WO2001032915A2 - Reversed-phase hplc assay for plasminogen activators - Google Patents

Reversed-phase hplc assay for plasminogen activators Download PDF

Info

Publication number
WO2001032915A2
WO2001032915A2 PCT/US2000/030252 US0030252W WO0132915A2 WO 2001032915 A2 WO2001032915 A2 WO 2001032915A2 US 0030252 W US0030252 W US 0030252W WO 0132915 A2 WO0132915 A2 WO 0132915A2
Authority
WO
WIPO (PCT)
Prior art keywords
tpa
cho
human
sample
buffer
Prior art date
Application number
PCT/US2000/030252
Other languages
French (fr)
Other versions
WO2001032915A3 (en
Inventor
Yuan Xu
Original Assignee
Genentech, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to DE60003449T priority Critical patent/DE60003449T2/en
Priority to MXPA02004401A priority patent/MXPA02004401A/en
Application filed by Genentech, Inc. filed Critical Genentech, Inc.
Priority to DK00978343T priority patent/DK1226269T3/en
Priority to CA2384756A priority patent/CA2384756C/en
Priority to SI200030178T priority patent/SI1226269T1/en
Priority to AU15816/01A priority patent/AU781023B2/en
Priority to JP2001535595A priority patent/JP4659320B2/en
Priority to BRPI0015283 priority patent/BRPI0015283B8/en
Priority to NZ518187A priority patent/NZ518187A/en
Priority to HU0202973A priority patent/HU226202B1/en
Priority to IL14900600A priority patent/IL149006A0/en
Priority to EP00978343A priority patent/EP1226269B1/en
Priority to AT00978343T priority patent/ATE243263T1/en
Priority to PL355389A priority patent/PL204011B1/en
Publication of WO2001032915A2 publication Critical patent/WO2001032915A2/en
Publication of WO2001032915A3 publication Critical patent/WO2001032915A3/en
Priority to NO20022122A priority patent/NO329876B1/en
Priority to HK02107782.5A priority patent/HK1046158B/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/37Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving peptidase or proteinase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/914Hydrolases (3)
    • G01N2333/948Hydrolases (3) acting on peptide bonds (3.4)
    • G01N2333/972Plasminogen activators
    • G01N2333/9726Tissue plasminogen activator
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N30/00Investigating or analysing materials by separation into components using adsorption, absorption or similar phenomena or using ion-exchange, e.g. chromatography or field flow fractionation
    • G01N30/02Column chromatography

Definitions

  • This invention is directed to an assay for determining the amount of Chinese Hamster Ovary (CHO) -produced tPA is present in samples of recombmant human tPA with native sequence or its variants produced n CHO cells.
  • Tissue-type plasmmogen activators are endogenous se ⁇ ne proteases involved n a cascade of events leading to the dissolution of a blood clot (Astrup and Permm, Nature, 159, 681-682 (1947); Camiolo et al . , Proc. Soc. Exp. Biol. Med. , 138, 277-280 (1971) ; Collen, J. Biol . Chem. , 33, 77-86 (1987); Hoylaerts et al . , J. Biol. Chem., 257, 2912-2919 (1982)).
  • ACTIVASE 0 is the recombmant form of human tPA (r-tPA) , used in the management of acute myocardial infarction and pulmonary embolism (Grossbard, Pharm. Res . , 4., 375-378 (1987)). ACTIVASE 0 is also now approved for treating lschemic stroke (Smith et al . , Acad. Emergency Medicine, .6(6), 618-25 (1999); Kwiatkowski et al . , New Eng. J. Med., 340(23), 1781-1787 (1999)). It is a glycoprotem produced by expressing the complementary DNA (cDNA) for natural human tPA m Chinese hamster ovary (CHO) cells.
  • cDNA complementary DNA
  • TNK-tPA is a genetically engineered variant of human tPA cloned and expressed in CHO cells (Keyt et al . , Proc. Natl. Acad. Sci USA., 91, 3670-3674 (1994)). Site-directed mutations were introduced at three specific sites of human tPA to create the TNK-tPA variant. They are Thrl03 to Asn (T103N), Asn 117 to Gin (N117Q) , and Lys-His-Arg-Arg 296-299 to Ala-Ala-Ala-Ala (KHRR296-299AAAA) .
  • TNK-tPA When compared to tPA, TNK-tPA exhibits similar m vi tro biological activity, an increased resistance to plasmmogen activator inhibitor and an enhanced fibrin specificity, and is cleared more slowly from plasma (Keyt et al . , Proc. Natl. Acad. Sci USA., 91, 3670-3674 (1994); Thomas et al . , Stroke, 25:10, 2072-2079 ( 1994); Benedict et al . , Circulation, 92:10, 3032- 3040 (1995); Modi et al . , Thromb Haemost, 79, 134-139 ( 1998)). It is currently awaiting regulatory approval as a single bolus administered form of r-tPA.
  • CHO-PA CHO cells biosynthesize endogenous hamster tPA called CHO-PA.
  • CHO-PA has a similar fib ⁇ nolytic activity to human tPA as determined by the clot lysis assay.
  • the ammo acid sequence of CHO-PA is 80% identical to that of human tPA. Many of the substitutions are semi-conservative such as: Arg ⁇ -->Lys, Glu ⁇ -->Asp, Phe ⁇ -->Tyr, Val ⁇ -->Ala, Ile ⁇ —>Leu or Thr ⁇ —> Ser.
  • r-tPA, TNK-tPA, and CHO-PA are all single polypeptide chains composed of 527 ammo acids with 17 disulfide bonds (Nguyen and Carole, "Stability Characterization and Formulation Development of Altepase, a Recombmant Tissue Plasmmogen Activator," in Stability and Characterization of Protein and Peptide Drugs: Case Histories, Y. J. Wang, R. Pearlman, eds . , (Plenum Press: New York, 1993), pp.
  • the peptide bond between Arg 275 and Ile 276 is particularly susceptible to protease cleavage.
  • the cleavage results two fragments: one consisting of the N-termmal 275 ammo acids and the other consisting of the C-termmal 252 ammo acids.
  • the N-termmal chain contains regions which are homologous to the krmgle regions found in plasmmogen and prothrombm and, therefore, is often referred to as the "krmgle fragment" (Nguyen and Carole, supra ; de Vos et al . , Biochem. , 31, 270-279 (1992) ) .
  • the C-termmal chain contains the catalytically active site and, therefore, is commonly referred to as the "protease fragment” (Pennica et al . , Nature, 301, 214-221 (1983)).
  • the cleaved two chains are linked by a single disulfide bond formed between Cys 264 and Cys 395 .
  • the cleaved molecule is commonly referred to as "two-chain tPA” as opposed to " single-chain tPA” or the intact form.
  • r-tPA contains four potential sites for N-lmked glycosylation identified by the sequence Asn-X-Ser/Thr (Nguyen and Carole, supra ) .
  • r-tPA exists as two glycosylation isozymes designated type I and type II.
  • Type I r-tPA is glycosylated at Asn 117 , Asn 184 , and Asn 448 ; whereas type II r-tPA is glycosylated only at Asn 117 and Asn 448 .
  • Asn 218 is not glycosylated m either isoforms.
  • TNK-tPA has the same glycosylation pattern as r-tPA, except that the Thrl03 to Asn and Asnll7 to Gin mutations effectively moved the glycosylation site from position 117 to 103 (Keyt et al . , supra) .
  • the glycosylation pattern for CHO-PA is not fully characterized (Ri ⁇ ken and Collen, J. Biol. Chem., 256: 7035-7041 (1981)
  • ACTIVASE ® is a trademark for the recombmant form of human tissue-type plasmmogen activator (r-tPA) , used m the management of acute myocardial infarction and pulmonary embolism.
  • ACTIVASE & brand tPA is also now approved for treating lschemic stroke. It is produced by expressing the complementary DNA (cDNA) for natural human tPA in Chinese hamster ovary (CHO) cells (U.S. Pat. No. 5,753,486) .
  • TNK-tPA is a genetically engineered variant of r-tPA with enhanced efficacy and lower incidence of bleeding compared with ACTIVASE ® r-tPA.
  • CHO-PA biosynthesize endogenous hamster tPA called CHO-PA.
  • ammo acid sequence of CHO-PA is highly homologous (80% identical) to that of r-tPA. All three thrombolytic proteins exist as heterogeneous isoforms, mainly due to proteolysis/hydrolysis and differential glycosylation.
  • a method for purifying human tPA from CHO-PA is described in U.S. Pat. No. 5,411,864.
  • This method comprises contacting a fluid containing the human tPA with antibodies specifically binding the corresponding endogenous CHO-PA and recovering the human tPA.
  • the contacting step involves passing the fluid through a chromatographic bed having the antibodies immobilized thereon.
  • Peptide mapping is a key method for monitoring the ammo acid sequence and is able to detect small changes small to moderate size proteins, for example, insulin and human growth hormone.
  • a reversed-phase HPLC method was developed herein for the analysis of the three thrombolytic molecules, CHO-tPA, recombmant human tPA with native sequence, and TNK-tPA.
  • This method not only has the ability to resolve human tPA and/or TNK-tPA from CHO-PA, but also is capable of identifying and quantifying different isoforms of each molecule.
  • the present invention provides a process for monitoring the effectiveness of a purification process in removing plasmmogen activator (PA) endogenous to Chinese hamster ovary (CHO) cells from a sample containing human tPA or variants thereof, which process comprises incubating the sample with a protease capable of specifically cleaving the Arg 275 - Ile 276 bond of human wild- type tPA and then with denaturing and reducing agents in amounts effective to reduce the disulfide bonds of human wild-type tPA; subjecting the sample to a reversed-phase high-performance liquid chromatography step, and analyzing the elution profile from the chromatography step for the amount of PA endogenous to the CHO cells present therein.
  • PA plasmmogen activator
  • CHO Chinese hamster ovary
  • Figure 1 shows a reversed-phase HPLC analysis of native r-tPA, TNK-tPA and CHO-PA.
  • Figure 2 shows a reversed-phase HPLC analysis of DTT/urea treated r-tPA, TNK-tPA and CHO-PA. Plasmmogen activators were treated with DTT/urea prior to chromatography .
  • Figure 3 shows a reversed-phase HPLC analysis of plasmm and DTT/urea treated r-tPA, TNK-tPA and CHO-PA. Plasmmogen activators were subjected to plasmm treatment followed by DTT/urea treatment prior to chromatography.
  • tissue plasmmogen activator and "tPA” refer to human extrinsic (tissue-type) plasmmogen activator having fibr olytic activity that typically has a structure with five domains (finger, growth factor, krmgle-1, krmgle-2, and protease domains) , but nonetheless may have fewer domains or may have some of its domains repeated if it still functions as a thrombolytic agent and retains the N-lmked glycosylation sites at positions 117, 184, and 448.
  • the tPA consists of a protease domain that is capable of converting plasmmogen to plasmm, and an N-termmal region believed to be at least partially responsible for fibrm binding, and retains the N-lmked glycosylation sites at positions corresponding to ammo acid positions 117, 184, and 448 of wild-type human tPA.
  • the retention of these glycosylation sites is due to the fact that variable site occupancy of recombmant and melanoma-derived wild-type tPA leads to production of two variants, designated as "Type I tPA" and "Type
  • Type I tPA contains N-lmked oligosaccha ⁇ des at positions 117, 184, and 448.
  • Type II tPA contained N-lmked oligosaccharides at positions 117 and 448. It will be understood that natural allelic variations exist and can occur among individuals, as demonstrated by one or more ammo acid differences in the ammo acid sequence of tPA of each individual.
  • wild-type human tissue plasmmogen activator refers to native-sequence human tPA, i.e., that encoded by the cDNA sequence reported in U.S. Pat. No. 4,766,075, issued 23 August 1988. Ammo acid site numbers or positions m the tPA molecule are labeled in accordance with U.S. Pat. No. 4,766,075.
  • references to various domains of tPA mean the domains of wild-type human tPA as heremabove defined, and functionally equivalent portions of human tPA having ammo acid alterations as compared to the native human tPA sequence, or of (native or variant) tPA from other sources, such as bat tissue plasmmogen activator (bat-PA) .
  • bat-PA bat tissue plasmmogen activator
  • proteease domain refers to the region extending from ammo acid position 264 to ammo acid position 527, inclusive, of the mature form of wild-type human tPA, and to functionally equivalent portions of human tPA having ammo acid alterations as compared to the native human tPA sequence, or of tPA from other sources, such as bat-PA.
  • tPA variants refers to molecules that differ from native tPA by one or more amino acid changes or modifications to existing ammo acids. TNK-tPA is the preferred variant herein.
  • the modification to change or insert the appropriate ammo ac ⁇ d(s) n the native molecule to effect the desired sequence variations is accomplished by any means known m the art, such as e.g. site-directed mutagenesis or ligation of the appropriate sequence into the DNA encoding the relevant protein.
  • TNK-tPA refers to a tPA molecule wherein Thrl03 of wild- type tPA is changed to Asn (T103N) , Asnll7 of wild-type tPA is changed to Gin (N117Q) , and Lys-His-Arg-Arg 296-299 of wild-type tPA is changed to Ala-Ala-Ala- Ala (KHRR296-299AAAA) .
  • TNK is further described n U.S. Pat. No. 5,612,029.
  • Chomiecki et al . in Advances m Animal Cell Biology and Technology for Bioprocesses, Spier et al . , eds . (1989), pp. 442-451), as well as CHO derivatives such as CHO/-DHFR (Urlaub and Chasm, Proc. Natl. Acad. Sci.
  • Preferred host cells include CHO-Kl DUX Bll and dpl2.CHO cells.
  • CHO cells developed for large-scale production of tPA are maintained cryogenically in a MCB/workmg cell bank (WCB) system as described by Wiebe et al . , n Large Scale Mammalian Cell Culture Technology, Lubmiecki, ed., (Marcel Dekker: New York, 1990), pp. 147-160.
  • DHFR+ CHO-Kl cells transfected with DNA encoding human tPA have been deposited at the American Type Culture Collection, Manassas, Virginia (ATCC) , and are available under accession number CCL 61.
  • a sample of another tPA-producmg CHO cell line (CHO cell line 1-15 15 ) has been deposited under ATCC accession number CRL 9606. The latter cell line was reported to result in human tPA levels approaching 50 pg/cell/day.
  • CHO plasmmogen activator or "CHO-PA” refers to plasmmogen activator that is produced endogenously by CHO cells. This endogenous PA expressed by CHO cells has a sequence slightly different (about
  • the CHO-PA is not a tissue-type
  • proteases refers to an enzyme that is capable of cleaving the Arg 27S - Ile 276 bond of human wild-type tPA specifically.
  • examples include plasmm (or plasmmogen, which converts to plasmm), tissue kallikrem, or Factor Xa, as well as any trypsm-like proteases that can effect this specific, limited proteolysis. Eligible proteases are further described in Ichmosi et al . , FEBS Letters, 175: 412-418 (1984). Preferred herein is plasmm/plasmmogen .
  • denaturing/reducing agents or “denaturing agent and reducing agent” refers to a combination of denaturant and reductant that reduces the disulfide bonds of human wild-type tPA.
  • the denaturing agent is guanidme or urea and the reducing agent is dithiothreitol (DTT) or 2- mercaptoethanol .
  • the tPA or tPA variant is recovered from the CHO culture medium, either as a secreted protein or from host cell lysates when directly expressed without a secretory signal. It is necessary to purify the tPA or variant thereof from host cell proteins to obtain preparations that are substantially homogeneous as to protein.
  • the culture medium or lysate is centrifuged or filtered to remove particulate cell debris.
  • the human tPA or variant thereof is then purified from corresponding contaminant endogenous proteins such as CHO-PA by such techniques as fractionation on lmmunoaffmity or ion- exchange columns as described, for example, m U.S. Pat. No. 5,411,864; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation exchange resm such as DEAE; chromatofocus g; SDS-PAGE; ammonium sulfate precipitation; or gel electrophoresis using, for example, Sephadex G-75.
  • contaminant endogenous proteins such as CHO-PA by such techniques as fractionation on lmmunoaffmity or ion- exchange columns as described, for example, m U.S. Pat. No. 5,411,864; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation exchange resm such as DEAE; chromatofocus g; SDS-PAGE
  • a protease inhibitor that does not interfere with the tPA activity such as phenylmethylsulfonyl fluoride (PMSF) also may be useful to inhibit proteolytic degradation during purification, and antibiotics may be included to prevent the growth of adventitious contaminants.
  • PMSF phenylmethylsulfonyl fluoride
  • purification methods suitable for native tPA may require modification to account for changes m the character of tPA or its variants upon expression in recombmant cell culture.
  • a tPA variant is being produced, it is secreted and the supernatant is passed over a PBS-preconditioned column of glass beads coupled to anti-tPA goat polyclonal A6 antibody, the column is equilibrated with a buffer, and the tPA variant is then eluted.
  • the invention herein is directed to monitoring (including qualifying and quantifying) levels of native CHO-PA in a sample taken from such purification systems that contains at least one form of human tPA that is produced in CHO cells.
  • the process comprises incubating the sample with a protease that is capable of cleaving Arg 275 - Ile 276 bond specifically. This is followed by incubation of the protease-treated sample with a combination of a denaturing and reducing agent in proper relative and absolute amounts to effect reduction of the disulfide bonds m human wild-type tPA. Since the treatment with denaturing and reducing agents causes the loss of enzyme activity, the incubation with protease occurs first.
  • the sample is subjected to a reversed-phase high- performance liquid chromatography step, and the elution profile from the chromatography step analyzed for the amount of PA endogenous to the CHO cells present therein.
  • the protease is plasmmogen, which converts to the active form, plasmm
  • the human tPA is native-sequence tPA
  • the consecutive incubation step with the protease followed by the denaturing/reducing agents typically takes place at a temperature of about 30- 40°C, more preferably about 36-38°C, and most preferably about 37°C, for a minimum of about 15 minutes, more preferably about 20-40 minutes.
  • the sample is diluted with a digestion buffer, which preferably has a pH of about 7 to 8, more preferably phosphate buffer at pH 7.4-7.6, and more preferably also containing argin ne.
  • a digestion buffer which preferably has a pH of about 7 to 8, more preferably phosphate buffer at pH 7.4-7.6, and more preferably also containing argin ne.
  • HPLC column on which the sample is loaded may be utilized for the purposes of this invention, including preparative or analytical scale.
  • the column is typically equilibrated for at least about 15 minutes prior to sample injection.
  • Column size, column material, flow rate, elution buffers, type of gradient, injection volume, and particle size of column ⁇ epend on various factors, including the size of the sample being examined, the type of mobile phase composition and gradient, and the forms of tPA being distinguished.
  • the loading solvent may be any solvent but is preferably an acetonitrile- based solvent such as water, acetonit ⁇ le, and t ⁇ fluoroacetic acid (TFA) .
  • the column is a Zorbax C8, Vydac, or Baker C-18 column packed with a medium having a particle diameter of about 4-40 ⁇ m, more preferably about 5-15 ⁇ m, and a pore size of about 100-4000 A, more preferably about 150-350 A.
  • the medium preferably has a C4, C8, or C18 alkyl group, ana most preferably is a C8 silica medium.
  • the elution is carried out with a solvent comprising acetonit ⁇ le, such as water, acetonit ⁇ le, and TFA, in a gradient format over 60-100 minutes, preferably a linear gradient, wherein the relative amount of acetonit ⁇ le is increased in the solvent.
  • a shallow gradient ramp at about 0.25% acetonitrile per mmute is employed.
  • the tPA or variant thereof can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the tPA product is combined in admixture with a pharmaceutically acceptable carrier.
  • Such formulations are well described in the literature as well as dosages and uses.
  • the tPA or its variant is suitably administered parenterally to subjects suffering from cardiovascular diseases or conditions and strokes.
  • ACTIVASE ® (r-tPA) and TNK-tPA were obtained from Genentech, Inc. (South San Francisco, CA) m a form purified from CHO cells. See also, for example, U.S. Pat. Nos. 4,766,075 and 5,753,486 for ACTIVASE ® r-tPA and US Pat. No. 5,612,029 for TNK-tPA.
  • Monoclonal antibody #354 for CHO-PA was produced as described in U.S. Pat. No. 5,411,864. Briefly, a female Balb/c mouse was immunized over a period of 12 weeks with protein solutions substantially enriched in CHO plasmmogen activator purified from host cell lacking the human t-PA. There were five injections each consisting of approximately 30 ⁇ g . The initial injection was emulsified with complete Freund's adjuvant and administered in subcutaneous s ⁇ te(s). The second injection given 1.5 weeks later was emulsified with incomplete Freund's adjuvant and half was administered subcutaneously and half mtraperitoneally . The remaining three injections were given on weeks 3, 6 and 12 in phosphate buffered saline (PBS) administered in one mtraperitoneal site.
  • PBS phosphate buffered saline
  • mice The spleen from the immunized mouse was removed on week 13 and spleen cells were fused with the mouse myeloma cell line NP3X63-Ag8.653 using the general procedures of Fazekas et al . , J . Immunol. Methods, 35 : 1 (1980) and
  • Hybridoma cells from wells containing CHO-PA-specifIC antibodies were expanded and cloned by limiting dilution (Oi and Herzenberg, "Immunoglobm-Producmg Hybrid Cell Lines", p. 351-372, in Selected Methods m Cellular Immunology, Mishell and Shngi, eds . (W. H. Freeman and Co., 1980)). Large quantities of specific monoclonal antibodies were produced by cell culture of the hybridoma cells or by injection of hybridoma cells in mice thereby producing ascites tumors .
  • MAb 354 was one resulting antibody, which lowered CHO-PA levels greater than about 100 fold m a single column pass when immobilized and is stable to several different immobilization chemistries and harsh washing conditions.
  • MAb 354 was purified using the steps set forth below, where all steps were carried out at room temperature. Concentration was carried out as follows: The MAb hybridoma suspension culture harvest fluid (HF) was filtered through a 0.2 ⁇ m filter. The culture fluid was concentrated by ultraflltration or by chromatography on an ion-exchange resin. Affinity purification was carried out as follows: Thimerosal was added to the concentrated MAb HF solution to approximately 0.02%.
  • the solution was adjusted to approximately 1.5 M glycme, 3.0 M NaCl, pH 9.0 by addition of 3.0 M glycme, followed by addition of crystalline NaCl. Protein A has poor Ab binding, so addition of NaCl and glycme increases hydrophobic interaction, so as to facilitate Ab binding.
  • the solution was clarified by filtration.
  • the clarified concentrated MAb HF was applied to a column of protein A immobilized to agarose.
  • the bound MAb was washed with the buffer used to equilibrate the column, having the approximate composition of 1.5 M glycme, 3.0 M NaCl, 0.02 M EDTA, pH 9.0.
  • the MAb was eluted with 0.1 M sodium citrate, 0.15 M NaCl, pH 3.0 buffer.
  • the protein A column was regenerated by washing with 3.0 M NaSCN, 0.03 M TRIS, pH 8.5 and re-equilibrated.
  • the eluted MAb peak was collected based on absorbance profile at 280 nm.
  • the citrate-eluted MAb peak was immediately neutralized by collection into buffer with the approximate composition: 1.5 M TRIS-HC1, pH 9.0.
  • the protein A column was unpacked and stored in sealed containers at 2-8°C in approximately 0.02% thimerosal as storage buffer.
  • the 354 MAb was then buffer exchanged by diaflltration . Diaflltration proceeded until the conductivity and pH were similar to the values for 0.03 M TRIS, 0.05 M NaCl, pH 8.5 buffer.
  • the MAb was subsequently applied to an amon-exchange chromatograpny column containing DEAE-FAST FLOWTM agarose support and washed with 0.03 M TRIS, 0.05 M NaCl, pH 8.5 buffer.
  • the MAb was step eluted with a buffer having the approximate composition: 0.03 M TRIS, 0.15 M NaCl, pH 8.5. The eluted MAb peak was collected based on absorbance profile at 280 nm.
  • the MAb was filtered (0.2 ⁇ m) into sanitized sealable containers and stored below -40°C. Plasmmogen was obtained from Fluka (Switzerland) . HPLC-grade acetonitrile was obtained from Burdick & Jackson (Muskgon, MI) and t ⁇ fluoroacetic acid (TFA) was from Pierce (Rockford, IL) . Water for the HPLC mobile phase and sample solutions was purified with a MILLI-QTM system from Millipore (Milford, MA) . All other chemicals were of reagent grade from Sigma (St. Louis, MO). METHODS
  • CHO-PA was isolated from CHO cell culture fluid by affinity chromatography followed by lmmunoabsorption. Lysme hyper D resm from BioSepra (Paris, France) was used for the affinity chromatography, as lysme binds to the krmgle 2 region of plasmmogen activators (Cleary et al . , Biochem. 28, 1884-1890 (1989) ) . The lmmunoabsorption was conducted by using CHO-PA specific monoclonal antibody #354 (MAb#354) . MAb#354 was coupled to CNBr-activated SEPHAROSE 4BTM gel according to the vendor's protocol (Pharmacia Biotech, Piscataway, NJ) . About 10 mg of MAb #354 was coupled to per ml of the CNBr-activated Sepharose 4B gel. After coupling, a MAb#354-SEPHAROSE 4BTM column was packed.
  • CHO cell culture fluid containing secreted CHO-PA was loaded onto a lysme-affinity column pre-equilibrated with an equilibration buffer containing 50 mM sodium phosphate and 0.01% POLYSORBATE 80TM detergent at pH 7.5. After loading, the lysme-affmity column was washed three times: first with the equilibration buffer, followed by a buffer containing 40 mM TRIS, 800 mM NaCl, and 0.008% POLYSORBATE 80TM at pH 8.0, and finally with the equilibration buffer.
  • CHO-PA was then eluted from the lysme affinity column with a buffer containing 50 mM sodium phosphate, 200 mM L-argmme, and 0.01% POLYSORBATE 80TM at pH 7.5.
  • a buffer containing 50 mM sodium phosphate, 200 mM L-argmme, and 0.01% POLYSORBATE 80TM at pH 7.5 After equilibrating with phosphate-buffered saline (PBS, 8 g/L NaCl, 0.2 g/L KC1, 1.44 g/L Na 2 HP0 4 and 0.24 g/L KH 2 P0 4 at pH 7.4), the MAb#354-SEPHAROSETM 4B column was loaded with the lysme-affinity column elution pool.
  • PBS phosphate-buffered saline
  • the column was washed with a buffer containing 9.5 mM Na 2 HP0 4 , 1 M NaCl, and 5% propylene glycol (v/v) at pH 7.4.
  • the bound CHO-PA was eluted from the column with 0.2 M glycme-HCl at pH 2.5.
  • the elution was monitored spectrophotomet ⁇ cally at 280 nm and the CHO-PA- containing fractions were neutralized with 0.14 volumes of 1.5 M argmme-phosphate (pH 8.0) immediately upon collection.
  • the identity and purity of the eluted CHO-PA was confirmed by SDS-PAGE and ammo ac d sequence analysis.
  • DTT/urea treatment The solution containing plasmmogen activator (s) was diluted 1:1 (v/v) with a denaturation buffer (8 M urea, 0.5 M TRIS, and 3.2 mM EDTA at pH 8.4). Dithiothreitol (DTT) was added from a 1 M stock solution to a final concentration of 20 mM, and the mixture was incubated at 37°C for 30 mm. Plasmm treatment
  • the solution containing plasmmogen activator (s) was diluted 1:3 (v/v) with the digestion buffer (125 mM Na 2 HP0 4 , 200 mM argmme, and 0.01% NaN 3 at pH 7.5). One hundredth (w/w) of plasmmogen was added, and the mixture was incubated at 37°C for 30 mm.
  • the assay was performed on a Hewlett-Packard 1090MTM HPLC system (Hewlett Packard, Avondale, PA) with a 4.6 mm x 250 mm, 5- ⁇ m particle size, 300 A pore resm, Zorbax SB-C8 column (Mac-Mod, Chadds Ford, PA) .
  • the column was equilibrated for at least 15 minutes prior to sample injection.
  • the initial mobile phase composition was 70/30/0.1 (v/v/v) of water/acetonitrile/TFA. After a five-mmute initial hold, a linear gradient was performed in 80 minutes (for Figs. 1 and 2) and in 60 minutes (for Fig.
  • ACTIVASE ® r-tPA
  • TNK-tPA TNK-tPA
  • CHO-PA CHO-PA
  • Analytical and preparative methods capable of resolving these three plasmmogen activators from each other or capable of resolving tPA from CHO-PA or TNK-tPA from CHO-PA are needed for recovery process development and to estimate the purity of each molecule for clinical studies and commercial production. Described herein is a simple reverse-phase HPLC method that accomplishes these goals.
  • Plasmm is a ser ne protease with low specificity and is capable of cleaving the Arg 275 -Ile 276 peptide bond m r-tPA, TNK-tPA, and CHO-PA.
  • incubation with plasmmogen converts the single-chain form of the three plasmmogen activators to the two-chain form.
  • FIG. 3 shows the reversed-phase HPLC profiles for the plasmm- and DTT/urea-treated plasmmogen activators.
  • the respective protease fragments of the three proteins were well separated from each other, while the krmgle fragments of the three molecules were not resolved. Therefore, the protease fragment was used for the integration and quantification of each plasmmogen activator.
  • the krmgle fragments from the type I and type II isozymes were well separated.
  • this method is also useful for the quantification of the type I to type II ratio for both r-tPA and TNK-tPA.
  • the availability of this reversed-phase HPLC method greatly facilitates the manufacturing process development. It has been used to evaluate the effect of different fermentation conditions on product quality regarding the integrity of the product (i.e. single chain percent) and the ratio of type I and type II isozymes. It has also been used to aid the purification process development and to ensure consistency between production batches. All those applications exemplify the crucial role of analytical and commercial methods in the development of new pharmaceutics.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Devices For Conveying Motion By Means Of Endless Flexible Members (AREA)
  • Mechanical Treatment Of Semiconductor (AREA)
  • Measuring And Recording Apparatus For Diagnosis (AREA)
  • Signal Processing For Digital Recording And Reproducing (AREA)
  • Glass Compositions (AREA)
  • Transition And Organic Metals Composition Catalysts For Addition Polymerization (AREA)
  • Investigating Or Analysing Materials By The Use Of Chemical Reactions (AREA)
  • Investigating Or Analysing Materials By Optical Means (AREA)
  • Measuring Pulse, Heart Rate, Blood Pressure Or Blood Flow (AREA)

Abstract

A process is described for monitoring the effectiveness of a purification process in removing plasminogen activator (PA) endogenous to Chinese hamster ovary (CHO) cells from a sample containing human tPA or variants thereof. This process comprises incubating the sample with a protease capable of specifically cleaving the Arg275 - Ile276 bond of human wild-type tPA and then with denaturing/reducing agents in respective amounts effective to reduce the disulfide bonds of human wild-type tPA; subjecting the sample to a reversed-phase high-performance liquid chromatography step, and analyzing the elution profile from the chromatography step for the amount of PA endogenous to the CHO cells present therein.

Description

REVE SED-PHASE HPLC ASSAY FOR PLASMINOGEN ACTIVATORS Background of the Invention Field of the Invention
This invention is directed to an assay for determining the amount of Chinese Hamster Ovary (CHO) -produced tPA is present in samples of recombmant human tPA with native sequence or its variants produced n CHO cells. Description of Related Art
Tissue-type plasmmogen activators (tPA) are endogenous seπne proteases involved n a cascade of events leading to the dissolution of a blood clot (Astrup and Permm, Nature, 159, 681-682 (1947); Camiolo et al . , Proc. Soc. Exp. Biol. Med. , 138, 277-280 (1971) ; Collen, J. Biol . Chem. , 33, 77-86 (1987); Hoylaerts et al . , J. Biol. Chem., 257, 2912-2919 (1982)). ACTIVASE0 is the recombmant form of human tPA (r-tPA) , used in the management of acute myocardial infarction and pulmonary embolism (Grossbard, Pharm. Res . , 4., 375-378 (1987)). ACTIVASE0 is also now approved for treating lschemic stroke (Smith et al . , Acad. Emergency Medicine, .6(6), 618-25 (1999); Kwiatkowski et al . , New Eng. J. Med., 340(23), 1781-1787 (1999)). It is a glycoprotem produced by expressing the complementary DNA (cDNA) for natural human tPA m Chinese hamster ovary (CHO) cells. TNK-tPA is a genetically engineered variant of human tPA cloned and expressed in CHO cells (Keyt et al . , Proc. Natl. Acad. Sci USA., 91, 3670-3674 (1994)). Site-directed mutations were introduced at three specific sites of human tPA to create the TNK-tPA variant. They are Thrl03 to Asn (T103N), Asn 117 to Gin (N117Q) , and Lys-His-Arg-Arg 296-299 to Ala-Ala-Ala-Ala (KHRR296-299AAAA) . When compared to tPA, TNK-tPA exhibits similar m vi tro biological activity, an increased resistance to plasmmogen activator inhibitor and an enhanced fibrin specificity, and is cleared more slowly from plasma (Keyt et al . , Proc. Natl. Acad. Sci USA., 91, 3670-3674 (1994); Thomas et al . , Stroke, 25:10, 2072-2079 ( 1994); Benedict et al . , Circulation, 92:10, 3032- 3040 (1995); Modi et al . , Thromb Haemost, 79, 134-139 ( 1998)). It is currently awaiting regulatory approval as a single bolus administered form of r-tPA. CHO cells biosynthesize endogenous hamster tPA called CHO-PA. CHO-PA has a similar fibπnolytic activity to human tPA as determined by the clot lysis assay. The ammo acid sequence of CHO-PA is 80% identical to that of human tPA. Many of the substitutions are semi-conservative such as: Arg<-->Lys, Glu<-->Asp, Phe<-->Tyr, Val<-->Ala, Ile<—>Leu or Thr<—> Ser. Using a model of the human tPA protease domain based upon the bovine chymotrypsm structure, it is observed that virtually all of the substitutions in CHO-PA are localized at or near the protein surface. r-tPA, TNK-tPA, and CHO-PA are all single polypeptide chains composed of 527 ammo acids with 17 disulfide bonds (Nguyen and Carole, "Stability Characterization and Formulation Development of Altepase, a Recombmant Tissue Plasmmogen Activator," in Stability and Characterization of Protein and Peptide Drugs: Case Histories, Y. J. Wang, R. Pearlman, eds . , (Plenum Press: New York, 1993), pp. 91-135. For all three proteins, the peptide bond between Arg275 and Ile276 is particularly susceptible to protease cleavage. The cleavage results two fragments: one consisting of the N-termmal 275 ammo acids and the other consisting of the C-termmal 252 ammo acids. The N-termmal chain contains regions which are homologous to the krmgle regions found in plasmmogen and prothrombm and, therefore, is often referred to as the "krmgle fragment" (Nguyen and Carole, supra ; de Vos et al . , Biochem. , 31, 270-279 (1992) ) .
The C-termmal chain contains the catalytically active site and, therefore, is commonly referred to as the "protease fragment" (Pennica et al . , Nature, 301, 214-221 (1983)). The cleaved two chains are linked by a single disulfide bond formed between Cys264 and Cys395. The cleaved molecule is commonly referred to as "two-chain tPA" as opposed to " single-chain tPA" or the intact form. r-tPA contains four potential sites for N-lmked glycosylation identified by the sequence Asn-X-Ser/Thr (Nguyen and Carole, supra ) . These are Asn117, Asn184, Asn218, and Asn448. r-tPA exists as two glycosylation isozymes designated type I and type II. Type I r-tPA is glycosylated at Asn117, Asn184, and Asn448; whereas type II r-tPA is glycosylated only at Asn117 and Asn448. Asn218 is not glycosylated m either isoforms. TNK-tPA has the same glycosylation pattern as r-tPA, except that the Thrl03 to Asn and Asnll7 to Gin mutations effectively moved the glycosylation site from position 117 to 103 (Keyt et al . , supra) . The glycosylation pattern for CHO-PA is not fully characterized (Riηken and Collen, J. Biol. Chem., 256: 7035-7041 (1981)
ACTIVASE® is a trademark for the recombmant form of human tissue-type plasmmogen activator (r-tPA) , used m the management of acute myocardial infarction and pulmonary embolism. ACTIVASE& brand tPA is also now approved for treating lschemic stroke. It is produced by expressing the complementary DNA (cDNA) for natural human tPA in Chinese hamster ovary (CHO) cells (U.S. Pat. No. 5,753,486) . TNK-tPA is a genetically engineered variant of r-tPA with enhanced efficacy and lower incidence of bleeding compared with ACTIVASE® r-tPA. It was created by three site-directed mutations (T103N, N117Q and KHRR296-299AAAA) , and is also cloned and expressed in CHO cells (U.S. Pat. No. 5,612,029). CHO cells biosynthesize endogenous hamster tPA called CHO-PA. The ammo acid sequence of CHO-PA is highly homologous (80% identical) to that of r-tPA. All three thrombolytic proteins exist as heterogeneous isoforms, mainly due to proteolysis/hydrolysis and differential glycosylation.
A method for purifying human tPA from CHO-PA is described in U.S. Pat. No. 5,411,864. This method comprises contacting a fluid containing the human tPA with antibodies specifically binding the corresponding endogenous CHO-PA and recovering the human tPA. Preferably the contacting step involves passing the fluid through a chromatographic bed having the antibodies immobilized thereon. The development of recombmant DNA-deπved protein pharmaceuticals has been facilitated by the introduction of new analytical methods that can be used to characterize protein and/or to demonstrate consistency of manufacture of a protein. Peptide mapping is a key method for monitoring the ammo acid sequence and is able to detect small changes small to moderate size proteins, for example, insulin and human growth hormone. The analysis of a much larger protein, e.g., fibrmogen (molecular mass of 350,000), or the heterogeneous glycoprotems, such as antibodies (molecular massof 150,000), is hindered by the complexity of the range of peptides generated by an enzymatic digestion. Such complexity makes a single reversed-phase high-performance liquid chromatography (RP-HPLC) separation combined with on-line ultraviolet detection of limited utility.
The advent of commercially available combined HPLC and electrospray lonization mass spectromety (LC-ES-MS) systems compatible with convention HPLC has increased the power of peptide mapping considerably (Ling et al . , Anal . Chem. , 63: 2909-2915 (1991) ; Guzetta et al . , Anal. Chem. , 65: 2953-2962 (1993) ) . LC-EM-MS in combination with m-source collisionally-mduced dissociation (CID) has been used effectively to identify sites of N- and O-lmked glycosylation (Carr et al . , Protein Sci. , 2: 183-196 (1993); Huddleston et al . , Anal. Chem., 65: 877-884 (1993); Conboy and Henion, J. Am. Soc. Mass Spectrom. , 3: 804-814 (1992)). However, even this technique is limited by insufficient resolution resulting from the large number of very similar peptides caused by variable protein glycosylation and enzymatic digests of moderately- sized glycoprotems. It is therefore necessary to employ a range of techniques with orthogonal selectivity to characterize such samples.
The use of combinations of high-performance capillary electrophoresis, HPLC, LC-ES-MS, and matrix-assisted laser desorption lonization-time of flight mass spectrometry has been investigated to allow for characterization of enzymatic digests of undeπvatized glycoprotem samples, as exemplified by DSPA l, a single-chain plasmmogen activator derived from vampire bat salivary glands (Apffel et al . , J. Chromatography A, 717: 41-60 (1995)). It was concluded that these four techniques are highly complimentary techniques for examining glycoprotems. Nonetheless, the authors acknowledge that more work needs to be done to improve the power of this approach, and that high-yield concentration steps will be required due to extensive carbohydrate heterogeneity.
There is a need for a technique to monitor the relative and absolute amounts of CHO-PA present after a purification procedure for tPA is carried out, such as the one reported in U.S. Pat. No. 5,411,864, supra . Summary of the Invention
Accordingly, a reversed-phase HPLC method was developed herein for the analysis of the three thrombolytic molecules, CHO-tPA, recombmant human tPA with native sequence, and TNK-tPA. This method not only has the ability to resolve human tPA and/or TNK-tPA from CHO-PA, but also is capable of identifying and quantifying different isoforms of each molecule.
Specifically, the present invention provides a process for monitoring the effectiveness of a purification process in removing plasmmogen activator (PA) endogenous to Chinese hamster ovary (CHO) cells from a sample containing human tPA or variants thereof, which process comprises incubating the sample with a protease capable of specifically cleaving the Arg275 - Ile276 bond of human wild- type tPA and then with denaturing and reducing agents in amounts effective to reduce the disulfide bonds of human wild-type tPA; subjecting the sample to a reversed-phase high-performance liquid chromatography step, and analyzing the elution profile from the chromatography step for the amount of PA endogenous to the CHO cells present therein.
Brief Description of the Drawings Figure 1 shows a reversed-phase HPLC analysis of native r-tPA, TNK-tPA and CHO-PA.
Figure 2 shows a reversed-phase HPLC analysis of DTT/urea treated r-tPA, TNK-tPA and CHO-PA. Plasmmogen activators were treated with DTT/urea prior to chromatography .
Figure 3 shows a reversed-phase HPLC analysis of plasmm and DTT/urea treated r-tPA, TNK-tPA and CHO-PA. Plasmmogen activators were subjected to plasmm treatment followed by DTT/urea treatment prior to chromatography.
Description of the Preferred Embodiments Definitions
The terms "tissue plasmmogen activator", and "tPA" refer to human extrinsic (tissue-type) plasmmogen activator having fibr olytic activity that typically has a structure with five domains (finger, growth factor, krmgle-1, krmgle-2, and protease domains) , but nonetheless may have fewer domains or may have some of its domains repeated if it still functions as a thrombolytic agent and retains the N-lmked glycosylation sites at positions 117, 184, and 448. At minimum, the tPA consists of a protease domain that is capable of converting plasmmogen to plasmm, and an N-termmal region believed to be at least partially responsible for fibrm binding, and retains the N-lmked glycosylation sites at positions corresponding to ammo acid positions 117, 184, and 448 of wild-type human tPA. The retention of these glycosylation sites is due to the fact that variable site occupancy of recombmant and melanoma-derived wild-type tPA leads to production of two variants, designated as "Type I tPA" and "Type
II tPA" , respectively. Type I tPA contains N-lmked oligosacchaπdes at positions 117, 184, and 448. Type II tPA contained N-lmked oligosaccharides at positions 117 and 448. It will be understood that natural allelic variations exist and can occur among individuals, as demonstrated by one or more ammo acid differences in the ammo acid sequence of tPA of each individual.
The terms "wild-type human tissue plasmmogen activator", "wild-type human tPA" , "native human tissue plasmmogen activator," and "native human tPA", where "human tPA" may be abbreviated as "htPA", refer to native-sequence human tPA, i.e., that encoded by the cDNA sequence reported in U.S. Pat. No. 4,766,075, issued 23 August 1988. Ammo acid site numbers or positions m the tPA molecule are labeled in accordance with U.S. Pat. No. 4,766,075. As used herein, references to various domains of tPA mean the domains of wild-type human tPA as heremabove defined, and functionally equivalent portions of human tPA having ammo acid alterations as compared to the native human tPA sequence, or of (native or variant) tPA from other sources, such as bat tissue plasmmogen activator (bat-PA) . Thus, as used herein, the term "protease domain" refers to the region extending from ammo acid position 264 to ammo acid position 527, inclusive, of the mature form of wild-type human tPA, and to functionally equivalent portions of human tPA having ammo acid alterations as compared to the native human tPA sequence, or of tPA from other sources, such as bat-PA. As used herein, "tPA variants" refers to molecules that differ from native tPA by one or more amino acid changes or modifications to existing ammo acids. TNK-tPA is the preferred variant herein. The modification to change or insert the appropriate ammo acιd(s) n the native molecule to effect the desired sequence variations is accomplished by any means known m the art, such as e.g. site-directed mutagenesis or ligation of the appropriate sequence into the DNA encoding the relevant protein.
As used herein, "TNK-tPA" refers to a tPA molecule wherein Thrl03 of wild- type tPA is changed to Asn (T103N) , Asnll7 of wild-type tPA is changed to Gin (N117Q) , and Lys-His-Arg-Arg 296-299 of wild-type tPA is changed to Ala-Ala-Ala- Ala (KHRR296-299AAAA) . Such TNK is further described n U.S. Pat. No. 5,612,029.
The term "Chinese hamster ovary cell" or "CHO cell" refers to cells or cell lines derived from Chinese hamster ovaries, as described, for example, in EP 117,159, published August 29, 1989; U.S. Pat. Nos. 4,766,075; 4,853,330; 5,185,259; Lubmiecki et al . , in Advances m Animal Cell Biology and Technology for Bioprocesses, Spier et al . , eds . (1989), pp. 442-451), as well as CHO derivatives such as CHO/-DHFR (Urlaub and Chasm, Proc. Natl. Acad. Sci. USA, 77: 4216 (1980)), CHO-Kl DUX Bll (Simonsen and Levmson, Proc. Natl. Acad. Sci. USA, 80: 2495-2499 (1983); Urlaub and Chasm, supra ) , and dpl2.CH0 cells (EP 307,247 published 15 March 1989) Preferred host cells include CHO-Kl DUX Bll and dpl2.CHO cells.
The CHO cells developed for large-scale production of tPA are maintained cryogenically in a MCB/workmg cell bank (WCB) system as described by Wiebe et al . , n Large Scale Mammalian Cell Culture Technology, Lubmiecki, ed., (Marcel Dekker: New York, 1990), pp. 147-160. DHFR+ CHO-Kl cells transfected with DNA encoding human tPA have been deposited at the American Type Culture Collection, Manassas, Virginia (ATCC) , and are available under accession number CCL 61. A sample of another tPA-producmg CHO cell line (CHO cell line 1-1515) has been deposited under ATCC accession number CRL 9606. The latter cell line was reported to result in human tPA levels approaching 50 pg/cell/day.
As used herein "CHO plasmmogen activator" or "CHO-PA" refers to plasmmogen activator that is produced endogenously by CHO cells. This endogenous PA expressed by CHO cells has a sequence slightly different (about
80% identical) from the human wild-type tPA. The CHO-PA is not a tissue-type
PA.
As used herein, "protease" refers to an enzyme that is capable of cleaving the Arg27S - Ile276 bond of human wild-type tPA specifically. Examples include plasmm (or plasmmogen, which converts to plasmm), tissue kallikrem, or Factor Xa, as well as any trypsm-like proteases that can effect this specific, limited proteolysis. Eligible proteases are further described in Ichmosi et al . , FEBS Letters, 175: 412-418 (1984). Preferred herein is plasmm/plasmmogen . As used herein, "denaturing/reducing agents" or "denaturing agent and reducing agent" refers to a combination of denaturant and reductant that reduces the disulfide bonds of human wild-type tPA. Preferably, the denaturing agent is guanidme or urea and the reducing agent is dithiothreitol (DTT) or 2- mercaptoethanol . Modes for Carrying Out the Invention
After recombmant production, the tPA or tPA variant is recovered from the CHO culture medium, either as a secreted protein or from host cell lysates when directly expressed without a secretory signal. It is necessary to purify the tPA or variant thereof from host cell proteins to obtain preparations that are substantially homogeneous as to protein. As a first step, the culture medium or lysate is centrifuged or filtered to remove particulate cell debris.
The human tPA or variant thereof is then purified from corresponding contaminant endogenous proteins such as CHO-PA by such techniques as fractionation on lmmunoaffmity or ion- exchange columns as described, for example, m U.S. Pat. No. 5,411,864; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation exchange resm such as DEAE; chromatofocus g; SDS-PAGE; ammonium sulfate precipitation; or gel electrophoresis using, for example, Sephadex G-75. A protease inhibitor that does not interfere with the tPA activity such as phenylmethylsulfonyl fluoride (PMSF) also may be useful to inhibit proteolytic degradation during purification, and antibiotics may be included to prevent the growth of adventitious contaminants. One skilled m the art will appreciate that purification methods suitable for native tPA may require modification to account for changes m the character of tPA or its variants upon expression in recombmant cell culture.
In a preferred embodiment, if a tPA variant is being produced, it is secreted and the supernatant is passed over a PBS-preconditioned column of glass beads coupled to anti-tPA goat polyclonal A6 antibody, the column is equilibrated with a buffer, and the tPA variant is then eluted.
The invention herein is directed to monitoring (including qualifying and quantifying) levels of native CHO-PA in a sample taken from such purification systems that contains at least one form of human tPA that is produced in CHO cells. The process comprises incubating the sample with a protease that is capable of cleaving Arg275 - Ile276 bond specifically. This is followed by incubation of the protease-treated sample with a combination of a denaturing and reducing agent in proper relative and absolute amounts to effect reduction of the disulfide bonds m human wild-type tPA. Since the treatment with denaturing and reducing agents causes the loss of enzyme activity, the incubation with protease occurs first.
After incubation, the sample is subjected to a reversed-phase high- performance liquid chromatography step, and the elution profile from the chromatography step analyzed for the amount of PA endogenous to the CHO cells present therein. Preferably, the protease is plasmmogen, which converts to the active form, plasmm, the human tPA is native-sequence tPA, and the tPA variant
The consecutive incubation step with the protease followed by the denaturing/reducing agents typically takes place at a temperature of about 30- 40°C, more preferably about 36-38°C, and most preferably about 37°C, for a minimum of about 15 minutes, more preferably about 20-40 minutes.
Also, preferably before incubation, the sample is diluted with a digestion buffer, which preferably has a pH of about 7 to 8, more preferably phosphate buffer at pH 7.4-7.6, and more preferably also containing argin ne.
Any suitable HPLC column on which the sample is loaded may be utilized for the purposes of this invention, including preparative or analytical scale. The column is typically equilibrated for at least about 15 minutes prior to sample injection. Column size, column material, flow rate, elution buffers, type of gradient, injection volume, and particle size of column αepend on various factors, including the size of the sample being examined, the type of mobile phase composition and gradient, and the forms of tPA being distinguished.
The loading solvent may be any solvent but is preferably an acetonitrile- based solvent such as water, acetonitπle, and tπfluoroacetic acid (TFA) . Preferably, the column is a Zorbax C8, Vydac, or Baker C-18 column packed with a medium having a particle diameter of about 4-40 μm, more preferably about 5-15 μm, and a pore size of about 100-4000 A, more preferably about 150-350 A. Also, the medium preferably has a C4, C8, or C18 alkyl group, ana most preferably is a C8 silica medium. Preferably, the elution is carried out with a solvent comprising acetonitπle, such as water, acetonitπle, and TFA, in a gradient format over 60-100 minutes, preferably a linear gradient, wherein the relative amount of acetonitπle is increased in the solvent. In another preferred embodiment, a shallow gradient ramp at about 0.25% acetonitrile per mmute is employed.
If the analysis for purity herein indicates that the technique employed successfully removes CHO-PA, further purification steps can be carried out as necessary to remove any other contaminants. If the technique did not successfully remove CHO-PA to acceptable levels, a different purification scheme can be utilized and the process herein repeated to determine how effective that scheme is.
After final purification, the tPA or variant thereof can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the tPA product is combined in admixture with a pharmaceutically acceptable carrier. Such formulations are well described in the literature as well as dosages and uses. For example, the tPA or its variant is suitably administered parenterally to subjects suffering from cardiovascular diseases or conditions and strokes.
The following examples are intended to illustrate one embodiment now known for practicing the invention, but the invention is not to be considered limited to these examples. All open and patented literature citations herein are expressly incorporated by reference. EXAMPLE 1
MATERIALS
ACTIVASE® (r-tPA) and TNK-tPA were obtained from Genentech, Inc. (South San Francisco, CA) m a form purified from CHO cells. See also, for example, U.S. Pat. Nos. 4,766,075 and 5,753,486 for ACTIVASE® r-tPA and US Pat. No. 5,612,029 for TNK-tPA.
Monoclonal antibody #354 for CHO-PA was produced as described in U.S. Pat. No. 5,411,864. Briefly, a female Balb/c mouse was immunized over a period of 12 weeks with protein solutions substantially enriched in CHO plasmmogen activator purified from host cell lacking the human t-PA. There were five injections each consisting of approximately 30 μg . The initial injection was emulsified with complete Freund's adjuvant and administered in subcutaneous sιte(s). The second injection given 1.5 weeks later was emulsified with incomplete Freund's adjuvant and half was administered subcutaneously and half mtraperitoneally . The remaining three injections were given on weeks 3, 6 and 12 in phosphate buffered saline (PBS) administered in one mtraperitoneal site.
The spleen from the immunized mouse was removed on week 13 and spleen cells were fused with the mouse myeloma cell line NP3X63-Ag8.653 using the general procedures of Fazekas et al . , J . Immunol. Methods, 35 : 1 (1980) and
Lane, J. Immunol. Methods 81 : 223 (1985) . The fused cells were distributed into ten microtiter plates each containing 96 wells. Each well was screened for specific antibody production using differential reactivity m two ELISA's (enzyme linked lmmunoadsorbant assays) . One ELISA specifically detected antibodies against CHO-PA and the second detected antibodies that cross-reacted with recombmant human tPA.
Approximately 5% of the total wells were reactive only with CHO-PA, and 3% reacted with CHO-PA and human t-PA. Hybridoma cells from wells containing CHO-PA-specifIC antibodies were expanded and cloned by limiting dilution (Oi and Herzenberg, "Immunoglobm-Producmg Hybrid Cell Lines", p. 351-372, in Selected Methods m Cellular Immunology, Mishell and Shngi, eds . (W. H. Freeman and Co., 1980)). Large quantities of specific monoclonal antibodies were produced by cell culture of the hybridoma cells or by injection of hybridoma cells in mice thereby producing ascites tumors . MAb 354 was one resulting antibody, which lowered CHO-PA levels greater than about 100 fold m a single column pass when immobilized and is stable to several different immobilization chemistries and harsh washing conditions. MAb 354 was purified using the steps set forth below, where all steps were carried out at room temperature. Concentration was carried out as follows: The MAb hybridoma suspension culture harvest fluid (HF) was filtered through a 0.2 μm filter. The culture fluid was concentrated by ultraflltration or by chromatography on an ion-exchange resin. Affinity purification was carried out as follows: Thimerosal was added to the concentrated MAb HF solution to approximately 0.02%. The solution was adjusted to approximately 1.5 M glycme, 3.0 M NaCl, pH 9.0 by addition of 3.0 M glycme, followed by addition of crystalline NaCl. Protein A has poor Ab binding, so addition of NaCl and glycme increases hydrophobic interaction, so as to facilitate Ab binding. The solution was clarified by filtration. The clarified concentrated MAb HF was applied to a column of protein A immobilized to agarose. The bound MAb was washed with the buffer used to equilibrate the column, having the approximate composition of 1.5 M glycme, 3.0 M NaCl, 0.02 M EDTA, pH 9.0. The MAb was eluted with 0.1 M sodium citrate, 0.15 M NaCl, pH 3.0 buffer. The protein A column was regenerated by washing with 3.0 M NaSCN, 0.03 M TRIS, pH 8.5 and re-equilibrated. The eluted MAb peak was collected based on absorbance profile at 280 nm. The citrate-eluted MAb peak was immediately neutralized by collection into buffer with the approximate composition: 1.5 M TRIS-HC1, pH 9.0. After use, the protein A column was unpacked and stored in sealed containers at 2-8°C in approximately 0.02% thimerosal as storage buffer.
The 354 MAb was then buffer exchanged by diaflltration . Diaflltration proceeded until the conductivity and pH were similar to the values for 0.03 M TRIS, 0.05 M NaCl, pH 8.5 buffer. The MAb was subsequently applied to an amon-exchange chromatograpny column containing DEAE-FAST FLOW™ agarose support and washed with 0.03 M TRIS, 0.05 M NaCl, pH 8.5 buffer. The MAb was step eluted with a buffer having the approximate composition: 0.03 M TRIS, 0.15 M NaCl, pH 8.5. The eluted MAb peak was collected based on absorbance profile at 280 nm.
The MAb was filtered (0.2 μm) into sanitized sealable containers and stored below -40°C. Plasmmogen was obtained from Fluka (Switzerland) . HPLC-grade acetonitrile was obtained from Burdick & Jackson (Muskgon, MI) and tπfluoroacetic acid (TFA) was from Pierce (Rockford, IL) . Water for the HPLC mobile phase and sample solutions was purified with a MILLI-Q™ system from Millipore (Milford, MA) . All other chemicals were of reagent grade from Sigma (St. Louis, MO). METHODS
Purification of CHO-PA
CHO-PA was isolated from CHO cell culture fluid by affinity chromatography followed by lmmunoabsorption. Lysme hyper D resm from BioSepra (Paris, France) was used for the affinity chromatography, as lysme binds to the krmgle 2 region of plasmmogen activators (Cleary et al . , Biochem. 28, 1884-1890 (1989) ) . The lmmunoabsorption was conducted by using CHO-PA specific monoclonal antibody #354 (MAb#354) . MAb#354 was coupled to CNBr-activated SEPHAROSE 4B™ gel according to the vendor's protocol (Pharmacia Biotech, Piscataway, NJ) . About 10 mg of MAb #354 was coupled to per ml of the CNBr-activated Sepharose 4B gel. After coupling, a MAb#354-SEPHAROSE 4B™ column was packed.
CHO cell culture fluid containing secreted CHO-PA was loaded onto a lysme-affinity column pre-equilibrated with an equilibration buffer containing 50 mM sodium phosphate and 0.01% POLYSORBATE 80™ detergent at pH 7.5. After loading, the lysme-affmity column was washed three times: first with the equilibration buffer, followed by a buffer containing 40 mM TRIS, 800 mM NaCl, and 0.008% POLYSORBATE 80™ at pH 8.0, and finally with the equilibration buffer. CHO-PA was then eluted from the lysme affinity column with a buffer containing 50 mM sodium phosphate, 200 mM L-argmme, and 0.01% POLYSORBATE 80™ at pH 7.5. After equilibrating with phosphate-buffered saline (PBS, 8 g/L NaCl, 0.2 g/L KC1, 1.44 g/L Na2HP04 and 0.24 g/L KH2P04 at pH 7.4), the MAb#354-SEPHAROSE™ 4B column was loaded with the lysme-affinity column elution pool. After loading, the column was washed with a buffer containing 9.5 mM Na2HP04, 1 M NaCl, and 5% propylene glycol (v/v) at pH 7.4. The bound CHO-PA was eluted from the column with 0.2 M glycme-HCl at pH 2.5. The elution was monitored spectrophotometπcally at 280 nm and the CHO-PA- containing fractions were neutralized with 0.14 volumes of 1.5 M argmme-phosphate (pH 8.0) immediately upon collection. The identity and purity of the eluted CHO-PA was confirmed by SDS-PAGE and ammo ac d sequence analysis. DTT/urea treatment The solution containing plasmmogen activator (s) was diluted 1:1 (v/v) with a denaturation buffer (8 M urea, 0.5 M TRIS, and 3.2 mM EDTA at pH 8.4). Dithiothreitol (DTT) was added from a 1 M stock solution to a final concentration of 20 mM, and the mixture was incubated at 37°C for 30 mm. Plasmm treatment
The solution containing plasmmogen activator (s) was diluted 1:3 (v/v) with the digestion buffer (125 mM Na2HP04, 200 mM argmme, and 0.01% NaN3 at pH 7.5). One hundredth (w/w) of plasmmogen was added, and the mixture was incubated at 37°C for 30 mm.
Reversed-phase HPLC assay for plasmmogen activators
The assay was performed on a Hewlett-Packard 1090M™ HPLC system (Hewlett Packard, Avondale, PA) with a 4.6 mm x 250 mm, 5-μm particle size, 300 A pore resm, Zorbax SB-C8 column (Mac-Mod, Chadds Ford, PA) . The column was equilibrated for at least 15 minutes prior to sample injection. The initial mobile phase composition was 70/30/0.1 (v/v/v) of water/acetonitrile/TFA. After a five-mmute initial hold, a linear gradient was performed in 80 minutes (for Figs. 1 and 2) and in 60 minutes (for Fig. 3) to 50/50/0.1 (v/v/v) of water/acetonitrile/TFA. Immediately following the gradient, the column was regenerated for 10 minutes with 100/0.1 (v/v) of acetonitπle/TFA. The composition was then brought back to the initial conditions m 5 minutes, and the system was re-equilibrated for the next injection. The injection volume was 250 rtiL, and the flow rate was 1 mL/min. The chromatography was conducted at 40°C. Fluorescence was measured with a Hewlett-Packard 1046A™ programmable fluorescence detector (Ex = 275 nm and Em = 340 nm) . The chromatograms were recorded and analyzed with Hewlett-Packard CHEMSTATION™ software. RESULTS AND DISCUSSION
Due to high sequence homology, ACTIVASE® (r-tPA) , TNK-tPA, and CHO-PA have very similar biochemical/biophysical properties. Analytical and preparative methods capable of resolving these three plasmmogen activators from each other or capable of resolving tPA from CHO-PA or TNK-tPA from CHO-PA are needed for recovery process development and to estimate the purity of each molecule for clinical studies and commercial production. Described herein is a simple reverse-phase HPLC method that accomplishes these goals. With a shallow gradient ramp at 0.25% acetonitrile per mmute and no protease or denaturing/reducing agents, reversed-phase HPLC was not able to separate the native form of r-tPA from native CHO-PA (Figure 1). However, the native form of TNK-tPA was resolved very well from both native r-tPA and CHO-PA under these conditions. Next, DTT/urea treatment was performed to reduce the disulfide bonds and denature the proteins. For all three proteins, the peptide bond between Arg275 and Ile276 is very susceptible to protease cleavage. Over time, this susceptibility leads to heterogeneity for r-tPA, TNK-tPA, and CHO-PA m solution. A small amount of the single-chain form is converted to the two- chain form due to the protease cleavage. DTT/urea treatment reduces the disulfide bond between Cys264 and Cys395 that holds the two-chain form of the molecule together, resulting m the dissociation of the molecule into two fragments (the krmgle fragment and the protease fragment) . Figure 2 shows that, under the same gradient ramp, reversed-phase HPLC was able to resolve the single-chain form of the three thrombolytic molecules from each other after DTT/urea treatment. All three proteins exhibited similar elution profiles with the krmgle fragment of the two-chain form eluting first, the single-chain form eluting second, and the protease fragment of the two-chain form eluting last. The respective protease fragments of the three plasmmogen activators were also well resolved from each other, while the krmgle fragments for the three molecules were not well separated. Consequently, this method can be used to detect and quantify the fragmentation of the single-chain form into the two-chain form of the plasmmogen activators.
The heterogeneity observed in the r-tPA, TNK-tPA, and CHO-PA profiles (Figure 2) makes the quantification of these molecules very difficult, especially when trying to quantify each individual molecule in a mixture of rtPA, TNK-tPA, and CHO-PA. To eliminate the heterogeneity associated with proteolysis, all of the single-chain form was converted to two-chain form by incubating with plasmmogen. Plasmmogen is the substrate of plasmmogen activator m the natural fibrmolytic system. r-tPA, TNK-tPA, and CHO-PA all have the enzymatic activity of cleaving the Arg560-Val561 peptide bond of plasmmogen. Such cleavage converts plasmmogen into its active form, plasmm. Plasmm is a ser ne protease with low specificity and is capable of cleaving the Arg275-Ile276 peptide bond m r-tPA, TNK-tPA, and CHO-PA. As a result, incubation with plasmmogen converts the single-chain form of the three plasmmogen activators to the two-chain form.
Following the plasmm treatment, the samples were treated with DTT/urea to reduce disulfide bonds and thus dissociate the two-chain form of the molecule into two discrete fragments. Figure 3 shows the reversed-phase HPLC profiles for the plasmm- and DTT/urea-treated plasmmogen activators. The respective protease fragments of the three proteins were well separated from each other, while the krmgle fragments of the three molecules were not resolved. Therefore, the protease fragment was used for the integration and quantification of each plasmmogen activator. For both r-tPA and TNK-tPA, the krmgle fragments from the type I and type II isozymes were well separated. As a result, this method is also useful for the quantification of the type I to type II ratio for both r-tPA and TNK-tPA. The availability of this reversed-phase HPLC method greatly facilitates the manufacturing process development. It has been used to evaluate the effect of different fermentation conditions on product quality regarding the integrity of the product (i.e. single chain percent) and the ratio of type I and type II isozymes. It has also been used to aid the purification process development and to ensure consistency between production batches. All those applications exemplify the crucial role of analytical and commercial methods in the development of new pharmaceutics.

Claims

WHAT IS CLAIMED IS:
1. A process for monitoring the effectiveness of a purification process m removing plasmmogen activator (PA) endogenous to Chinese hamster ovary (CHO) cells from a sample containing human tPA or variants thereof, which process comprises mcubatmg the sample with a protease capable of specifically cleaving the Arg275 - Ile276 bond of human wild-type tPA and then with a denaturing agent and a reducing agent in amounts effective to reduce the disulfide bonds of human wild-type tPA; subjecting the sample to a reversed-phase high-performance liquid chromatography step, and analyzing the elution profile from the chromatography step for the amount of PA endogenous to the CHO cells present therein.
2. The process of claim 1 wherein the protease is plasmmogen.
3. The process of claim 1 wherein the human tPA is native-sequence tPA.
4. The process of claim 1 wherein the tPA variant is TNK-tPA.
5. The process of claim 1 wherein before incubation, the sample is diluted with a digestion buffer.
6. The process of claim 5 wherein the buffer has a pH of about 7 to 8.
7. The process of claim 6 wherein the buffer is a phosphate buffer.
8. The process of claim 7 wherein the buffer further comprises argmme.
9. The process of claim 1 wherein the denaturing agent comprises urea or guanidme.
10. The process of claim 1 wherein the reducing agent comprises dithiothreitol or 2-mercaptoethanol.
11. The process of claim 1 wherein the chromatography step is carried out by eluting with a solvent comprising acetonitrile m a gradient format.
PCT/US2000/030252 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators WO2001032915A2 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
AT00978343T ATE243263T1 (en) 1999-11-04 2000-11-01 TEST PROCEDURE
BRPI0015283 BRPI0015283B8 (en) 1999-11-04 2000-11-01 Process for monitoring the effectiveness of a purification process.
DK00978343T DK1226269T3 (en) 1999-11-04 2000-11-01 Assay
CA2384756A CA2384756C (en) 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators
SI200030178T SI1226269T1 (en) 1999-11-04 2000-11-01 Assay
AU15816/01A AU781023B2 (en) 1999-11-04 2000-11-01 Reversed-phase HPLC assay for plasminogen activators
JP2001535595A JP4659320B2 (en) 1999-11-04 2000-11-01 Reversed phase HPLC assay of plasminogen activator
DE60003449T DE60003449T2 (en) 1999-11-04 2000-11-01 TEST METHODS
NZ518187A NZ518187A (en) 1999-11-04 2000-11-01 Reversed-phase HPLC assay for plasminogen activators
IL14900600A IL149006A0 (en) 1999-11-04 2000-11-01 Reversed-phase hplc for plasminogen activators
HU0202973A HU226202B1 (en) 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators
EP00978343A EP1226269B1 (en) 1999-11-04 2000-11-01 Assay
MXPA02004401A MXPA02004401A (en) 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators.
PL355389A PL204011B1 (en) 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators
NO20022122A NO329876B1 (en) 1999-11-04 2002-05-03 Method for monitoring the efficiency of a purification process by removing endogenous plasminogen activator (PA) from Chinese hamster ovary (CHO) cells, from a sample containing human tPA or a tPA molecule in which Thr103 from human wild-type tPA is changed to Asn, Asn117 from human wild type tPA is changed to Gln and Lys-His-Arg-Arg from human wild type tPA is changed to Ala-Ala-Ala-Ala at positions 296-299
HK02107782.5A HK1046158B (en) 1999-11-04 2002-10-28 Assay

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16360799P 1999-11-04 1999-11-04
US60/163,607 1999-11-04

Publications (2)

Publication Number Publication Date
WO2001032915A2 true WO2001032915A2 (en) 2001-05-10
WO2001032915A3 WO2001032915A3 (en) 2002-03-07

Family

ID=22590762

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/030252 WO2001032915A2 (en) 1999-11-04 2000-11-01 Reversed-phase hplc assay for plasminogen activators

Country Status (24)

Country Link
US (2) US6593097B1 (en)
EP (1) EP1226269B1 (en)
JP (1) JP4659320B2 (en)
KR (1) KR100649043B1 (en)
CN (1) CN1167808C (en)
AT (1) ATE243263T1 (en)
AU (1) AU781023B2 (en)
BR (1) BRPI0015283B8 (en)
CA (1) CA2384756C (en)
CZ (1) CZ302023B6 (en)
DE (1) DE60003449T2 (en)
DK (1) DK1226269T3 (en)
ES (1) ES2200973T3 (en)
HK (1) HK1046158B (en)
HU (1) HU226202B1 (en)
IL (1) IL149006A0 (en)
MX (1) MXPA02004401A (en)
NO (1) NO329876B1 (en)
NZ (1) NZ518187A (en)
PL (1) PL204011B1 (en)
PT (1) PT1226269E (en)
SI (1) SI1226269T1 (en)
WO (1) WO2001032915A2 (en)
ZA (1) ZA200202687B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1885488A1 (en) * 2005-05-24 2008-02-13 GE Healthcare Bio-Sciences AB Regeneration of a chromatography matrix

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4729222B2 (en) 1999-12-10 2011-07-20 ライフ テクノロジーズ コーポレーション Use of multiple recombination sites with unique selectivity in recombination cloning
EP1349917B1 (en) 2000-12-08 2010-03-10 Life Technologies Corporation Methods and compositions for synthesis of nucleic acid molecules using multiple recognition sites
US6972327B1 (en) * 2001-05-08 2005-12-06 Immunex Corporation Regeneration of chromatography material
KR100701265B1 (en) * 2006-01-16 2007-04-02 대한민국 Method for generating transgenic pig secreting human tissue-plasminogen activator in its urine
MX2009002576A (en) * 2006-09-08 2009-03-20 Wyeth Corp Arginine wash in protein purification using affinity chromatography.
EP2089710B1 (en) * 2006-10-27 2014-10-01 Janssen Pharmaceutica NV A method for pharmacologically profiling compounds
CN101539550B (en) * 2009-04-28 2012-05-23 李绍平 Qualitative and quantitative analysis method for polyoses
US20200017771A1 (en) 2017-03-09 2020-01-16 Universitaet Des Saarlandes Means and methods for lignin pyrolysis
CN108333281A (en) * 2018-02-08 2018-07-27 吉林大学 A method of measuring recombinant human granulocyte colony stimulating factor disulfide bond composition
CN108918749B (en) * 2018-07-17 2020-05-05 北京赛升药业股份有限公司 Reversed phase HPLC detection method of snake venom fibrinolytic enzyme
CN114426962B (en) * 2021-12-21 2024-06-18 佛山汉腾生物科技有限公司 T-PA purification method

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989004367A1 (en) * 1987-11-05 1989-05-18 Genentech, Inc. Method of purifying recombinant proteins from corresponding host cell proteins

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FI831484L (en) 1982-05-05 1983-11-06 Genentech Inc PLASMINOGEN AKTIVATOR FOER MAENSKOVAEVNAD
US4766075A (en) 1982-07-14 1988-08-23 Genentech, Inc. Human tissue plasminogen activator
US4777043A (en) * 1985-12-17 1988-10-11 Genentech, Inc. Stabilized human tissue plasminogen activator compositions
US5120535A (en) * 1986-11-26 1992-06-09 Oncogen Oncostatin M and novel compositions having anti-neoplastic activity
GB8919803D0 (en) * 1989-09-01 1989-10-18 Ciba Geigy Pharmaceutical compositions
ATE155816T1 (en) 1992-06-03 1997-08-15 Genentech Inc VARIANTS OF TISSUE PLASMINOGEN ACTIVATOR WITH IMPROVED THERAPEUTIC EFFECTS
US5753702A (en) * 1996-05-22 1998-05-19 University Of Vermont Arachidonic acid metabolite, 16-hete

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1989004367A1 (en) * 1987-11-05 1989-05-18 Genentech, Inc. Method of purifying recombinant proteins from corresponding host cell proteins

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ANCINETTI, V AND HANCOCK,WS: "Analytical considerations in the development of protein purification processes" BIOPROCESS. TECHNOL., vol. 18, 1994, pages 11-36, XP001006204 *
APFFEL A ET AL: "Application of new analytical technology to the production of a "well-characterized biological"." DEVELOPMENTS IN BIOLOGICAL STANDARDIZATION, vol. 96, 1998, pages 11-25, XP001006250 Symposium;Washington, D.C., USA; December 11-13, 1995, biotechnology pharmaceutical products. 1998 S. Karger AG; S. Karger AG P.O. Box, Allschwilerstrasse 10, CH-4009 Basel, Switzerland; New York, New York, USA ISBN: 3-8055-6805-3 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1885488A1 (en) * 2005-05-24 2008-02-13 GE Healthcare Bio-Sciences AB Regeneration of a chromatography matrix
EP1885488B1 (en) * 2005-05-24 2015-07-08 GE Healthcare Bio-Sciences AB Regeneration of a chromatography matrix

Also Published As

Publication number Publication date
NO329876B1 (en) 2011-01-17
US6828118B2 (en) 2004-12-07
HU226202B1 (en) 2008-06-30
CN1167808C (en) 2004-09-22
HUP0202973A2 (en) 2002-12-28
CA2384756C (en) 2010-09-28
PL204011B1 (en) 2009-12-31
CZ302023B6 (en) 2010-09-08
NO20022122L (en) 2002-07-03
MXPA02004401A (en) 2002-09-02
BRPI0015283B1 (en) 2015-03-17
DE60003449T2 (en) 2004-05-06
SI1226269T1 (en) 2003-12-31
JP2003513638A (en) 2003-04-15
US6593097B1 (en) 2003-07-15
KR100649043B1 (en) 2006-11-27
BRPI0015283B8 (en) 2021-07-06
CA2384756A1 (en) 2001-05-10
ZA200202687B (en) 2003-06-25
PT1226269E (en) 2003-11-28
WO2001032915A3 (en) 2002-03-07
DE60003449D1 (en) 2003-07-24
CZ20021196A3 (en) 2002-08-14
US20030199016A1 (en) 2003-10-23
HK1046158B (en) 2003-11-28
NZ518187A (en) 2002-12-20
KR20020070431A (en) 2002-09-09
HUP0202973A3 (en) 2005-11-28
PL355389A1 (en) 2004-04-19
NO20022122D0 (en) 2002-05-03
IL149006A0 (en) 2002-11-10
HK1046158A1 (en) 2002-12-27
CN1387579A (en) 2002-12-25
EP1226269A2 (en) 2002-07-31
AU1581601A (en) 2001-05-14
JP4659320B2 (en) 2011-03-30
ATE243263T1 (en) 2003-07-15
ES2200973T3 (en) 2004-03-16
AU781023B2 (en) 2005-04-28
EP1226269B1 (en) 2003-06-18
DK1226269T3 (en) 2003-10-13
BRPI0015283A (en) 2002-07-02

Similar Documents

Publication Publication Date Title
Lundblad et al. Preparation and partial characterization of two forms of bovine thrombin
White et al. The isolation and characterization of plasminogen activators (urokinase) from human urine
Boissel et al. Covalent structures of beta and gamma autolytic derivatives of human alpha-thrombin.
EP1226269B1 (en) Assay
EP1841863B1 (en) Method for purification of factor vii
Do et al. Characterization of pure human renal renin. Evidence for a subunit structure.
Winkler et al. Purification and characterization of recombinant single-chain urokinase produced in Escherichia coli
HOLVOET et al. Characterization of functional domains in human tissue‐type plasminogen activator with the use of monoclonal antibodies
Robbins et al. The Primary Structure of Human Plasminogen: I. THE NH2-TERMINAL SEQUENCES OF HUMAN PLASMINOGEN AND THE S-CARBOXYMETHYL HEAVY (A) AND LIGHT (B) CHAIN DERIVATIVES OF PLASMIN
Lim et al. Affinity purification and enzymatic cleavage of inter-alpha inhibitor proteins using antibody and elastase immobilized on CIM monolithic disks
van Zonneveld et al. Mapping of epitopes on human tissue-type plasminogen activator with recombinant deletion mutant proteins
Wiman et al. Amino‐Acid Sequence of the Cyanogen‐Bromide Fragment from Human Plasminogen that Forms the Linkage between the Plasmin Chains
Nguyen et al. Stability characterization and formulation development of alteplase, a recombinant tissue plasminogen activator
KR100583537B1 (en) Monoclonal antibody which is specific for activated coagulation factor VII, and its use
Pohl et al. Tissue plasminogen activator mutants lacking the growth factor domain and the first kringle domain: I: DNA Constructions, Expression in Mammalian Cells, Protein Structure, Fibrin Affinity and Enzymatic Properties
Reddy et al. Differential autolysis of human and canine plasmins
WO1989009820A1 (en) PURIFIED TYPE I AND TYPE II tPA
Xu et al. A reversed-phase HPLC assay for plasminogen activators
Opdenakker et al. Heterogeneity of human tissue-type plasminogen activator
Kentzer et al. Characterization of plasmin digested peptide maps for recombinant high molecular weight urokinase and pro-Urokinase. Its use for the estimation of low molecular weight urokinase in pro-UK/HMW-UK
Shikata et al. Intelligent peptide-mapping method for recombinant human tissue-type plasminogen activator by lysyl-endopeptidase digestion
JPH0759191B2 (en) Method for producing human plasmin-α2-plasmin inhibitor complex and method for separating the same
Malhotra Purification and characterization of dicoumarol-induced prothrombins. Evidence of 5-Gla variant with two (or more) polypeptide chains
JPH04218368A (en) Production of plasminogen activator precursor

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 BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 2384756

Country of ref document: CA

Ref document number: 15816/01

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 149006

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: PV2002-1196

Country of ref document: CZ

Ref document number: 2002/02687

Country of ref document: ZA

Ref document number: 200202687

Country of ref document: ZA

Ref document number: 518187

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2000978343

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2002/004401

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1020027005727

Country of ref document: KR

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2001 535595

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 008153434

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2000978343

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: PV2002-1196

Country of ref document: CZ

WWP Wipo information: published in national office

Ref document number: 1020027005727

Country of ref document: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 518187

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 518187

Country of ref document: NZ

WWG Wipo information: grant in national office

Ref document number: 2000978343

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 15816/01

Country of ref document: AU