0.000 claims description 22
- 238000005277 cation exchange chromatography Methods 0.000 claims description 21
- 102000004169 proteins and genes Human genes 0.000 claims description 20
- 108090000623 proteins and genes Proteins 0.000 claims description 20
- 238000011068 load Methods 0.000 claims description 18
- 235000018102 proteins Nutrition 0.000 claims description 17
- 230000005526 G1 to G0 transition Effects 0.000 claims description 15
- ODKSFYDXXFIFQN-BYPYZUCNSA-N L-arginine Chemical compound OC(=O)[C@@H](N)CCCN=C(N)N ODKSFYDXXFIFQN-BYPYZUCNSA-N 0.000 claims description 14
- 229920001213 Polysorbate 20 Polymers 0.000 claims description 12
- 229940068977 Polysorbate 20 Drugs 0.000 claims description 12
- 229920002684 Sepharose Polymers 0.000 claims description 12
- 239000000256 polyoxyethylene sorbitan monolaurate Substances 0.000 claims description 12
- 235000010486 polyoxyethylene sorbitan monolaurate Nutrition 0.000 claims description 12
- 238000002415 sodium dodecyl sulfate polyacrylamide gel electrophoresis Methods 0.000 claims description 12
- 229960005480 Sodium caprylate Drugs 0.000 claims description 11
- 241000701093 Suid alphaherpesvirus 1 Species 0.000 claims description 11
- 150000001768 cations Chemical class 0.000 claims description 11
- BYKRNSHANADUFY-UHFFFAOYSA-M sodium;octanoate Chemical compound [Na+].CCCCCCCC([O-])=O BYKRNSHANADUFY-UHFFFAOYSA-M 0.000 claims description 11
- 239000000126 substance Substances 0.000 claims description 11
- 239000004472 Lysine Substances 0.000 claims description 10
- 238000003306 harvesting Methods 0.000 claims description 10
- 238000002360 preparation method Methods 0.000 claims description 10
- 238000000108 ultra-filtration Methods 0.000 claims description 10
- 238000010828 elution Methods 0.000 claims description 9
- 239000000919 ceramic Substances 0.000 claims description 8
- 229910052588 hydroxylapatite Inorganic materials 0.000 claims description 8
- 150000002500 ions Chemical class 0.000 claims description 8
- 239000012528 membrane Substances 0.000 claims description 8
- 239000008363 phosphate buffer Substances 0.000 claims description 8
- NBIIXXVUZAFLBC-UHFFFAOYSA-N phosphoric acid Chemical compound OP(O)(O)=O NBIIXXVUZAFLBC-UHFFFAOYSA-N 0.000 claims description 8
- 230000037250 Clearance Effects 0.000 claims description 7
- 235000014852 L-arginine Nutrition 0.000 claims description 7
- 230000035512 clearance Effects 0.000 claims description 7
- 238000001542 size-exclusion chromatography Methods 0.000 claims description 7
- 239000001488 sodium phosphate Substances 0.000 claims description 7
- 229910000162 sodium phosphate Inorganic materials 0.000 claims description 7
- RYFMWSXOAZQYPI-UHFFFAOYSA-K trisodium phosphate Chemical compound [Na+].[Na+].[Na+].[O-]P([O-])([O-])=O RYFMWSXOAZQYPI-UHFFFAOYSA-K 0.000 claims description 7
- 241000713893 Xenotropic murine leukemia virus Species 0.000 claims description 6
- 238000004113 cell culture Methods 0.000 claims description 6
- 229920003013 deoxyribonucleic acid Polymers 0.000 claims description 6
- 150000003839 salts Chemical class 0.000 claims description 6
- AWLUSOLTCFEHNE-UHFFFAOYSA-N sodium;urea Chemical compound [Na].NC(N)=O AWLUSOLTCFEHNE-UHFFFAOYSA-N 0.000 claims description 6
- 230000003750 conditioning Effects 0.000 claims description 5
- 239000012465 retentate Substances 0.000 claims description 5
- VMHLLURERBWHNL-UHFFFAOYSA-M sodium acetate Chemical compound [Na+].CC([O-])=O VMHLLURERBWHNL-UHFFFAOYSA-M 0.000 claims description 5
- 239000001632 sodium acetate Substances 0.000 claims description 5
- 235000017281 sodium acetate Nutrition 0.000 claims description 5
- 238000011100 viral filtration Methods 0.000 claims description 5
- 239000002253 acid Substances 0.000 claims description 4
- SLXKOJJOQWFEFD-UHFFFAOYSA-N Aminocaproic acid Chemical compound NCCCCCC(O)=O SLXKOJJOQWFEFD-UHFFFAOYSA-N 0.000 claims description 3
- 239000000178 monomer Substances 0.000 claims description 2
- 239000007974 sodium acetate buffer Substances 0.000 claims description 2
- 241000495102 Maize mosaic nucleorhabdovirus Species 0.000 claims 3
- 208000009305 Pseudorabies Diseases 0.000 claims 1
- 239000012141 concentrate Substances 0.000 claims 1
- 239000012064 sodium phosphate buffer Substances 0.000 claims 1
- LSNNMFCWUKXFEE-UHFFFAOYSA-N sulfonic acid Chemical compound OS(O)=O LSNNMFCWUKXFEE-UHFFFAOYSA-N 0.000 claims 1
- 238000001042 affinity chromatography Methods 0.000 abstract description 28
- 108090000373 Tissue plasminogen activator Proteins 0.000 abstract description 6
- 102000003978 Tissue plasminogen activator Human genes 0.000 abstract description 6
- 229960000187 tissue plasminogen activator Drugs 0.000 abstract description 5
- 210000004027 cells Anatomy 0.000 description 26
- 235000002639 sodium chloride Nutrition 0.000 description 20
- 239000012149 elution buffer Substances 0.000 description 18
- 239000000523 sample Substances 0.000 description 18
- 229940045136 Urea Drugs 0.000 description 16
- 229960000539 carbamide Drugs 0.000 description 16
- 238000009295 crossflow filtration Methods 0.000 description 13
- 239000012535 impurity Substances 0.000 description 12
- 239000000047 product Substances 0.000 description 10
- 238000004458 analytical method Methods 0.000 description 8
- 230000002829 reduced Effects 0.000 description 8
- 239000008186 active pharmaceutical agent Substances 0.000 description 7
- 239000006167 equilibration buffer Substances 0.000 description 7
- 239000004475 Arginine Substances 0.000 description 6
- 241000702623 Minute virus of mice Species 0.000 description 6
- 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 5
- 239000002033 PVDF binder Substances 0.000 description 5
- 235000009697 arginine Nutrition 0.000 description 5
- 239000003814 drug Substances 0.000 description 5
- 229940079593 drugs Drugs 0.000 description 5
- 239000007788 liquid Substances 0.000 description 5
- 229960000485 methotrexate Drugs 0.000 description 5
- 229920002981 polyvinylidene fluoride Polymers 0.000 description 5
- BQCADISMDOOEFD-UHFFFAOYSA-N silver Chemical compound [Ag] BQCADISMDOOEFD-UHFFFAOYSA-N 0.000 description 5
- 229910052709 silver Inorganic materials 0.000 description 5
- 239000004332 silver Substances 0.000 description 5
- KEAYESYHFKHZAL-UHFFFAOYSA-N sodium Chemical compound [Na] KEAYESYHFKHZAL-UHFFFAOYSA-N 0.000 description 5
- 229910052708 sodium Inorganic materials 0.000 description 5
- 239000011734 sodium Substances 0.000 description 5
- 206010061256 Ischaemic stroke Diseases 0.000 description 4
- 230000001154 acute Effects 0.000 description 4
- 239000012504 chromatography matrix Substances 0.000 description 4
- 230000009089 cytolysis Effects 0.000 description 4
- 238000004128 high performance liquid chromatography Methods 0.000 description 4
- 230000002934 lysing Effects 0.000 description 4
- 239000000463 material Substances 0.000 description 4
- 238000011069 regeneration method Methods 0.000 description 4
- SXGZJKUKBWWHRA-UHFFFAOYSA-N 2-(N-morpholino)ethanesulfonic acid Chemical compound [O-]S(=O)(=O)CC[NH+]1CCOCC1 SXGZJKUKBWWHRA-UHFFFAOYSA-N 0.000 description 3
- 206010000891 Acute myocardial infarction Diseases 0.000 description 3
- 238000002965 ELISA Methods 0.000 description 3
- 229950003499 FIBRIN Drugs 0.000 description 3
- BWGVNKXGVNDBDI-UHFFFAOYSA-N Fibrin Chemical compound CNC(=O)CNC(=O)CN BWGVNKXGVNDBDI-UHFFFAOYSA-N 0.000 description 3
- 102000009123 Fibrin Human genes 0.000 description 3
- 108010073385 Fibrin Proteins 0.000 description 3
- 239000012190 activator Substances 0.000 description 3
- 235000001014 amino acid Nutrition 0.000 description 3
- 238000004166 bioassay Methods 0.000 description 3
- 238000005341 cation exchange Methods 0.000 description 3
- 239000001913 cellulose Substances 0.000 description 3
- 229920002678 cellulose Polymers 0.000 description 3
- 230000003247 decreasing Effects 0.000 description 3
- 239000003599 detergent Substances 0.000 description 3
- 238000011026 diafiltration Methods 0.000 description 3
- -1 diethylamino ethyl Chemical group 0.000 description 3
- 238000010790 dilution Methods 0.000 description 3
- 238000004255 ion exchange chromatography Methods 0.000 description 3
- 238000002156 mixing Methods 0.000 description 3
- 230000001264 neutralization Effects 0.000 description 3
- 210000001519 tissues Anatomy 0.000 description 3
- 229940019746 Antifibrinolytic amino acids Drugs 0.000 description 2
- 229960001230 Asparagine Drugs 0.000 description 2
- 241000699802 Cricetulus griseus Species 0.000 description 2
- 241000272780 Cygnus Species 0.000 description 2
- 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 2
- DCXYFEDJOCDNAF-REOHCLBHSA-N L-asparagine Chemical compound OC(=O)[C@@H](N)CC(N)=O DCXYFEDJOCDNAF-REOHCLBHSA-N 0.000 description 2
- AYFVYJQAPQTCCC-GBXIJSLDSA-N L-threonine Chemical compound C[C@@H](O)[C@H](N)C(O)=O AYFVYJQAPQTCCC-GBXIJSLDSA-N 0.000 description 2
- 210000001672 Ovary Anatomy 0.000 description 2
- 229940012957 Plasmin Drugs 0.000 description 2
- 102000013566 Plasminogen Human genes 0.000 description 2
- 108010051456 Plasminogen Proteins 0.000 description 2
- 239000004473 Threonine Substances 0.000 description 2
- 230000002378 acidificating Effects 0.000 description 2
- 229960002684 aminocaproic acid Drugs 0.000 description 2
- 229910052921 ammonium sulfate Inorganic materials 0.000 description 2
- 239000001166 ammonium sulphate Substances 0.000 description 2
- 235000011130 ammonium sulphate Nutrition 0.000 description 2
- 108090001123 antibodies Proteins 0.000 description 2
- 102000004965 antibodies Human genes 0.000 description 2
- 235000009582 asparagine Nutrition 0.000 description 2
- 239000003795 chemical substances by application Substances 0.000 description 2
- 239000012501 chromatography media Substances 0.000 description 2
- 238000011143 downstream manufacturing Methods 0.000 description 2
- LFQSCWFLJHTTHZ-UHFFFAOYSA-N ethanol Chemical compound CCO LFQSCWFLJHTTHZ-UHFFFAOYSA-N 0.000 description 2
- 239000000706 filtrate Substances 0.000 description 2
- 238000009472 formulation Methods 0.000 description 2
- 239000012537 formulation buffer Substances 0.000 description 2
- 238000001641 gel filtration chromatography Methods 0.000 description 2
- XKUUMWKWUZRRPD-UHFFFAOYSA-N heptan-2-amine;sulfuric acid Chemical compound [O-]S([O-])(=O)=O.CCCCCC(C)[NH3+].CCCCCC(C)[NH3+] XKUUMWKWUZRRPD-UHFFFAOYSA-N 0.000 description 2
- 238000004191 hydrophobic interaction chromatography Methods 0.000 description 2
- 238000005342 ion exchange Methods 0.000 description 2
- KFZMGEQAYNKOFK-UHFFFAOYSA-N iso-propanol Chemical compound CC(C)O KFZMGEQAYNKOFK-UHFFFAOYSA-N 0.000 description 2
- ZOTBXTZVPHCKPN-HTXNQAPBSA-N kresoxim-methyl Chemical compound CO\N=C(\C(=O)OC)C1=CC=CC=C1COC1=CC=CC=C1C ZOTBXTZVPHCKPN-HTXNQAPBSA-N 0.000 description 2
- 125000003588 lysine group Chemical class [H]N([H])C([H])([H])C([H])([H])C([H])([H])C([H])([H])C([H])(N([H])[H])C(*)=O 0.000 description 2
- 210000004962 mammalian cells Anatomy 0.000 description 2
- 239000011159 matrix material Substances 0.000 description 2
- 238000001471 micro-filtration Methods 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000006011 modification reaction Methods 0.000 description 2
- 238000001728 nano-filtration Methods 0.000 description 2
- 230000000737 periodic Effects 0.000 description 2
- 235000011007 phosphoric acid Nutrition 0.000 description 2
- 229920000406 phosphotungstic acid polymer Polymers 0.000 description 2
- 230000000896 plasminic Effects 0.000 description 2
- WCUXLLCKKVVCTQ-UHFFFAOYSA-M potassium chloride Chemical compound [Cl-].[K+] WCUXLLCKKVVCTQ-UHFFFAOYSA-M 0.000 description 2
- 238000001556 precipitation Methods 0.000 description 2
- 238000011002 quantification Methods 0.000 description 2
- 230000008929 regeneration Effects 0.000 description 2
- SQVRNKJHWKZAKO-OQPLDHBCSA-N sialic acid Chemical compound CC(=O)N[C@@H]1[C@@H](O)C[C@@](O)(C(O)=O)OC1[C@H](O)[C@H](O)CO SQVRNKJHWKZAKO-OQPLDHBCSA-N 0.000 description 2
- VYPSYNLAJGMNEJ-UHFFFAOYSA-N silicium dioxide Chemical compound O=[Si]=O VYPSYNLAJGMNEJ-UHFFFAOYSA-N 0.000 description 2
- 238000003998 size exclusion chromatography high performance liquid chromatography Methods 0.000 description 2
- 239000000243 solution Substances 0.000 description 2
- 235000000346 sugar Nutrition 0.000 description 2
- 230000001225 therapeutic Effects 0.000 description 2
- 239000011534 wash buffer Substances 0.000 description 2
- ZBMRKNMTMPPMMK-UHFFFAOYSA-N 2-amino-4-[hydroxy(methyl)phosphoryl]butanoic acid;azane Chemical compound [NH4+].CP(O)(=O)CCC(N)C([O-])=O ZBMRKNMTMPPMMK-UHFFFAOYSA-N 0.000 description 1
- 102100001249 ALB Human genes 0.000 description 1
- 101710027066 ALB Proteins 0.000 description 1
- 206010002368 Anger Diseases 0.000 description 1
- 210000001367 Arteries Anatomy 0.000 description 1
- 210000004369 Blood Anatomy 0.000 description 1
- 241000283707 Capra Species 0.000 description 1
- MTCFGRXMJLQNBG-UWTATZPHSA-N D-serine Chemical compound OC[C@@H](N)C(O)=O MTCFGRXMJLQNBG-UWTATZPHSA-N 0.000 description 1
- 102000033147 ERVK-25 Human genes 0.000 description 1
- 229940012952 Fibrinogen Drugs 0.000 description 1
- 102000008946 Fibrinogen Human genes 0.000 description 1
- 108010049003 Fibrinogen Proteins 0.000 description 1
- 229940019698 Fibrinogen containing hemostatics Drugs 0.000 description 1
- 102000003886 Glycoproteins Human genes 0.000 description 1
- 108090000288 Glycoproteins Proteins 0.000 description 1
- 239000000899 Gutta-Percha Substances 0.000 description 1
- 206010018987 Haemorrhage Diseases 0.000 description 1
- 241000726306 Irus Species 0.000 description 1
- FFEARJCKVFRZRR-BYPYZUCNSA-N L-Methionine Natural products CSCC[C@H](N)C(O)=O FFEARJCKVFRZRR-BYPYZUCNSA-N 0.000 description 1
- ZDXPYRJPNDTMRX-VKHMYHEASA-N L-glutamine Chemical compound OC(=O)[C@@H](N)CCC(N)=O ZDXPYRJPNDTMRX-VKHMYHEASA-N 0.000 description 1
- HNDVDQJCIGZPNO-YFKPBYRVSA-N L-histidine Chemical class OC(=O)[C@@H](N)CC1=CN=CN1 HNDVDQJCIGZPNO-YFKPBYRVSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 239000007987 MES buffer Substances 0.000 description 1
- 241001480512 Mammalian orthoreovirus 3 Species 0.000 description 1
- 208000010125 Myocardial Infarction Diseases 0.000 description 1
- 101710003043 NFKBIZ Proteins 0.000 description 1
- 108091005771 Peptidases Proteins 0.000 description 1
- 230000037289 Plasma half life Effects 0.000 description 1
- 230000037240 Plasma half-life Effects 0.000 description 1
- 229950008882 Polysorbate Drugs 0.000 description 1
- 229960000380 Propiolactone Drugs 0.000 description 1
- 239000004365 Protease Substances 0.000 description 1
- 208000010378 Pulmonary Embolism Diseases 0.000 description 1
- 102000012479 Serine Proteases Human genes 0.000 description 1
- 108010022999 Serine Proteases Proteins 0.000 description 1
- DBMJMQXJHONAFJ-UHFFFAOYSA-M Sodium laurylsulphate Chemical compound [Na+].CCCCCCCCCCCCOS([O-])(=O)=O DBMJMQXJHONAFJ-UHFFFAOYSA-M 0.000 description 1
- 108010051181 TNK-tissue plasminogen activator Proteins 0.000 description 1
- 229940035295 Ting Drugs 0.000 description 1
- STCOOQWBFONSKY-UHFFFAOYSA-N Tributyl phosphate Chemical compound CCCCOP(=O)(OCCCC)OCCCC STCOOQWBFONSKY-UHFFFAOYSA-N 0.000 description 1
- FPKOPBFLPLFWAD-UHFFFAOYSA-N Trinitrotoluene Chemical compound CC1=CC=C([N+]([O-])=O)C([N+]([O-])=O)=C1[N+]([O-])=O FPKOPBFLPLFWAD-UHFFFAOYSA-N 0.000 description 1
- 229960004441 Tyrosine Drugs 0.000 description 1
- 238000002835 absorbance Methods 0.000 description 1
- 230000004913 activation Effects 0.000 description 1
- 238000005377 adsorption chromatography Methods 0.000 description 1
- 230000002776 aggregation Effects 0.000 description 1
- 238000004220 aggregation Methods 0.000 description 1
- 229940050528 albumin Drugs 0.000 description 1
- 229960003318 alteplase Drugs 0.000 description 1
- 229910000147 aluminium phosphate Inorganic materials 0.000 description 1
- 150000001413 amino acids Chemical class 0.000 description 1
- 238000005349 anion exchange Methods 0.000 description 1
- 229910052586 apatite Inorganic materials 0.000 description 1
- 238000010420 art technique Methods 0.000 description 1
- 230000000740 bleeding Effects 0.000 description 1
- 231100000319 bleeding Toxicity 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 230000000295 complement Effects 0.000 description 1
- 239000000356 contaminant Substances 0.000 description 1
- 230000000875 corresponding Effects 0.000 description 1
- 238000004185 countercurrent chromatography Methods 0.000 description 1
- 239000012228 culture supernatant Substances 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 230000004059 degradation Effects 0.000 description 1
- 238000006731 degradation reaction Methods 0.000 description 1
- 230000036425 denaturation Effects 0.000 description 1
- 238000004925 denaturation Methods 0.000 description 1
- 230000018109 developmental process Effects 0.000 description 1
- 239000012470 diluted sample Substances 0.000 description 1
- 230000000694 effects Effects 0.000 description 1
- 235000020673 eicosapentaenoic acid Nutrition 0.000 description 1
- 239000002158 endotoxin Substances 0.000 description 1
- 108010072542 endotoxin binding proteins Proteins 0.000 description 1
- 238000005265 energy consumption Methods 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 238000011067 equilibration Methods 0.000 description 1
- 230000002349 favourable Effects 0.000 description 1
- 238000000855 fermentation Methods 0.000 description 1
- 230000004151 fermentation Effects 0.000 description 1
- 230000020764 fibrinolysis Effects 0.000 description 1
- 239000012467 final product Substances 0.000 description 1
- 238000004108 freeze drying Methods 0.000 description 1
- 238000002523 gelfiltration Methods 0.000 description 1
- 239000004220 glutamic acid Substances 0.000 description 1
- 238000010438 heat treatment Methods 0.000 description 1
- 238000002318 immunoblotting Methods 0.000 description 1
- 230000000415 inactivating Effects 0.000 description 1
- 239000004615 ingredient Substances 0.000 description 1
- 125000001449 isopropyl group Chemical group [H]C([H])([H])C([H])(*)C([H])([H])[H] 0.000 description 1
- 230000001665 lethal Effects 0.000 description 1
- 231100000518 lethal Toxicity 0.000 description 1
- 108020004707 nucleic acids Proteins 0.000 description 1
- 150000007523 nucleic acids Chemical class 0.000 description 1
- 239000003960 organic solvent Substances 0.000 description 1
- 238000009928 pasteurization Methods 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
- 239000000546 pharmaceutic aid Substances 0.000 description 1
- 239000008194 pharmaceutical composition Substances 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 230000033885 plasminogen activation Effects 0.000 description 1
- 239000002797 plasminogen activator inhibitor Substances 0.000 description 1
- 238000005498 polishing Methods 0.000 description 1
- 238000002264 polyacrylamide gel electrophoresis Methods 0.000 description 1
- 108091008117 polyclonal antibodies Proteins 0.000 description 1
- 229920000136 polysorbate Polymers 0.000 description 1
- 239000001103 potassium chloride Substances 0.000 description 1
- 235000011164 potassium chloride Nutrition 0.000 description 1
- 239000012264 purified product Substances 0.000 description 1
- 238000003753 real-time PCR Methods 0.000 description 1
- 238000011084 recovery Methods 0.000 description 1
- 238000004064 recycling Methods 0.000 description 1
- 238000004366 reverse phase liquid chromatography Methods 0.000 description 1
- 239000000377 silicon dioxide Substances 0.000 description 1
- NRHMKIHPTBHXPF-TUJRSCDTSA-M sodium cholate Chemical compound [Na+].C([C@H]1C[C@H]2O)[C@H](O)CC[C@]1(C)[C@@H]1[C@@H]2[C@@H]2CC[C@H]([C@@H](CCC([O-])=O)C)[C@@]2(C)[C@@H](O)C1 NRHMKIHPTBHXPF-TUJRSCDTSA-M 0.000 description 1
- 235000011008 sodium phosphates Nutrition 0.000 description 1
- 239000002904 solvent Substances 0.000 description 1
- 238000011146 sterile filtration Methods 0.000 description 1
- 230000001954 sterilising Effects 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 150000008163 sugars Chemical class 0.000 description 1
- QAOWNCQODCNURD-UHFFFAOYSA-L sulfate Chemical compound [O-]S([O-])(=O)=O QAOWNCQODCNURD-UHFFFAOYSA-L 0.000 description 1
- 238000002211 ultraviolet spectrum Methods 0.000 description 1
- 241001430294 unidentified retrovirus Species 0.000 description 1
- 239000004474 valine Substances 0.000 description 1
- 238000005406 washing Methods 0.000 description 1
- 239000008215 water for injection Substances 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- VEZXCJBBBCKRPI-UHFFFAOYSA-N β-Propiolactone Chemical compound O=C1CCO1 VEZXCJBBBCKRPI-UHFFFAOYSA-N 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- A61K38/43—Enzymes; Proenzymes; Derivatives thereof
- A61K38/46—Hydrolases (3)
- A61K38/48—Hydrolases (3) acting on peptide bonds (3.4)
- A61K38/482—Serine endopeptidases (3.4.21)
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/02—Inorganic compounds
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/16—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing nitrogen, e.g. nitro-, nitroso-, azo-compounds, nitriles, cyanates
- A61K47/18—Amines; Amides; Ureas; Quaternary ammonium compounds; Amino acids; Oligopeptides having up to five amino acids
- A61K47/183—Amino acids, e.g. glycine, EDTA or aspartame
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/06—Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
- A61K47/26—Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P9/00—Drugs for disorders of the cardiovascular system
- A61P9/10—Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
-
- C—CHEMISTRY; METALLURGY
- 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)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/64—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
- C12N9/6402—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from non-mammals
- C12N9/6405—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from non-mammals not being snakes
- C12N9/6408—Serine endopeptidases (3.4.21)
-
- 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)
- C12N9/48—Hydrolases (3) acting on peptide bonds (3.4)
- C12N9/50—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
- C12N9/64—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
- C12N9/6421—Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
- C12N9/6424—Serine endopeptidases (3.4.21)
- C12N9/6456—Plasminogen activators
- C12N9/6459—Plasminogen activators t-plasminogen activator (3.4.21.68), i.e. tPA
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12Y—ENZYMES
- C12Y304/00—Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
- C12Y304/21—Serine endopeptidases (3.4.21)
- C12Y304/21068—Tissue plasminogen activator (3.4.21.68), i.e. tPA
Abstract
The present invention relates to a process of isolating and purifying tissue plasminogen activator and its variants more specifically TNK-tPA from CHO cells, the process uses a series of affinity chromatography and filtration steps to overcome known difficulties in the industrial scale purification of TNK-tPA. of TNK-tPA.
Description
A NOVEL ERR}E‘iCA’i‘EON PROCESS FOR ESQLATEON AME) C(EMIWERCEAL
PROBUCTEGN 0F RECQMBENANT TNKnT?A "TENECTEPLASE
HELD 917 THE ENVENTEON
Present invention is related to a novel process of isolation, purification and production of
tissue plasminogen activator PA) from mammalian cells, more specifically from
Chinese Hamster Ovary (CHO) cells.
BACKGROUND 0? TEE EN‘VENTION
Tenecteplase (TNK—tPA) is a recombinant glycoprotein of serine protease family with six
amino acids tution in the native human tissue nogen activator (t—PA) with 17
disulphide bridges and having molecular weight of . In the pment of TNK—tPA,
the modifications made in native t—PA includes tution of threonine 103 with asparagine,
tution of asparagine 117 with glutamine both within the kringle 1 domain, and the
substitution of lysine, histidine and two arginine with tetra—alanine amino acids at 296—299
positions in the protease domain to make the resulting protein highly fibrin specific with
longer plasma half life and 80% decreased susceptibility to degradation by nogen
activator inhibitor—1 (PAI—l) compared to native t—PA.
The TNK in TNK—TPA refers to the sites of the t—PA molecule that have been modified
i.e.TlO3; N1 17; and KHRR 296—299.The aforementioned modifications of TNK—tPA renders
its use as an improved eutic agent for the treatment of acute myocardial infarction
which has better therapeutic compliance because the greater fibrin specificity allows for
faster and complete clot lysis with decreased ng complications and the long pan
permits a single bolus dose with less systemic fibrinolysis and lesser bleeding complications
from the previous clot buster drugs.
The mechanism of TNK—tPA is initiated on binding of TNK—tPA to the fibrin component of
the thrombus (blood clot) which selectively converts inalctive plasminogen into plasmin and
consequently the resultant plasmin degrades the matrix of thrombus in occluded artery while
conserving fibrinogen and zing systemic plasminogen activation due to its highly
specific nature.
The benefits of TNK—tPA seen in myocardial infarction patients and the encouraging results
from animal studies in the context of Acute Ischemic Stroke (AIS), suggested that TNK—tPA
might prove to be a safer and more effective y than ase, the only drug ed
by USFDA for AIS. Over the past few years, several clinical trials evaluated the use of TNK—
tPA in AIS and proved that TNK—tPA has a better pharmacological profile than alteplase and
also suggested that it could be an effective and safe therapeutic option in treating AIS in
patients reporting within 4.5 h after symptom onset. ly, TNK—tPA has been considered
for the treatment of patients with pulmonary embolism and several clinical trials showed
promising outcomes. A large number of clinical trials are still being conducted to assess the
complete conclusive picture of TNK—tPA in l indications.
In the last few years, development and manufacturing of recombinant giycoproteins was
carried out by batch, fed hatch, semi~fed batch and perfusion bioreactors processes and for
purification of these proteins adsorption and ion exchange chromatography were majorly
employed.
For t~PA and its variants certain purification ois are known in prior art cg. purification
by immuno affinity (anti—tPA goat polyclonal antibody), ion exchange, ethanol precipitation,
reverse phase chromatography, chromatography on silica or anion exchange, such as
diethylamino ethyl, ammonium sulphate precipitation, sephadex— G—75 etc.
Some of the approaches for the purification of TNK t—PA is listed in prior art includes W0
til5922, sets out a purification process where series of ion ge chrornatt‘rgraphy
steps, immunoaffinity chromatography and ultrafil'tration/diafiitration steps are used for
purification of 'E‘NK—tPA. Wt} zeta/usessa A, sets out a purification process prirnatiiy
drawn to the use of hydrophobic interaction chromatography.
The immune affinity tography used in the prior art is not suitable technique for
commercial cturing of TNK—tPA. Not only it could raise lot of tory coneems
but the cost of immune affinity chromatography media is also very high compared to
com'entional chn'irnatography matrices owing to their use of rnonocioual antibodies for
preparation. Hydrophobic interaction chromatography described in certain prior art for IN Kr-
tPA cation uses isopropyl alcohoi (IPA) in the process which is an organic solvent and
known for ng aggregation and denaturation of proteins and may he considered as one
of the disadvantages of the prier art. "l‘NK-tl’A is a highly unstable molecule and hence use of
{PA in the purificatitm prneess sheuld be avoided as it may lead to the denaturatien of the
protein. ln additinn, the large volume usage of EPA at commercial scale tvnuld require
recycling of lPA whieh again demands additienal energy consumption and extra investment
such as snlvent recovery unit.
'l‘herel’ore, none (if the aforementioned processes are capable 0f prnviding an efficient,
scalable and robust pnrifieatinn solution, which could consistently produce TNK—tPA drug
substance at emnrnereial seale, meeting all the required Sp ations
Renee, there is a need fer an effective and cmnrnercially viable prneess fer cation of
’l‘NKutl’A.
GREECE {39F THE lNVlEN’E‘EON
The object of the present invention is to develop an efficient, robust, scalable, and
commercially viable purification process for the production of TNK—tPA resulting yield not
less than 60% and purity more than 95% as measured by Size exclusion chromatography.
SUMMARY 0F ’l‘HlEl ENVEN’E‘EQN
The present invention relates to a novel process of isolating and purifying tissue plasminogen
tor and its ts more specifically TNK—tPA from CHO cells and bes an
industrially applicable, simple, cost effective, robust and highly efficient process of A
purification.
A s for isolation and purification of TNK—tPA of the present invention comprising
steps of:
i) subjecting cell free harvest obtained from CHO cell culture to affinity
chromatography to capture TNK—tPA and obtaining an eluate containing
partially purified TNK—tPA.
(ii) subjecting the eluate of step (i) to ty chromatography for additional
purification of TNK—tPA and obtain an eluate containing ily A.
(iii) viral inactivation of eluate of step (ii) to obtain the viral vated sample;
4 [followed by page 4A]
(iv) subjecting the viral inactivated sample of step (iii) to a further affinity
chromatography for additional purification to obtain an eluate containing
primarily highly purified TNK-tPA;
(v) subjecting the eluate of step (iv) to cation exchange chromatography to obtain an
eluate containing highly purified preparation of TNK-tPA;
(vi) subjecting the eluate of step (v) to virus reduction filtration for removal of virus
present;
(vii) concentrating the sample of step (vi) to obtain TNK- tPA; wherein the yield of the
process is more than 60 % and purity of TNK-tPA obtained is more than 95% as
measured by Size exclusion chromatography.
In one particular aspect the invention provides a process for ion and purification of
TNK-tPA comprising steps of:
(i) subjecting cell free harvest obtained from CHO cell culture to affinity
chromatography-I to capture A and obtaining an eluate containing partially
purified TNK-tPA;
wherein the affinity tography-I comprises Blue ose 6 FF as
a stationary phase and a mobile phase, n the mobile phase is a mixture of
sodium ate buffer, sodium chloride and urea;
(ii) subjecting the eluate of step (i) to ty chromatography-II for
additional purification of TNK-tPA and obtain an eluate containing primarily TNK-tPA;
wherein the affinity chromatography-II comprises Lysine Hyper D as a
stationary phase and a mobile phase, wherein the mobile phase is a mixture of sodium
acetate buffer, urea and epsilon aminocarproic acid (EACA);
(iii) viral inactivation of eluate of step (ii) to obtain the viral inactivated
sample;
wherein viral inactivation is conducted by low pH and chemical
inactivation, wherein chemical inactivation is done by ng the sample with sodium
caprylate in the presence of urea;
4A [followed by page 4B]
(iv) subjecting the viral inactivated sample of step (iii) to a further affinity
chromatography-III for additional cation to obtain an eluate containing primarily
highly purified TNK-tPA;
wherein the affinity chromatography-III comprises Ceramic Hydroxy
Apatite (CHT) as a stationary phase and a mobile phase, wherein the mobile phase is a
mixture of phosphate buffer, 2-(N-Morpholino) ethanesulfonic acid hydrate (MESHydrate
), sodium chloride and urea;
(v) subjecting the eluate of step (iv) to cation exchange chromatography to
obtain an eluate containing highly purified preparation of TNK-tPA;
wherein the cation exchange chromatography comprises Fractogel SO3 as
a stationary phase and a mobile phase, wherein the mobile phase is a mixture of
L-Arginine, phoric acid and polysorbate-20;
(vi) subjecting the eluate of step (v) to virus reduction filtration for removal of
virus t;
wherein virus filtration is d out by polyethersulphone (PES)
nanofilter having a size in the range from 15-20 nm; and
(vii) concentrating the sample of step (vi) to obtain TNK- tPA;
wherein the yield of the s is more than 60% and purity of TNK-tPA
obtained is more than 95% as measured by Size exclusion chromatography.
BRIEF DESCRIPTION OF DRAWINGS
Figure 1: Depicts a chromatogram of affinity-I cation where first peak represents
ties and second peak corresponds to eluate ning TNK-tPA.
Figure 2: Depicts a SDS PAGE r stained) profile of affinity-I purification where lane no 5,
6 and 7 showing wash fractions (1, 2 and 3 tively) and Lane No. 9 corresponds to eluate
containing TNK-tPA.
Figure 3: Depicts a chromatogram of affinity- II purification, UV 280 peak corresponding to
eluate containing TNK-tPA.
Figure 4: Depicts a SDS PAGE (silver stained) profile of affinity-II purification where lane No.
9 corresponds to eluate containing TNK-tPA
Figure 5: Depicts a chromatogram of affinity- III purification, UV 280 peak (first) corresponds
to eluate containing TNK-tPA
Figure 6: Depicts a SDS PAGE (silver d) profile of affinity-III purification where lane No.
9 corresponds to eluate containing TNK-tPA
Figure 7: Depicts a togram of cation exchange purification, UV 280 peak corresponds to
eluate containing TNK-tPA.
Figure 8: Depicts a SDS PAGE (silver stained) profile of cation exchange purification where
lane No. 5 corresponds to eluate containing TNK-tPA.
Figure 9: Depicts a ison of ng SDS-PAGE (silver stained) profile of drug
nce obtained after purification using steps described in present invention and innovator
product (Metalyse).
[FOLLOWED BY PAGE 5]
Figure it}: Depicts a comparison of non reducing SDS RAGE (silver stained) profile of drug
substance obtained after purification using steps described in present invention and innovator
product (Metalyse).
Figure ii: Depicts a peptide inap cin‘oniatogram of drug substance obtained after
purification using step described in present invention resembles with tor product
(Metaiyse)
Figure 12: Depicts a immune blotting of drug substance obtained after purification using
steps described in present invention resembles with innovator product {Metaiyse}
Figure 13: Depicts the breakthrough curve of Affinity-l (Blue Sepahrose FF) in batci'i mode.
Figure 14: Depicts Chromatograms for the PCC run over two cycles for (a) Column A —first
column in the loading zone ; (b) Column B — the second column in the loading zone ; (0)
Column C — the third column in the loading zone. ; (d) Column chromatograms mposed
on each other showing full PCC run. UV is measured at 280 nm. Run performed on an
XKl6—5ml Blue Sepharose Fast Flow.
Figure 15: Depicts Chromatogram from the first coiumn (Column A) overioading and
capture in Column 8, UV measured at 280 um. Run performed on an Xtil i Blue
Sepharose Fast Flow.
Figure 16: Depicts Chromatogram from the third column (Column C) g all post load
washing. Impact of wash steps are denoted by A — Wash 1, B— Wash 2, C— wash 3, D —
n, E— Regeneration 1, F— Regeneration 2, G—Regeneration 3, H—Regeneration 4. UV
measured at 280 nm. Run performed on an XKl6—5ml Blue Sepharose Fast Flow.
DETAILED DESCRIPTION OF INVENTION
The present ion s to a novel process of isolating and purifying tissue nogen
activator and its variants more ically TNK—tPA. The cell free t is obtained from
the cells cultured in bioreactors. The list of symbols and abbreviations used in specification
of the present invention are listed at Table A below:
Table A: List of symbols and iations
tPA Tissue Plasminogen Activator
CHO Chinese Hamster Ovary
kDa Kilo—Dalton
PAI Plasminogen Activator Inhibitor
WO 63299
AIS Acute Ischemic Stroke
SDS PAGE Sodium l Sulfate Polyacrylamide Gel Electrophoresis
IPA Isopropyl Alcohol
CHT Ceramic y Apatite
FF Fast Flow
mM Milli Molar
MTX Methotrexate
EACA Epsilon—aminocaproic acid
MES— Hydrate 2—(N—Morpholino) ethanesulfonic acid hydrate
TFF Tangential Flow Filtration
UF Ultrafiltration
DF Diafiltration
PES Polyethersulphone
PVDF Polyvinylidene Fluoride
u Micron or Micrometer
XMuLV Xenotropic Murine Leukemia Virus
PRV Pseudorabies Virus
Reo—3 Reovirus Type 3
MMV Murine Minute Virus
CV Column Volume
DBC Dynamic Binding ty
HCP Host Cell n
DNA De—oxyribo Nucleic Acid
HCD Host Cell DNA
SEC— HPLC Size Exclusion Chromatography
HPLC High Performance Liquid Chromatography
LAL Limulous Amoebocyte Lysate
UV Ultraviolet
MMC Mixed Mode Chrmnatography
The present invention relates to a novel process of isolating and purifying tissue plasminogen
activator and its variants more specifically TNK—tPA from CHO cells and describes an
industrially applicable, , cost effective, robust and highly efficient process of TNK—tPA
purification
A process for isolation and cation of TNK—tPA of the present invention comprising
steps of:
i) subjecting cell free harvest obtained from CHO cell culture to ty
chromatography to e TNK—tPA and obtaining an eluate containing
partially purified TNK—tPA.
(ii) subjecting the eluate of step (i) to affinity chromatography for additional
purification of A and obtain an eluate containing primarily TNK—tPA.
(iii) viral inactivation of eluate of step (ii) to obtain the viral inactivated sample;
(iv) subjecting the viral inactivated sample of step (iii) to a further affinity
chromatography for additional purification to obtain an eluate containing
ily highly purified TNK—tPA;
(v) subjecting the eluate of step (iv) to cation exchange chromatography to obtain
an eluate containing highly purified preparation of A;
(vi) subjecting the eluate of step (v) to virus reduction filtration for removal of
virus present;
(vii) concentrating the sample of step (vi) to obtain TNK— tPA; wherein the yield of
the process is more than 60 % and purity of TNK—tPA obtained is more than
95% as measured by Size exclusion chromatography.
The process of the present invention may be explained by rating the steps as below:
(i) Subjecting cell free harvest obtained from CHO cell culture to affinity
chromatography to e TNK-tPA and obtaining an eluate containing
partially purified TNK-tPA.
The cell free harvest containing TNK—tPA may be obtained from perfusion technology
based fermentation system by CHO cells. The harvest ning TNK—tPA may be
filtered with 0.2 u and collected in sterile ners and stored at 2—8°C till further
use. The cell free harvest containing TNK—tPA is subjected to affinity
chromatography. The stationary phase of affinity chromatography may be selected
from the group comprising Blue Sepharose 6 fast flow, Lysine Hyper D, Ceramic
Hydroxy Apatite (CHT), preferably the stationary phase is Blue Sepharose 6 FF. The
column may be equilibrated by using a buffer comprising Phosphate buffer, sodium
chloride and polysorbate 20 or their mixtures. The elution buffer or mobile phase
employed for capture step ity Chromatography) may be individually or in
ation selected from phosphate buffer, urea, and sodium chloride. More
ably, the elution buffer or mobile phase is used in combination. The
concentration of sodium phosphate in buffer used is preferably 20—50 mM sodium
phosphate, more preferably 20—40mM. The concentration of sodium chloride used in
the buffer is 1—2.5 M NaCl, more preferably l.5—2M.The tration of urea in the
buffer may be in the range of 1—4 molar, preferably in the range of 2—3 molar. The pH
of the elution buffer or mobile phase may be maintained in the range of 7—8,
preferably in the range of 7—7.6, more preferably in the range of 4.
The removal of host cell proteins from products produced in mammalian cells is
always a difficult task. In the present invention the step optimized as capture
chromatography in current invention selectively removes host cell proteins to an
extent of 0.8 to 1.5 log, more specifically 1.0 log which helps in achieving final
concentration of HCP in purified TNK—tPA preparation, below 100 ppm.
Not only the host cell proteins (HCP) are reduced but process related impurities such
as albumin, gentamycin, Methotrexate (MTX) etc. are also removed ively by the
capture chromatography described in present invention.
(ii) ting the eluate of step (i) to affinity chromatography for additional
ation of TNK-tPA to obtain an eluate containing primarily TNK-tPA.
The eluate of step (i) may be subjected to affinity chromatography. The stationary
phase or column material may be selected from the group comprising of Blue
Sepharose 6 fast flow, Lysine Hyper D, Ceramic Hydroxy Apatite (CHT), etc.,
preferably the stationary phase is Lysine Hyper D. The equilibration buffer ed
in ty chromatography step may be individually or in combination selected from
Phosphate buffer, sodium chloride and polysorbate. More preferably the equilibration
buffer is used in ation.
The column may be eluted by a elution buffer or mobile phase selected from the
group comprising individually or in combination selected from sodium acetate, urea
and Epsilon—aminocaproic acid (EACA). The pH of the affinity chromatography
WO 63299
elution buffer may be done in the range of pH 3.5—6, preferably 4—5, more preferably
4.5-5.
The chromatography matrix ed for intermediate cation e.g. Affinity—II
chromatography step may be selected from the group comprising of Blue Sepharose 6
fast flow, Lysine Hyper D and Ceramic Hydroxy Apatite (CHT), preferably the
chromatography matrix used is Lysine Hyper D.
The n buffer ed in Affinity chromatography step may be 5—25 mM
sodium acetate, 1—4 M urea, 01—04 M EACA having pH 4.0—5 .0. The concentration
of sodium acetate in elution buffer is 5—25 mM, preferably in the range of 5—15 mM.
The concentration of EACA in the buffer is 01—04 mM, preferably in the range of
0.1—0.2 M. The concentration of urea is 1—4 M in the buffer, preferably in the range of
2—3 M.
The inventive merit of elution buffer by using EACA at acidic pH in this step is to
e the most suitable condition. The process of the present invention as set out
herein is most suitable for viral inactivation and reduces the handling and material
consumption by several folds. Approximately 2—4 log reduction of viral clearance and
1—1.5 log reduction of host cell ns (HCP) are achieved by Affinity
chromatography used in present invention. On an average the 1.69 log reduction with
XMuLV and 4.30 log reduction with PRV are achieved by Affinity chromatography
used in present ion.
(iii) Viral inactivation of eluate of step (ii) to obtain the viral inactivated sample;
The viral inactivation of the eluate obtain from step (ii) may be conducted by any
method such as heat treatment (Pasteurization, Lyophilisation/dry heat), irradiation,
ultraviolet (UV), high tatic pressure, low pH incubation, chemical and
solvent/detergent treatments. Chemical inactivation or viral inactivation is done by
treating the sample with chemical selected from the group comprising sodium cholate,
triton, Beta—propiolactone, tri(n—butyl)phosphate (TNBP), and sodium caprylate,
preferably treating the sample with sodium caprylate at low pH in the presence of
Urea. The tration of chemical/detergent is in the range of 0.001% to 0.10%
W/v, preferably 0.01% — 0.07% (W/v), more preferably 0.05%.The concentration of
urea is used in the range of 1—4 M, ably 2—3 M. The viral activation may also be
carried out by incubating the sample for a period of 40—180 minutes, more preferably
for a period of 40—80 minutes, in the temperature range of l5—45°C, more preferably
in the range of 20—300C.
Generally low pH is the choice and most Widely used method of viral inactivation in
purification processes. Low pH is known to vate enveloped viruses but it cannot
inactivate highly resistant non enveloped viruses. The pH is in range from 4.0 to 4.7,
more preferably in range from 4.3 to 4.7.
The composition of viral vation buffer described in present invention is
optimized in such a manner that it could inactivate both ped and non enveloped
viruses efficiently. Use of the als/detergents, urea, EACA and low pH of the
t invention makes the conditions lethal for enveloped and non enveloped
viruses and makes the combination an optimum choice for inactivating highly
resistant viruses. On an average 5.65 log reduction With XMuLV and 5.38 log
reduction with PRV are achieved by low pH and chemical inactivation of the present
invention.
(M Subjecting the viral inactivated sample of step (iii) to a further affinity
chromatography to obtain an eluate containing TNK-tPA;
The viral inactivated sample of step (iii) may be subjected to a further affinity
chromatography. The stationary phase or column material of affinity tography
may be selected from the group of Blue Sepharose 6 fast flow, Lysine Hyper D,
Ceramic Hydroxy Apatite (CHT), etc., preferably the nary phase or
chromatography matrix used is Ceramic Hydroxy Apatite (CHT). The stationary
phase or column may be eluted by a mobile phase or buffer selected from the group
comprising individually or in combination selected from Phosphate buffer, 2—(N—
Morpholino) ethanesulfonic acid e (MES— Hydrate), sodium chloride and urea.
The pH of the affinity chromatography elution buffer may be done at pH g from
6—9, preferably 6—8, more preferably 6—7.
The mobile phase or elution buffer employed in Affinity chromatography step may be
—50 mM phospahte buffer, preferably in the range of 5—15 mM. The concentration of
urea is used in the range of 1—4 M in buffer, ably in the range of 1—3 M. The
concentration of MES— e used may be 2—20 mM in the buffer. Elution may be
d by increasing concentration of salt. The salt of this n may be selected
from group of potassium chloride, sodium chloride, sodium phosphate and
ammonium sulphate, preferably the salt is sodium chloride and concentration of
sodium chloride is in range of 0.1—1.0 M sodium chloride. The elution type may be
linear, step or in combination of both, preferably the elution used is linear salt
gradient.
Affinity chromatography (multimode chromatography) used in present invention is to
remove traces of impurities, specifically host cell DNA (HCD), host cell
proteins(HCP) and viral impurities if any. Approximately 2—4 log reduction of viral
clearance and 0.5 —l log reduction of HCP is achieved using affinity—III
chromatography mentioned in t invention. On an average 4.23 log reduction
with XMuLV, 4.06 log reduction with PRV, 3.73 log reduction With Reo—3 and 2.97
log reduction with MMV are achieved by Affinity tography used in present
invention.
Subjecting the eluate of step (iv) to cation exchange chromatography to obtain
an eluate containing highly purified ation of A in formulation
buffer;
The cation exchange chromatography of the eluate of step (iv) may be conducted by
using a matrix or stationary phase selected from the group comprising Fractogel S03,
Fratogel SE Hicap, SP Sepharose FF, CM Sepharose FF etc., preferably the
chromatography matrix or stationary phase used is Fractogel S03. The equilibration
buffer employed in cation exchange tography may be individually or in
combination selected from group comprising phosphate buffer, urea, MES and
sodium chloride, preferably the equilibration buffer is used in combination.
The elution buffer or mobile phase employed in cation exchange chromatography
may be individually or in combination selected from nine, O—phosphoric acid
and rbate—20, ably, the elution buffer is used in combination or mixture.
The mobile phase or elution buffer employed in cation exchange chromatography
comprising L—Arginine present in range from 10—350 mM, preferably in range from
250—350 mM, O—phosphoric acid present in range from 0.5—1%, preferably in range
from 0.6%—0.8% and polysorbate—20 present in range from 0.01—0.05%, preferably in
range from 0.04 to 0.05 % . The pH of the cation exchange chromatography elution
buffer or mobile phase may be done in the range of 7.0 to 7.5, preferably in the range
7.3— 7.5.
Generally the ion exchange chromatography is used as e, intermediate and
polishing chromatography to either remove bulk or traces of impurities. In present
invention the cation exchange chromatography is somewhat used differently than it is
conventionally used. Herein, the eluate of us chromatography is directly loaded
on to cation exchange chromatography to obtain the highly purified ation of
TNK—tPA in final formulation buffer containing arginine, hosphoric acid and
polysorbate 20. The advantage of using cation exchange chromatography differently
is that buffer exchange and simultaneous concentration and purification are achieved
in a single step. In prior art techniques like gel filtration tography and
diafiltration using Tangential Flow Filtration (TFF) are employed for buffer
exchange. These techniques are efficient and most commonly used for buffer
exchange but cannot further purify target protein. Due to the fact that one can only
load m 30% of sample to column volume in gel filtration chromatography,
bigger size of columns are required compared to ion exchange tography. Gel
filtration chromatography also causes the dilution of target protein during buffer
exchange which further requires some additional concentration step like TFF. Buffer
exchange using diafiltration is also not feasible When volumes to be buffer exchanged
are higher since it es very high amount of buffer and demands large size of
UF/DF unit. Considering entioned limitations of conventional buffer exchange
techniques the cation ge chromatography described in present invention is
capable to provides a buffer exchange step and assists in further cation also. It is
possible to achieve a viral reduction factor of 1.21 log with non enveloped virus e.g.
MMV using ion exchange chromatography of the present invention.
(vi) subjecting the eluate of step (v) to virus reduction tion for removal of virus
present;
The virus filtration may be performed after affinity chromatography, cation exchange
chromatography, and tangential flow filtration step, more preferably the virus
tion step is performed after cation exchange chromatography. Nanofilter ed
for this step may be of 15 nm, 20 nm, and higher. Preferably the nanofilter size is
between 15— 20 nm. The nanofilter used may be made up of cellulose, PES, PVDF
etc. More preferably the filter used is PBS.
The filtration performed after virus filtration may be ed from microfiltration,
ultra filtration, nano filtration, macro filtration, tial flow filtration, etc. More
preferably the filtration is tangential flow filtration. On an average 4.15 log reduction
with XMuLV, 3.40 log reduction with PRV, 4.41 log reduction with Reo—3, 4.76 log
reduction with MMV are achieved by virus reduction filtration used in present
invention.
The Overall downstream process provides more than 15 log reduction with XMuLV,
more than 17 log reduction with PRV, more than 8 log reduction with Reo—3, and
more than 8 log reduction with MMV.
The most probable contaminant in the process of TNK t—PA would be CHO cell
derived retrovirus and XMuLV represents a non—defective C type irus for CHO
cells, a more than 15 log reduction obtained for XMuLV is considered most nt
and gives a high assurance in terms of viral safety.
(vii) Concentrating the sample of step (vi) to obtain TNK-tPA.
The filtrate obtained in step (vii) is subjected to filtration method selected from the
group comprising iltration, ultrafiltration, nanofiltration, microfiltration and
tangential flow filtration, preferably the filtration method is tangential flow filtration.
Ultra filtration membrane selected for this step may be of 5, 10, 30 or 50kDa.
Preferably the ultra filtration membrane used is in the range of 5—30 kDa, more
preferably the size of ultra filtration membrane used is 10 kDa. The Ultra tion
membrane used may be made up of cellulose, PES, PVDF etc. More preferably the
filter used is PBS. The concentration of TNK—tPA retentate may be in the range of
1.0i0.4mg/ml to 4mg/ml. More preferably, the concentration of TNK—tPA may
be in the range of l.0i0.4mg/ml to 6.0i0.4mg/ml. Most preferably, the concentration
of TNK—tPA is 5.5i0.4mg/ml.
The TNK—tPA drug substance (Tenecteplase) may ably be obtained by sterile
filtration of TFF Retentate using 0.2 u sterilizing grade filters made up of PBS,
PVDF, and Cellulose. More preferably sterile filter is made up of PBS.
The present ion also ses a process, wherein the batch Affinity—I
chromatography may also be operated in continuous mode using periodic counter
current chromatography (PCC). The use of FCC, provides additional advantage e.g.
reduced buffer ption, increased productivity, steady state operation and better
process controls. .
The present invention, includes Within its scope, the use of inline buffer and
chromatography load conditioning cum preparation by five pump based customized
AKTA process system With a maximum flow rate of 600 L/h. The said activity can be
performed by Flow feedback or pH—Flow feedback mode of control for buffer and
chromatography load preparation of Affinity—I, II, III and IEC tography steps
as mentioned in Example 1 to 4.
The process of the present invention results in a purified product of TNK—tPA with
increased yield and purity. The attributes of TNK—tPA obtained by the process of the
present invention is set out in detail at Table B.
Table B: s pertaining to Quality of TNK-tPA
S.No. Critical Quality Attribute Quality of Purified A BULK
1 Appearance Clear colorless to ly ish liquid
2 pH 7.0 -7.6
3 Protein (mg/ml) Not less than 5.0 mg/ml
4 No additional band other than principal band
SDS PAGE
Identified with specific antibody and resembles
Immunoblotting
with qualified standard
6 Bioactivity (U/mg) l60 U/mg to 240 U/mg
7 Monomer (%) More than 95%
8 Single Chain t (%) More than 60 %
9 HCP (ppm) Less than 100 ppm
Sialic Acid (mol/mol of
2.9 to 5.7 moles/mol of TNK—tPA
TPA)
11 l Sugar ol of
.5 to 13.5 moles/mol of TNK—tPA
TPA)
12 Type40 %,
Type I 11 content
, Type
Type—II 60—72%
13 HCD Less than 10ng/dose
14 BET < 1EU/mg
Serine(S)—Tyrosine(Y)—Glutamine(Q)—
Valine(V)—Isoleucine(I)—Cysteine(C)—Arginine(
N—Terminal Sequence (First
R)—Aspartic acid(D)—Glutamic acid(E)—
amino acid )
Lysine(K)_Threonine(T)—Glutamine(Q)—
Methionine(M)—Isoleucine(I)—Tyrosine(Y)
16 Osmolality 260-320 mOsm/Kg
17 Arginine Content 50—60 mg/ml
18 Chromatogram n resembles with qualified
Peptide mapping
standard
19 UV Spectrum (Amax) 280i2nm
In an embodiment the process of the present invention is e to remove or inactivate
viruses as potential itious agents as assessed using a scaled down purification process.
The high log clearance values obtained for XMuLV, PRV, Reo—3 and MMV provides a
very good assurance that any adventitious viruses which could not be detected, or might gain
access to the production process, would be cleared/or inactivated, during highly capable
purification process, mentioned in the current invention and thus reducing the l risk to
patient safety.
In addition to higher assurance of viral safety, the aforementioned improvements in the
cation process of TNK—tPA are also beneficial in terms of decreased human
intervention, lower capital and operational expenditures for higher yield TNK—tPA
preparation.
In an embodiment the t invention provides a pharmaceutical composition comprising
the TNK—tPA retentate obtained from the process of present invention in liquid parenteral I.V
formulation with pharmaceutically acceptable excipients for acute myocardial infarction and
acute ischemic stroke.
In an embodiment the ceutical ition of present invention comprises:
Ingredient Concentration
eplase 5 mg
Arginine 55 mg
Polysorbate — 20 0.43 mg
Phosphoric Acid 17 mg
Water for injection q.s to 1.0 ml
In r ment the present invention provides the use of the isolated and prepared
TNK—tPA in liquid eral I.V formulation for acute myocardial infarction and acute
ischemic stroke.
The invention is described in detail herein below with respect to the following examples
which are provided merely for ration and are not intended to restrict scope of invention
in any manner. Any embodiments that may be apparent to a person skilled in the art are
deemed to fall within the scope of present invention.
Example- 1
The cell free harvest containing TNK—tPA is subjected to Affinity chromatography column
packed with Blue Sepharose FF. before loading, the Column is equilibrated with 5 Column
Volume (CV) of equilibration buffer. The loading is stopped till column achieve saturation.
The loading capacity of column is decided based on the Dynamic Binding Capacity (DBC) of
column which is in range of l — 2 mg/ml. After loading, column is washed with equilibration
buffer until loosely bound s and t related impurities were washed away in
equilibration wash. For further removal of host cell proteins another wash buffer is used
which is composed of urea, sodium chloride, sodium phosphate and polysorbate 20.
After column wash, TNK—tPA is eluted using elution buffer containing 20—50 mM Sodium
Phosphate, 1—2 M NaCl, 2—3 M urea, and 0.04—0.l % polysorbate 20. Affinity—I eluate is
filtered with 0.2um filter. Samples are awn and analysed by reduced and non reduced
SDS PAGE to know the purity profile and single chain/double chain content. Those who
skilled in art can understand the ality of single chain/double chain composition in final
TNK—tPA drug nce.
The present invention is advantageous due to direct capture of clarified harvest without
mixing in large mixing tanks.
Example- 2
Affinity—I Chromatography Eluate is diluted with affinity—II on or affinity
chromatography equilibration buffer containing 20—50 mM Sodium Phosphate, 0.04—0.l %
polysorbate20 at pH 7.2 to reduce the conductivity up to less than 15 ms/cm. Diluted sample
is clarified using 0.2 u filter and loaded on to Affinity—II chromatography. Column is washed
with bration buffer to bring the UV280 absorbance to baseline. Column is further
washed to remove s and t related impurities with wash buffer containing 20—50
mM sodium ate, 1—3 M NaCl, 0.04—0.l% polysorbate 20 and pH 7.2.Purified TNK—
tPA is recovered and eluted from the column by passing elution buffer consisting of 5—25 mM
sodium acetate, 1—4 M urea, 0.1—0.4 M EACA and pH 4.0—5.0. All the chromatography
samples including load, flow through, washes, and elution were analyzed using following
analytical methods:
SDS PAGE (reduced/non reduced) for purity and single chain/double chain content.
Size Exclusion High Performance Chromatography (SEC—HPLC) For Aggregate Content
TNK —tPA content measured by Clot Lysis assay.
Total Protein by rd and UV280 nm.
HCP content using ELISA (Cygnus third generation kit)
Affinity chromatography is optimized for removing process & product related impurities. The
method of elution in this step is optimized in such a way that it complements to viral
inactivation step and the composition with condition of elution buffer e.g. urea, EACA, and
low pH are optimized to inline with viral inactivation. Approximately 2 to4 log reduction of
viral clearance and 1.0 to 1.5 log reduction of host cell proteins (HCP) are achieved after
Affinity—II chromatography step.
The other advantage is using EACA at acidic pH in ty tography—II in elution
buffer inspite of L—Arginine and EACA at neutral pH. This particular change in purification
step is valuable in reducing the cost of L—Arginine and also provides an optimum condition
for viral inactivation. Hence, it can be stated that the same step is not only favorable for
TNK—tPA n but also optimum for viral inactivation that in turn reduces the work load
and material consumption with time.
Example- 3
The Elution of Affinity Chromatography is subjected to low pH and chemical inactivation
using sodium caprylate. Mixture is incubated at 20 to 25°C for 60 min. In viral inactivation
step, sodium ate used is in very low amount that eliminates the need of large mixing
vessels. In prior art sodium caprylate was used to inactivate viruses present in before capture
chromatography where s are comparatively higher hence quantity of sodium caprylate
required was also high. In present invention, sodium Caprylate is added after second
chromatography steps where volume to be handled is low and therefore es less amount
of sodium ate and much smaller vessel for handling. Apart from that, the use of sodium
caprylate at pH 4.5 as compared to neutral or alkaline pH, provides a more effective and
robust viral inactivation in the process.
After viral inactivation the solution is diluted using ate buffer for loading on to
Affinity chromatography (mixed mode tography) to remove traces of impurities,
specifically HCD, HCP and viral impurities if any. Approximately 2—4 log reduction of viral
clearance and 0.5 —l log reduction of HCP clearance are achieved by affinity
chromatography. In prior art same c hydroxyl apatite is described for tissue
plasminogen activator purification, but none of the s have described the capability to
remove impurities e.g. HCP, DNA and viruses. Criticality of removing such ties is
evident by the fact that the amount of these impurities is tested in final product (except viral
load) and is part of final drug substance e specifications. All the chromatography
samples including load, flow through, washes, and elution are analyzed using following
analytical methods:
SDS PAGE (reduced/non reduced) for purity and single chain/double chain content.
Size Exclusion High performance Chromatography (SEC—HPLC) for ate t
TNK —tPA content measured by Clot Lysis assay.
Total Protein by Bradford and UV280 nm.
HCP content using ELISA (Cygnus third generation kit)
Example- 4
The Affinity chromatography eluate without any conditioning is directly loaded on to cation
exchange chromatography for concentration and buffer exchange of target protein. Cation
ge chromatography is optimized in such a way that it avoids cumbersome dilution
steps for feed conditioning and therefore the ty—III eluate can be directly loaded on to
the cation exchange chromatography. TNK—tPA is red from the column by passing
elution buffer containing 55 mg/ml L—Arginine, 17 mg/ml of orthophosphoric acid, 0.43
mg/ml Polysorbate 20 and pH 7.4.Cation exchange chromatography eluate is ted to
filtration for viral reduction and the resultant filtrate is further concentrated using Tangential
Flow Filtration (TFF) system to achieve the final drug substance concentration. After
concentration the TFF Retentate is sterile filtered and kept at —20°C for further use. Drug
nce produced by the purification process of present ion is thoroughly analyzed by
the state of art and validated analytical procedures which includes but not limited to; Identity
and purity check by SDS PAGE, Western Blot, N—terminal sequence analysis and peptide
map,
HCP determination using ELISA,
ivity and TNK—tPA quantification using clot lysis assay,
Host cell DNA quantification using qPCR,
Endotoxin fication using LAL test,
ate and single chain/double chain content using size ion HPLC,
Arginine content & Osmolality,
Sialic acid, neutral sugars, type—I and type—II glycoforms analysis,
Analysis of process related impurities e.g. Gentamycin, MTX, Urea, Sodium Caprylate, and
EACA using in—house developed methods.
After extensive analysis and biophysical comparison with innovator product it can be
concluded that the product purified by the process described in current invention is yielding
TNK—tPA product which is highly similar to innovator product with l process yield of
more than 60%.
Example 5:
A Periodic counter current tography (PCC) for affinity—I step has been med with
cell culture supernatant containing TNK—tPA from perfusion based bioreactor. In batch mode
dynamic binding capacity for affinity—I chromatography media were evaluated and based on
the information obtained from break through analysis a three column FCC has been
experimented on XKl6— 5 ml BLUE SEPAHROSE FF. The chromatographic buffer
compositions were kept same as mentioned in Example—l.
Example 6:
The method for ation of a liquid mixture of controlled pH & ionic strength for required
buffers, dilution and/or conditioning of chromatography load by five pump based customized
AKTA process system with a maximum flow rate of 600 L/h for TNK—tPA downstream
processing. The liquid mixtures prepared with the above system With d recipes are
suitable for purification of TNK—tPA at different chromatography stages as mentioned in
Example—1 to 4.
WE
Claims (19)
1. A process for ion and cation of TNK-tPA si ng steps of: (i) subjecting cell free harvest ed from CHO cell culture to affinity chromatography-I to capture TNK-tPA and ing an eluate containing partially purified TNK-tPA; wherein the affinity chromatography-I comprises Blue Sepharose 6 FF as a stationary phase and a mobile phase, wherein the mobile phase is a mixture of sodium phosphate buffer, sodium chloride and urea; (ii) subjecting the eluate of step (i) to affinity chromatography-II for additional purification of TNK-tPA and obtain an eluate containing primarily TNK-tPA; wherein the affinity chromatography-II comprises Lysine Hyper D as a stationary phase and a mobile phase, wherein the mobile phase is a mixture of sodium acetate buffer, urea and epsilon aminocarproic acid ; (iii) viral inactivation of eluate of step (ii) to obtain the viral inactivated sample; n viral inactivation is conducted by low pH and chemical inactivation, wherein chemical inactivation is done by treating the sample with sodium caprylate in the presence of urea; (iv) subjecting the viral inactivated sample of step (iii) to a further affinity chromatography-III for additional purification to obtain an eluate containing primarily highly purified TNK-tPA; wherein the affinity chromatography-III comprises Ceramic Hydroxy Apatite (CHT) as a stationary phase and a mobile phase, n the mobile phase is a e of phosphate buffer, 2-(N-Morpholino) sulfonic acid hydrate (MESHydrate ), sodium chloride and urea; (v) subjecting the eluate of step (iv) to cation exchange chromatography to obtain an eluate containing highly purified preparation of TNK-tPA; wherein the cation exchange chromatography comprises Fractogel SO3 as a nary phase and a mobile phase, wherein the mobile phase is a mixture of L-Arginine, O-phosphoric acid and polysorbate-20; (vi) subjecting the eluate of step (v) to virus reduction filtration for removal of virus present; wherein virus filtration is carried out by polyethersulphone (PES) lter having a size in the range from 15-20 nm; and (vii) concentrating the sample of step (vi) to obtain TNK- tPA; n the yield of the process is more than 60% and purity of A obtained is more than 95% as ed by Size exclusion chromatography.
2. The process as claimed in claim 1, wherein the TNK-tPA purity is more than 95% by Size Exclusion Chromatography and wherein the other critical quality attributes are: No additional band other than principal band SDS PAGE observed Monomer (%) More than 95% Single Chain Content (%) More than 60 % Host cell n (HCP) (ppm) Less than 100 Host cell DNA (HCD) (ng/dose) Less than 10
3. The process as claimed in step (i) of claim 1, wherein the sodium phosphate is present in range from 20–40 mM, sodium chloride is t in range from 1.5–2 M and urea is present in range from 2–3 M; wherein the pH of the mobile phase is present in range from 7.2–7.4.
4. The process as claimed in claim 3, wherein the affinity chromatography-I provides 0.8 – 1.5 log reduction in host cell proteins.
5. The process as claimed in step (ii) of claim 1, wherein the sodium acetate is present in range from 5–15 mM, urea is present in range from 2–3 M and EACA is present in range from 0.1–0.2 mM; wherein the pH of the affinity chromatography-II mobile phase is in range of pH 4.5–5.
6. The process as claimed in claim 5, wherein the affinity tography-II results 1–1.5 log reduction in host cell proteins and 1–4 log reduction of viral nce.
7. The process as claimed in claim 5, wherein the affinity chromatography-II results more than 1.5 log reduction of Xenotropic Murine Leukemia Virus (XMuLV) and more than 4 log reduction of Pseudorabies (PRV).
8. The process as d in step (iii) of claim 1, wherein sodium caprylate is present in the range from 0.01% - 0.07% (w/v), ably in the range of 0.05%; n concentration of urea is present in the range of 2–3 M; wherein viral inactivation is conducted by holding the sample for a period from 40–80 minutes; wherein holding of the sample at a ature range from 20–30°C; wherein the pH is present in range from 4.3 to 4.7.
9. The process as claimed in claim 8, wherein the Low pH and chemical inactivation results in more than 5 log reduction of XMuLV and PRV.
10. The process as d in step (iv) of claim 1, wherein the phosphate buffer is present in range from 5–15 mM, MES- Hydrate is present in range from 2–20 mM, urea is present in range from 1–3 M and sodium chloride is present in range from 0.1 to 0.5 M; wherein the mobile phase pH is in the range from 6–9; wherein the affinity chromatography-III elution type is linear salt gradient; wherein the salt of this elution is sodium chloride in a range from 0.1–1.0 M sodium chloride.
11. The process as claimed in claim 10, wherein the affinity chromatography-III results 0.5–1 log reduction in host cell proteins and 2 – 4 log of viral clearance.
12. The process as claimed in claim 10, wherein the affinity chromatography-III results more than 4 log reduction with XMuLV and PRV and more than 3 log reduction with MMV and Reo 3 viruses.
13. The process as claimed in step (v) of claim 1, wherein the L-Arginine is t in range from 250-350 mM, O-phosphoric acid is present in range from .8% and polysorbate-20 is present in range from 0.04 to 0.05 %; and wherein the mobile phase pH is in range from 7.3–7.5.
14. The s as claimed in claim 13, wherein the cation exchange chromatography results more than 1 log viral nce with MMV and more than 0.5 log nce for host cell proteins.
15. The process as claimed in step (vi) of claim 1, wherein the virus filtration results more than 4 log reduction of XMuLV, more than 3 log reduction of PRV, more than 4 log reduction of Reo-3 and more than 4.5 log reduction of MMV.
16. The process as claimed in step (vii) of claim 1, wherein the filter is ultra filtration membrane; wherein the ultra filtration membrane is PES; wherein the size of ultra filtration membrane is in a range from 5-30 kDa, preferably the size of ultra filtration membrane is 10 kDa.
17. The process as claimed in claim 16, wherein the concentrate is TNK-tPA retentate.
18. A process as d in claim 1, wherein ic r current chromatography is used for Affinity chromatography-I.
19. A process as claimed in claim 1, wherein Inline conditioning approach is used for buffer and load preparation for Affinity chromatography-I, Affinity chromatography-II, Affinity tography-III (Mixed Mode Chromatography) and cation exchange chromatography. WO 63299
Applications Claiming Priority (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
IN2343/MUM/2014 | 2014-10-21 | ||
IN2343MU2014 | 2014-10-21 | ||
PCT/IN2015/050137 WO2016063299A2 (en) | 2014-10-21 | 2015-10-19 | A novel purification process for isolation and commercial production of recombinant tnk-tpa (tenecteplase) |
Publications (2)
Publication Number | Publication Date |
---|---|
NZ731196A NZ731196A (en) | 2020-09-25 |
NZ731196B2 true NZ731196B2 (en) | 2021-01-06 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2015334455B2 (en) | A novel purification process for isolation and commercial production of recombinant TNK-tPA (Tenecteplase) | |
EP0954528B1 (en) | A method for chromatographic removal of prions | |
JP5730211B2 (en) | Viral arginine inactivation | |
RU2590726C2 (en) | Method for purifying vitamin k dependent proteins such as coagulation factor ix | |
JP6876655B2 (en) | Protein purification method | |
US9663553B2 (en) | Integrated process for the production of therapeutics (human albumin, immunoglobulins, clotting factor VIII and clotting factor IX) from human plasma | |
EP1519944B1 (en) | Processes for the preparation of fibrinogen | |
AU2006287833A1 (en) | An ultra-high yield intravenous immune globulin preparation | |
US20070049733A1 (en) | Ultra-high yield intravenous immune globulin preparation | |
CN103319592B (en) | Method for obtaining an IgG composition by heat treatment | |
RU2649363C2 (en) | Methods for reducing and/or removing fxi and fxia from solutions containing said clotting factors | |
TWI531576B (en) | Method for purifying transgenic factor vii | |
WO2014089954A1 (en) | Method for preparing antithrombin | |
NZ731196B2 (en) | A novel purification process for isolation and commercial production of recombinant tnk-tpa (tenecteplase) | |
WO2011107299A9 (en) | Method for the manufacture of recombinant dspa alpha1 | |
US9145448B2 (en) | Method for the isolation of haptoglobin | |
KR20140013925A (en) | Purification of cell culture derived alpha1 protease inhibitor | |
US20040106779A1 (en) | Modified factor IX preparation | |
BR112017006294B1 (en) | PROCESS FOR ISOLATION AND PURIFICATION OF TENECTEPLASE (TNK-TPA) | |
EP3271380B1 (en) | Purification of bone morphogenetic proteins (bmps) | |
CN113121638B (en) | Method for purifying recombinant protein | |
US20220056107A1 (en) | Method for Filtering Fibrinogen | |
EP2699594B1 (en) | Process for the preparation of a virus-inactivated fv concentrate starting from human plasma, scalable to industrial level |