WO2009137254A2 - Modified factor ix polypeptides and uses thereof - Google Patents

Modified factor ix polypeptides and uses thereof Download PDF

Info

Publication number
WO2009137254A2
WO2009137254A2 PCT/US2009/040813 US2009040813W WO2009137254A2 WO 2009137254 A2 WO2009137254 A2 WO 2009137254A2 US 2009040813 W US2009040813 W US 2009040813W WO 2009137254 A2 WO2009137254 A2 WO 2009137254A2
Authority
WO
WIPO (PCT)
Prior art keywords
fix
amino acid
polypeptide
polypeptides
glycosylation sites
Prior art date
Application number
PCT/US2009/040813
Other languages
French (fr)
Other versions
WO2009137254A3 (en
Inventor
Alan Brooks
John E. Murphy
Marian Seto
Xiaoqiao Jiang
Chandra Patel
Uwe Gritzan
Kornelia Kirchner
Ulrich Haupts
Original Assignee
Bayer Healthcare Llc
Bayer Healthcare Ag
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
Application filed by Bayer Healthcare Llc, Bayer Healthcare Ag filed Critical Bayer Healthcare Llc
Priority to CA2721683A priority Critical patent/CA2721683A1/en
Priority to MX2010011345A priority patent/MX2010011345A/en
Priority to JP2011505204A priority patent/JP2011517951A/en
Priority to BRPI0910702A priority patent/BRPI0910702A2/en
Priority to CN2009801227842A priority patent/CN102083856A/en
Priority to EP09743241A priority patent/EP2288622A4/en
Priority to AU2009244633A priority patent/AU2009244633A1/en
Publication of WO2009137254A2 publication Critical patent/WO2009137254A2/en
Publication of WO2009137254A3 publication Critical patent/WO2009137254A3/en
Priority to IL208718A priority patent/IL208718A0/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/644Coagulation factor IXa (3.4.21.22)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21022Coagulation factor IXa (3.4.21.22)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates to modified Factor IX polypeptides such as Factor IX polypeptides with one or more introduced glycosylation sites.
  • the modified Factor IX polypeptides may exhibit increased in vitro or in vivo stability such as a longer plasma half-life.
  • the invention also relates to methods of making modified Factor IX polypeptides, and methods of using modified Factor IX polypeptides, for example, to treat patients afflicted with hemophilia B.
  • Hemophilia B effects one out of 34,500 males and is caused by various genetic defects in the gene encoding coagulation Factor IX (FIX) that result in either low or undetectable FIX protein in the blood (Kurachi, et al., Hematol. Oncol. Clin. North Am. 6:991-997, 1992; Lillicrap, Haemophilia 4:350-357, 1998). Insufficient levels of FIX lead to defective coagulation and symptoms that result from uncontrolled bleeding.
  • FIX coagulation Factor IX
  • Hemophilia B is treated effectively by the intravenous infusion of either plasma-derived or recombinant FIX protein either to stop bleeds that have already initiated or to prevent bleeding from occurring (prophylaxis) (Dargaud, et al., Expert Opin. Biol. Ther. 7:651-663; Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005). Effective prophylaxis requires maintaining a minimum trough level of FIX of about 1% of normal levels (Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005).
  • FIX levels drop to less than 1% of normal levels within 3 to 4 days following bolus injection which necessitates repeat injection on average every three days to achieve effective prophylaxsis (Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005).
  • Such frequent intravenous injection is problematic for patients and is a hurdle for achieving effective prophylaxsis (Petrini, Haemophilia 13 Suppl 2:16-22, 2007), especially in children.
  • a FIX protein with a longer half-life would enable less frequent administration and thus, be of significant medical benefit.
  • the application provides FIX polypeptides (also referred to as modified FIX polypeptides, FIX muteins, or FIX variants) comprising amino acid sequences that have been modified by introducing one or more glycosylation sites.
  • the one or more glycosylation sites may be N-linked glycosylation sites.
  • the polypeptides have coagulation activity.
  • the modified polypeptides may comprise at least one substitution such as, but not limited to R338A and V86A. In some embodiments, the modified polypeptides may comprise both the R338A and V86A substitutions.
  • the application also provides FIX polypeptides comprising amino acid sequences that have been modified by introducing one or more glycosylation sites.
  • the FIX polypeptides may further comprise a carbohydrate chain attached to the one or more introduced glycosylation sites.
  • the carbohydrate chain may be an N-linked carbohydrate chain.
  • the carbohydrate chain may have a mammalian carbohydrate chain structure.
  • the carbohydrate chain may have a human carbohydrate chain structure.
  • the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites may increase serum half-life of the polypeptide by, for example, at least 30% relative to the polypeptide lacking the introduced glycosylation sites. In some embodiments, the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites does not reduce the amount of secreted polypeptide by, for example, more than 50% relative to the amount of the secreted polypeptide lacking the introduced glycosylation sites.
  • the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites does not inhibit interaction of the polypeptide with at least one of Factor VIII (FVIII), Factor XI (FXI), or Factor X (FX) by, for example, more than 50% relative to interaction of the polypeptide lacking the carbohydrate chain at the introduced glycosylation sites with FVIII, FXI, or FX.
  • the modified polypeptide may have a specific activity of, for example, at least 100 units per mg of polypeptide.
  • the one or more glycosylation sites may be introduced via one or more amino acid substitutions.
  • the substitutions may be at surface exposed residues and/or be limited to substitutions that do not introduce a mutation known to be associated with hemophilia B.
  • Exemplary embodiments include FIX polypeptides comprising one or more substitutions such as, but not limited to:
  • the one or more glycosylation sites may be introduced into the catalytic domain of FIX or activation peptide.
  • FIX polypeptides comprising one or more substitutions such as, but not limited to: R37N; D85N; K122T; S138N; A146N; A161N; Q170N; T172N; D177N; F178T; K201N; K228N; E239N; E242N; I251T; A262T; E294N; E374N; and E410N.
  • FIX polypeptides comprising one or more substitutions such as: G59N and S61T; K63N and D65T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; E119N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; V156N and S158T; S158N and E160T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; D166N and I168T; I168N and Q170T; S171N and Q173T; T172N and S174T; Q173N and F175
  • the one or more glycosylation sites may be introduced by converting an O-linked glycosylation site to an N-linked glycosylation site.
  • Exemplary embodiments include substitutions such as, but not limited to T169N; T172N; T148N and F150T; and T159N and A161T.
  • the substitutions may be T172N; T148N and F150T; and T159N and A161T.
  • the modified polypeptides may further comprise at least one substitution such as, but not limited to R338A and V86A.
  • the modified polypeptides may further comprise both the R338A and V86A substitutions.
  • the one or more glycosylation sites may be introduced by inserting between 1 and 12 amino acid residues between amino acid residues 160-164.
  • the glycosylation sites may be introduced between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163-164.
  • SEQ ID NO: 2 may be introduced between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163-164.
  • glycosylation sites may be introduced by adding between 1 and 12 amino acid residues to the C-terminus of the FIX polypeptide.
  • SEQ ID NOs: 4, 5, 6, or 7 may be introduced to the to the C-terminus of the FIX polypeptide.
  • the polypeptide may further comprise amino acid substitutions D177E and F178T.
  • the polypeptide may further comprise amino acid substitutions P151N and V153T.
  • the polypeptide may further comprise amino acid substitution T172N.
  • the polypeptide may further comprise amino acid substitutions Pl 5 IN, V153T, and T172N.
  • the polypeptide may further comprise amino acid substitutions T148N and Fl 5OT.
  • the polypeptide may further comprise amino acid substitutions G226N and K228T.
  • the modified polypeptides may further comprise at least one substitution such as R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
  • modified FIX polypeptides comprising one or more substitutions such as: D85N; K122T; S138N; T172N; K201N; K228N; E239N; E242N; I251T; A262T; E294N; G59N and S61T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; E119N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; S158N and E160T; E162N and I164T; T163N and L165T; T172N and S174T; Q173N and F175T; K201N and D203T; T225N and V227T; G2
  • the application also provides FIX polypeptides comprising an R338A substitution and a V86A substitution.
  • the polypeptide may have a specific activity of at least 700 units per mg of polypeptide.
  • the application also provides pharmaceutical preparations comprising modified FIX polypeptides and a pharmaceutically acceptable carrier.
  • the application also provides methods for treating hemophilia B comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical preparations described herein.
  • the application also provides DNA sequences encoding modified polypeptides, as well as eukaryotic host cells transfected with the DNA sequences. [015] The application also provides methods for producing modified FIX polypeptides comprising (i) modifying the amino acid sequence of the polypeptide by introducing one or more glycosylation sites; (ii) expressing the polypeptide in a manner which allows glycosylation at the one or more glycosylation sites; and (iii) purifying the polypeptide.
  • Figure 1 depicts a multiple sequence alignment of FIX sequences within the activation peptide from eight species
  • Figure 2 depicts Western Blot analysis of media from HKBl 1 cells transfected with glycosylation site muteins of FIX.
  • FIX protein was detected using an anti-FIX-HRP antibody.
  • Figure 3 depicts a table of expression and activity of FIX glycosylation site muteins in HKBI l cells. Expression was determined by ELISA and activity by aPTT assay. Specific activity is calculated as international units per mg of FIX protein. Values were converted to a percentage of FIX-R338A run in the same transfection experiment, (none): no change in mobility, (+): decreased mobility with the number of + indicating degree, (-): increased mobility.
  • Figure 4 depicts expression level, coagulation activity, and specific activity of FIX glycosylation site muteins in HKBl 1 cells. Values are expressed as a percentage of the FIX- R338A mutein. Constructs are described in Figure 3.
  • Figure 5 depicts Western Blot analysis of media from HKBl 1 cells transfected with the G226N, K228T glycosylation site mutein of FIX.
  • FIX protein was detected using an anti-FIX- HRP antibody.
  • the present application provides FIX polypeptides that include one or more O- or N- linked glycosylation sites.
  • Increased glycosylation of therapeutic proteins may be used to achieve 1) reduced immunogenicity; 2) less frequent administration of the protein; 3) increased protein stability such as increased serum half-life; and 4) reduction in adverse side effects such as inflammation.
  • the application provides variants of human FIX with one or more additional glycosylation sites.
  • the modified FIX polypeptides may also have an increased plasma half-life that would provide, for example, an extended time of protection against bleeding in hemophilia B patients.
  • the modified FIX polypeptides would enable hemophilia B patients to achieve protection against bleeding with fewer injections of FIX than is possible with the currently available therapy of wild type FIX protein.
  • the application provides a number of exemplary variants of FIX in which functional glycosylation sites were created in both the catalytic domain and in the activation peptide. Moreover, the application demonstrates that these variants may be expressed in mammalian cells, have increased apparent molecular weight indicative of increased glycosylation, and maintain between 50% and 100% activity in a coagulation assay. Finally, these modification sites may be combined with alterations that enhance the specific activity of FIX, including but not limited to the R338A substitution and/or the V86A substitution (Chang, et al., J. Biol. Chem. 273:12089-12094, 1998 and Chang, et al., J. Biol. Chem.
  • wild type FIX is a single chain glycoprotein of about 55,000 Daltons. It can structurally be considered as having four domains: the GIa or gamma carboxyglutamate-rich domain; the EGF-like regions; the activation peptide; and the catalytic domain containing the active site (Thomson, Blood 67:565-572, 1986).
  • FIX is synthesized in the liver as a single chain polypeptide of 461 amino acids and undergoes extensive posttranslational modification during passage through the golgi and endoplasmic reticulum (Nemerson, et al., CRC Crit. Rev. Biochem. 9:45-48, 1980; Stenflo, et al., Annu.
  • Efficient gamma carboxylation is essential for the coagulation activity of FIX and in humans 12 GIa residues are generated within the N terminal GIa domain, although gamma carboxylation on Gla36 and Gla40 are not required for function (DiScipio, et al., Biochemistry 18:899-904, 1979; Gillis, et al., Protein Sci. 6:185-196, 1997).
  • FIX contains two N- linked glycosylation sites (N157, N167), six O-linked glycosylation sites (S53, S61, T159, T169, T172, T179), and one site each for Ser phosphorylation (S158), tyrosine sulfation (Y155) and ⁇ - hydroxylation (D64) (McMullen, et al., Biochem. Biophys. Res. Comm. 115:8-14, 1983).
  • Activated Factor VII initiates the normal hemostatic process by forming a complex with tissue factor (TF), exposed as a result of injury to the vessel wall.
  • the complex subsequently activates FIX; the active form referred to as FIXa.
  • the activation peptide of FIX is removed by proteolytic cleavage at two sites by either Factor XIa (FXIa) or the tissue factor (TF)/Factor Vila complex to generate the catalytically active molecule, Factor IXa (FIXa).
  • FIXa and Factor Villa (FVIIIa) convert FX to Factor Xa (FXa), which in turn converts prothrombin to thrombin. Thrombin then converts fibrinogen to fibrin resulting in formation of a fibrin clot.
  • FIX As wild-type FIX has numerous post-translational modifications some of which have been suggested to play a role in the in vivo pharmacokinetic profile, an ectopic glycosylation site may be introduced at a position that does not affect these other modifications. Once produced, FIX should retain enzymatic activity and interact with FVIII, FXI, and FX in order to be an effective treatment for hemophilia B. The introduced glycosylation site should not perturb these interactions and function.
  • the application provides, in part, modifications to FIX which are likely to result in an increased number of glycosylation sites with minimal perturbation of function and thus have utility for increasing the bioavailability of FIX.
  • modification sites may be combined with alterations that enhance the specific activity of FIX, including but not limited to the R338A substitution and/or the V86A substitution. Alterations that enhance the specific activity of FIX may compensate for potential loss of coagulation activity and also potentially prolong the efficacy of modified molecules by conferring efficacy at lower levels of protein.
  • FIX polypeptides comprising one or more introduced glycosylation sites, that is, modified FIX polypeptides.
  • "Factor IX” as used herein refers to a human plasma FIX glycoprotein that is a member of the intrinsic coagulation pathway and is essential to blood coagulation. It is to be understood that this definition includes native as well as recombinant forms of the human plasma FIX glycoprotein. Unless otherwise specified or indicated, as used herein FIX means any functional human FIX protein molecule in its normal role in coagulation, including any fragment, analogue, variant, and derivative thereof.
  • fragment when referring to the polypeptides of the application, means fragments, derivatives, analogues, muteins, and variants of the polypeptides which retain substantially the same biological function or activity.
  • Non-limiting examples of FIX polypeptides include FIX, FIXa, and truncated versions of FIX having FIX activity. Biologically active fragments, deletion variants, substitution variants, or addition variants of any of the foregoing that maintain at least some degree of FIX activity can also serve as a FIX polypeptide.
  • the FIX polypeptides may comprise an amino acid sequence at least about 70, 80, 90, or 95% identical to SEQ ID NO: 1.
  • the modified FIX polypeptides are biologically active. Biological activity can be determined, for example, by coagulation assays described herein.
  • Modified FIX polypeptides may also contain conservative substitutions of amino acids.
  • a conservative substitution is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties and include, for example, the changes of alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalan
  • N-linked refers to the attachment of a carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences Asn-X-Ser and Asn-X-Thr, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the Asn side chain.
  • N-linked glycosylation site useful for the invention may also be represented as follows Xl-Asn-X2-X3-X4; where Xl is optionally Asp, VaI, GIu, GIy, or He; X2 is any amino acid except Pro; X3 is Ser or Thr; and X4 is optionally VaI, GIu, GIy, GIn, or He.
  • Xl is optionally Asp; X2 is Ser; X3 is Thr; and X4 is GIn.
  • Xl is Asp; X2 is He; X3 is Thr; and X4 is GIn.
  • Addition of N-linked glycosylation sites to a FIX polypeptide is accomplished by altering the amino acid sequence such that one or more of the above-described tripeptide sequences is introduced.
  • O-linked glycosylation refers to the attachment of one of the sugars N- aceytlgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly to serine or threonine, although attachment to 5-hydroxyproline or 5-hydroxylysine is also possible.
  • Addition of O-linked glycosylation sites to a FIX polypeptide may be accomplished by altering the amino acid sequence such that one or more Ser or Thr residues are introduced.
  • Glycosylation sites may be introduced, for example, by deleting one or more amino acid residues, substituting one or more endogenous FIX amino acid residues with another amino acid(s), or adding one or more amino acid residues.
  • the addition of an amino acid residue may be either between two existing amino acid residues or at the N- or C-terminal end of the native FIX molecule.
  • the terminology for amino acid substitutions used is as follows.
  • the first letter represents the amino acid residue naturally present at a position of human FIX.
  • the following number represents the position in the mature human FIX amino acid sequence (SEQ ID NO:1).
  • the second letter represent the different amino acid substituting for (r ⁇ lacing/substituting) the natural amino acid.
  • R338A denotes that the Arg residue at position 338 of SEQ ID NO: 1 has been replaced with an Ala residue.
  • SEQ ID NO: 26 which includes an additional 5' amino acid sequence (46 amino acids) of human FIX polypeptide
  • position 338 of SEQ ID NO: 1 corresponds to position 384 of SEQ ID NO: 26.
  • FIX residue number system used herein refers to that of the mature human FIX protein in which residue 1 represents the first amino acid of the mature FIX polypeptide following removal of both the signal sequence and the propeptide.
  • Native or wild type FIX is the full length mature human FIX molecule as shown in SEQ ID NO: 1.
  • the glycosylation sites are engineered in FIX at locations that will not abolish the function of the protein or its expression in cells.
  • the introduced glycosylation site is surface exposed. Surface exposure can be determined based on the solvent accessible surface area as determined in Autin, et al., (J. Thromb. Haemost. 3:2044-56, 2005).
  • the introduced glycosylation site does not introduce a mutation known to be associated with hemophilia B. Known mutations can be found on the world wide web at kcl.ac.uk/ip/petergreen/haemBdatabase.html and in Table 1.
  • the modified FIX polypeptides having one or more introduced glycosylation sites may be compared to a control polypeptide.
  • Properties for comparison include, for example, solubility, activity, plasma half-life, glycosylation state, and binding properties.
  • the modified FIX polypeptides may be glycosylated. It is within the purview of one skilled in the art to select the most appropriate control polypeptide for comparison.
  • the control polypeptide may be identical to the modified polypeptide except for the one or more introduced glycosylation sites.
  • Exemplary polypeptides include wild-type FIX polypeptide and FIX polypeptides comprising one or more activating substitutions, such as R338A and/or V86A.
  • modified FIX polypeptides having increased in vitro or in vivo stability over a control polypeptide.
  • Enhanced serum half-life and in vivo stability may be desirable to reduce the frequency of dosing that is required to achieve therapeutic effectiveness.
  • the glycosylated FIX polypeptides have a serum half-life increased by about 20, 30, 40, 60, 80, 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, or 1000% relative to a control protein.
  • the modified FIX polypeptides may have a serum half-life of at least one, at least two, at least three, at least four, at least five, at least ten, or at least twenty days or more.
  • half-life as used herein in the context of administering a polypeptide drug to a patient, is defined as the time required for plasma concentration of a drug in a patient to be reduced by one half.
  • Methods for pharmacokinetic analysis and determination of half-life and in vivo stability will be familiar to those skilled in the art. Details may be found in Kenneth, et al., Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters, et al., Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics,” M Gibaldi & D Perron, published by Marcel Dekker, 2nd Rev.
  • modified FIX polypeptides may be described either as an absolute value, such as in units, or as a percentage of the activity of a control polypeptide. In some embodiments, the modified FIX polypeptides may have a specific activity that is not reduced more than about 10, 20, 30, 40, 50, 60, 70, or 80% relative to a control protein.
  • a modified FIX polypeptide may have a specific activity that is not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide maintains at least about 20% of the specific activity as compared to the specific activity of the control.
  • FIX specific activity may be defined as the ability to function in the coagulation cascade, induce the formation of FXa via interaction with FVIIIa on an activated platelet, or support the formation of a blood clot. The activity may be assessed in vitro by techniques such as clot analysis, as described in, for example, McCarthy, et al., (Thromb. Haemost. 87:824-830,2002), and other techniques known to those skilled in the art.
  • the activity may also be assessed in vivo using one of the several animal lines that have been intentionally bred with a genetic mutation for hemophilia B such that an animal produced from such a line is deficient for FIX.
  • Such lines are available from a variety of sources such as, without limitation, the Division of Laboratories and Research, New York Department of Public Health, Albany, N. Y. and the Department of Pathology, University of North Carolina, Chapel Hill, N.C. Both of these sources, for example, provide canines suffering from canine hemophilia B.
  • mice deficient in FIX are also available (Sabatino, et al., Blood 104:2767-2774, 2005).
  • a test polypeptide is injected into the diseased animal, a small cut made and bleeding time compared to a healthy control.
  • Human wild-type FIX has a specific activity of around 200 units per mg.
  • One unit of FIX has been defined as the amount of FIX present in one millilitre of normal (pooled) human plasma (corresponding to a FIX level of 100%).
  • the modified FIX polypeptides may have a specific activity of at least about 100 units per mg of FIX polypeptide.
  • the modified FIX polypeptides may have a specific activity of at least about 120, 140, 160, 180, 200, 220, 240, 260 units, or more per mg of FIX polypeptide.
  • the specific activity of FIX may be measured using the APTT or activated partial thromboplastin time assay (described by, e.g., Proctor, et al., Am. J. Clin. Pathol. 36:212, 1961 and see Examples).
  • FIX polypeptide When expressed in cells, such as liver or kidney cells, FIX polypeptide may be synthesized by the cellular machinery, undergoes posttranslational modification, and is then secreted by the cells into the extracellular milieu. The amount of FIX polypeptide secreted from cells is therefore dependent on both processes of protein translation and extracellular secretion. In some embodiments, the modified FIX polypeptides may be secreted in an amount that is not reduced more than about 10, 20, 30, 40, 50, 60, 70, or 80% relative to the amount secreted of a control protein.
  • a modified FIX polypeptide may be secreted in an amount that is not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide is secreted in an amount of at least about 20% as compared to the control.
  • the amount of FIX polypeptide secreted may be measured, for example, by determining the protein levels in the extracellular medium using any art-known method.
  • Traditional methodologies for protein quantification include 2-D gel electrophoresis, mass spectrometry, and antibody binding.
  • Exemplary methods for assaying protein levels in a biological sample include antibody-based techniques, such as immunoblotting (western blotting), immunohistological assay, enzyme linked immunosorbent assay (ELISA), or radioimmunoassay (RIA).
  • the modified FIX polypeptides interact with at least one of FVIII, FXI, or FX at a level not reduced more than about 40, 50, 60, 70, or 80% relative to the interaction of a control protein with at least one of FVIII, FXI, or FX.
  • a modified FIX polypeptide may interact with at least one of FVIII, FXI, or FX at a level not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide interacts with at least one of FVIII, FXI, or FX at a level of at least about 20% as compared to the control.
  • FIX binding of FIX to other members of the coagulation cascade
  • binding of FIX to other members of the coagulation cascade can be determined by any method known to one skilled in the art, including for example, the methods described in Chang, et al., (J. Biol. Chem. 273:12089-12094, 1998).
  • FIX is composed of four structural domains with different biological functions.
  • One aspect of the invention is to provide FIX polypeptides that are modified in particular domains, such as in the activation peptide and in the catalytic domain.
  • the application provides, in part, FIX polypeptides comprising one more glycosylation sites introduced into the activation peptide of FIX.
  • the natural glycosylation sites in FIX are located in the EGFl domain (two O-linked sites on Ser53 and Ser ⁇ l) and in the activation peptide (four O-linked sites at Thrl59, Thrl69, Thrl72, and Thrl79 and 2 N-linked sites at Asnl57 and Asn 167).
  • the activation peptide may have less of a negative impact on activity or expression when introduced into regions of FIX that have natural glycosylation sites, such as the activation peptide.
  • the activation peptide is cleaved when FIX is activated and so is not required for the catalytic activity of FIXa.
  • the activation peptide is present in the zymogen, making the activation peptide an attractive region to introduce glycosylation sites to improve the circulating half-life of the zymogen.
  • FIX polypeptides comprising one or more glycosylation sites.
  • FIX polypeptides are provided comprising one or more substitutions selected from G4T; E33N; E36T; E36N; R37N; F75N; F77T; E83T; D85N; V86A; K91T; A103T; V107T; K122N; K122T; S138N; A146N; T148N; F150T; P151N; T159N; A161T; A161N; T169N; Q170N; T172N; D177N; D177E; F178T; K201N; K201T; K214T; V223N; G226N; Y226T; K228N; K228T; E239N; E242N; I251T; A262T; E294N; R338A; R338N; K341N; F353N;
  • FIX polypeptides comprising one or more substitutions selected from YlN and S3T; S3N and K5T; G4N and L6T; K5N and E7T; L6N and E8T; E7N and F9T; F9N and Ql IT; VlON and G12T; QI lN and N13T; G12N and L14T; N13 and E15T; L14N and R16T; E15N and E17T; M19N and E21T; E20N and K22T; S24N and E26T; F25N and E27T; E26N and A28T; E27N and R29T; A28N and E30T; R29N and V3 IT; E30N and F32T; V3 IN and E33T; F32N and N34T; T35N and R37T; T38N and E40T; T39N and F41T; E40N and W42T; F41
  • FIX polypeptides comprising one or more substitutions selected from D85N, K122T, and I251T; D85N, K122T, and E242N; E125N, P126A, and A127T; P126N, V128T, and P129A; T148N, F150T, and P151A; F150N, P151A, and D152T; P151N, V153T, and A161N; P151N, V153T, and T172N; V153N, Y155T, and E294N; T172N, G226N, and K228T; F353N, H354V, and E355T; F353N, H354I, and E355T; V370N, T371 V, and E372T; V370N, T371I, and E372T; M391N, K392V, and G393T; D85N, P151N, V153T,
  • FIX polypeptides comprising one or more substitutions selected from
  • FIX polypeptides comprising one more glycosylation sites introduced by converting an endogenous O-linked glycosylation site to an N-linked glycosylation site. It has been reported that N-linked glycosylation sites are more likely to be sialylated than O-linked glycosylation sites and there is evidence that higher sialic acid content confers increased protein half-life. It is generally believed that the increased sialic acid content provided by additional N-linked glycosylation may be responsible for the increased half-life in blood (White, et al., Thromb. Haemost. 78:261-265, 1997). Exemplary embodiments of such modified FIX polypeptides are as follows.
  • FIX polypeptides are provided comprising a T169N substitution. In some embodiments, FIX polypeptides are provided comprising a T172N substitution. In some embodiments, FIX polypeptides are provided comprising a T148N substitution and an F 150T substitution. In some embodiments, FIX polypeptides are provided comprising a T159N substitution and an A161T substitution.
  • Another aspect of the invention provides for the insertion of amino acids into the activation peptide in order to generate one or more glycosylation sites.
  • the application provides, in part, FIX polypeptides comprising one more glycosylation sites introduced between amino acid residues 160 to 164 of human FIX.
  • the amino acid residues introduced include a glycosylation site.
  • the amino acid residues introduced form a glycosylation site in combination with wild-type FIX amino acid residues.
  • the apparent site at which the additional amino acids in rat, mouse, and guinea pig are found can vary such that the site can be either between E160 and A161, between A161 and E162, between E162 and T163, or between T163 and 1164 of human FIX.
  • the amino acid sequence inserted between El 60 and Al 61 may be SEQ ID NO: 2.
  • the amino acid sequence inserted between Al 61 and E 162 may be SEQ ID NO: 2.
  • the amino acid sequence inserted between E162 and T163 may be SEQ ID NO: 2.
  • the amino acid sequence inserted between T163 and 1164 may be SEQ ID NO: 2.
  • one or more amino acids may be inserted between E160 and A161 and one or more amino acids may be inserted between A161 and E162.
  • one or more amino acids may be inserted between E160 and A161 and one or more amino acids may be inserted between E 162 and Tl 63.
  • one or more amino acids may be inserted between A161 and E162 and one or more amino acids may be inserted between E162 and T163.
  • one or more amino acids may be inserted between T163 and 1164.
  • up to about 30, 25, 20, 18, 16, 14, or 12 total amino acid residues may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, up to about 10 total amino acids may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, up to about 9 total amino acids may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, one, two, three, or four glycosylation sites may be introduced.
  • the application further provides modified FIX polypeptides comprising more than one of the introduced glycosylation sites disclosed herein.
  • FIX polypeptides are provided that comprise at least one introduced glycosylation site in the catalytic domain and at least one introduced glycosylation site in the activation peptide.
  • FIX polypeptides are provided that comprise at least two introduced glycosylation sites in the catalytic domain.
  • FIX polypeptides are provided that comprise at least two introduced glycosylation sites in the activation peptide.
  • the FIX polypeptides may comprise one or more of the following substitutions: R37N; D85N; K122T; S138N; A146N; A161N; Q170N; T172N; D177N; F178T; K201N; K228N; E239N; E242N; 125 IT; A262T; E294N; E374N; and E410N.
  • inventions may comprise one or more of the following substitutions: G59N and S61T; K63N and D65T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; El 19N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; V156N and S158T; S158N and E160T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; D166N and I168T; I168N and Q170T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T;
  • the application further provides modified FIX polypeptides that comprise at least one introduced glycosylation site at the C-terminus of the FIX polypeptide (i.e., following amino acid residue 415 of the FIX polypeptide).
  • FIX polypeptides are provided that comprise at least two, at least three, at least four, or more glycosylation sites at the C-terminus of the FIX polypeptide.
  • the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 4 at the C-terminus of the FIX polypeptide.
  • the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 5 at the C- terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 6 at the C-terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 7 at the C-terminus of the FIX polypeptide. [054]
  • modified FIX polypeptides comprising at least one or more glycosylation sites and one or more substitutions that increase the activity of FIX.
  • modified FIX polypeptides may comprise the R338A substitution.
  • modified FIX polypeptides may comprise the V86A substitution.
  • modified FIX polypeptides may comprise both the R338A and the V86A substitution.
  • FIX polypeptides with increased specific activity.
  • FIX polypeptides may comprise an R338A substitution and a V86A substitution.
  • the polypeptides may have a specific activity of at least about 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1400, 1600, 1800, or 2000 units per mg of polypeptide.
  • the specific activity can be determined as previously described, such as, for example, using the APTT assay.
  • These polypeptides are useful as therapeutic agents, particularly in patients afflicted with hemophilia B.
  • These polypeptides may comprise further substitutions or modifications, such as the glycosylation sites described herein.
  • modified Factor IX polypeptides comprising the following amino acid sequence:
  • X 85 is selected from D and E; wherein X 86 is selected from A, E, P, S, and V; wherein X 104 is selected from D, N, and T; wherein X 121 is selected from N, Q, and T; wherein X 138 is selected from N, S, and T; wherein X 148 and X 150 are selected from: (i) X 148 is T and X 150 is F; (ii) Xi 48 is N and Xi 50 is T; and (iii) Xi 48 is N and Xi 50 is S; wherein X 151 is selected from A, P, and T; wherein X151 and Xi 53 are selected from: (i) Xi 5I is P and Xi 53 is V; (ii) Xi 5 i is N and X i53 is T; and (iii) Xi 5 i is N and X i53 is S; wherein Zi, Z 2 , Z 3 , and Z 4
  • the introduction of at least one glycosylation site is the result of a substitution at least one of the X positions or an insertion in at least one of the Z positions.
  • the modified polypeptide additionally comprises between about 1-30, 1-20, or 1-10 conservative amino acid changes and maintains FIX activity.
  • the modified polypeptide is at least about 80, 85, 90, 95, or 99% identical to SEQ ID NO: 1 and maintains FIX activity.
  • Amino acid residues may be inserted, deleted, or substituted in order to introduce a non- native glycosylation site.
  • glycosylation sites may be introduced by altering the amino acid sequence of FIX.
  • Amino acid sequence alteration may be accomplished by a variety of techniques, such as, for example, by modifying the corresponding nucleic acid sequence by site- specific mutagenesis. Techniques for site-specific mutagenesis are well known in the art and are described in, for example, Zoller et al., (DNA 3:479-488, 1984) or Horton, et al., (Gene 77:61-68, 1989, pp. 61-68).
  • the nucleic acid construct encoding the FIX polypeptide may also be prepared synthetically by established standard methods, for example, the phosphoramidite method described by Beaucage, et al., (Gene Amplif. Anal. 3:1-26, 1983). According to the phosphoamidite method, oligonucleotides are synthesized, for example, in an automatic DNA synthesizer, purified, annealed, ligated, and cloned in suitable vectors. The DNA sequences encoding the human FIX polypeptides may also be prepared by polymerase chain reaction using specific primers, for example, as described in US Patent No.
  • nucleic acid construct may be of mixed synthetic and genomic, mixed synthetic and cDNA, or mixed genomic and cDNA origin prepared by ligating fragments of synthetic, genomic, or cDNA origin (as appropriate), corresponding to various parts of the entire nucleic acid construct, in accordance with standard techniques.
  • the DNA sequences encoding the FIX polypeptides may be inserted into a recombinant vector using recombinant DNA procedures.
  • the choice of vector will often depend on the host cell into which the vector is to be introduced.
  • the vector may be an autonomously replicating vector or an integrating vector.
  • An autonomously replicating vector exists as an extrachromosomal entity and its replication is independent of chromosomal replication, for example, a plasmid.
  • An integrating vector is a vector that integrates into the host cell genome and replicates together with the chromosome(s) into which it has been integrated.
  • the vector may be an expression vector in which the DNA sequence encoding the modified FIX is operably linked to additional segments required for transcription, translation, or processing of the DNA, such as promoters, terminators, and polyadenylation sites.
  • the expression vector may be derived from plasmid or viral DNA, or may contain elements of both.
  • operably linked indicates that the segments are arranged so that they function in concert for their intended purposes, for example, transcription initiates in a promoter and proceeds through the DNA sequence coding for the polypeptide.
  • Expression vectors for use in expressing FIX polypeptides may comprise a promoter capable of directing the transcription of a cloned gene or cDNA.
  • the promoter may be any DNA sequence that shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
  • Suitable promoters for directing the transcription of the DNA encoding the FIX polypeptides in mammalian cells are, for example, the SV40 promoter (Subramani, et al., MoI. Cell Biol. 1 :854-864, 1981), the MT-I (metallothionein gene) promoter (Palmiter, et al., Science 222:809-814, 1983), the CMV promoter (Boshart, et al., Cell 41 :521-530, 1985), or the adenovirus 2 major late promoter (Kaufman et al.,, MoI. Cell Biol, 2:1304-1319, 1982).
  • the DNA sequences encoding the FIX polypeptide may also, if necessary, be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter, et al.,
  • the expression vectors may also contain a polyadenylation signal located downstream of the insertion site.
  • Polyadenylation signals include the early or late polyadenylation signal from SV40, the polyadenylation signal from the adenovirus 5 EIb region, the human growth hormone gene terminator (DeNoto, et al., Nucl. Acids Res. 9:3719-3730, 1981), or the polyadenylation signal from the human FIX gene.
  • the expression vectors may also include enhancer sequences, such as the SV40 enhancer.
  • the native FIX secretory signal sequence may be used.
  • a secretory signal sequence also known as a leader sequence, prepro sequence, or pre sequence
  • the secretory signal sequence may be joined to the DNA sequences encoding the FIX analogues in the correct reading frame.
  • Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide.
  • Exemplary signal sequences include, for example, the MPIF-I signal sequence and the stanniocalcin signal sequence.
  • Cloned DNA sequences may be introduced into cultured mammalian cells by, for example, lipofection, DEAE-dextran-mediated transfection, microinj ection, protoplast fusion, calcium phosphate precipitation, retroviral delivery, electroporation, sonoporation, laser irradiation, magnetofection, natural transformation, and biolistic transformation (see, e.g., Mehier- Humbert, et al., Adv. Drug Deliv. Rev. 57:733-753, 2005).
  • a gene that confers a selectable phenotype is generally introduced into cells along with the gene or cDNA of interest.
  • Selectable markers include, for example, genes that confer resistance to drugs such as neomycin, puromycin, hygromycin, and methotrexate.
  • the selectable marker may be an amplifiable selectable marker, which permits the amplification of the marker and the exogenous DNA when the sequences are linked.
  • Exemplary amplifiable selectable markers include dihydrofolate reductase (DHFR) and adenosine deaminase. It is within the purview of one skilled in the art to choose suitable selectable markers (see, e.g., US Patent No. 5,238,820).
  • appropriate growth medium means a medium containing nutrients and other components required for the growth of cells and the expression of the active FIX polypeptides.
  • Media generally include, for example, a carbon source, a nitrogen source, essential amino acids, essential sugars, vitamins, salts, phospholipids, protein, and growth factors, and in the case of vitamin K dependent proteins such as FIX, vitamin K may also be provided.
  • Drug selection is then applied to select for the growth of cells that are expressing the selectable marker in a stable fashion. For cells that have been transfected with an amplifiable selectable marker the drug concentration may be increased to select for an increased copy number of the cloned sequences, thereby increasing expression levels. Clones of stably transfected cells are then screened for expression of the FIX polypeptide.
  • Examples of mammalian cell lines for use in the present invention are the COS-I (ATCC CRL 1650), baby hamster kidney (BHK), HKBl 1 cells (Cho, et al., J. Biomed. Sci, 9:631-638, 2002), and HEK-293 (ATCC CRL 1573; Graham, et al., J. Gen. Virol. 36:59-72, 1977) cell lines.
  • COS-I ATCC CRL 1650
  • BHK baby hamster kidney
  • HKBl 1 cells Cho, et al., J. Biomed. Sci, 9:631-638, 2002
  • HEK-293 ATCC CRL 1573; Graham, et al., J. Gen. Virol. 36:59-72, 1977
  • rat Hep I rat hepatoma; ATCC CRL 1600
  • rat Hep II rat hepatoma; ATCC CRL 1548
  • TCMK-I ATCC CCL 139
  • Hep-G2 ATCC HB 8065
  • NCTC 1469 ATCC CCL 9.1
  • CHO-Kl ATCC CCL 61
  • CHO-DUKX cells Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77:4216-4220, 1980).
  • FIX polypeptides may be recovered from cell culture medium and may then be purified by a variety of procedures known in the art including, but not limited to, chromatography (e.g., ion exchange, affinity, hydrophobic, chromatofocusing, and size exclusion), electrophoretic procedures (e.g., preparative isoelectric focusing (IEF), differential solubility (e.g., ammonium sulfate precipitation)), extraction (see, e.g., Protein Purification, Janson and Lars Ryden, editors, VCH Publishers, New York, 1989), or various combinations thereof.
  • the polypeptides may be purified by affinity chromatography on an anti-FIX antibody column.
  • Additional purification may be achieved by conventional chemical purification means, such as high performance liquid chromatography.
  • Other methods of purification are known in the art, and may be applied to the purification of the modified FIX polypeptides (see, e.g., Scopes, R., Protein Purification, Springer- Verlag, N. Y., 1982).
  • purified shall refer to a protein or peptide composition that has been subjected to fractionation to remove various other components, and which substantially retains its expressed biological activity. Where the term “substantially purified” is used, this designation shall refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or more of the proteins in the composition.
  • Various methods for quantifying the degree of purification of the polypeptide are known to those of skill in the art. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis.
  • An exemplary method for assessing the purity of a fraction is to calculate the specific activity of the fraction, compare the activity to the specific activity of the initial extract, and to thus calculate the degree of purity, herein assessed by a "-fold purification number.”
  • the actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique.
  • FIX polypeptides are recombinantly expressed in tissue culture cells and glycosylation is the result of the normal post-translational cell functioning of the host cell, such as a mammalian cell.
  • glycosylation may be achieved through chemical or enzymatic modification.
  • FIX polypeptides may be glycosylated by using, for example, an enzyme that adds alpha-(2,6)- linked sialic acid to protein.
  • an enzyme that adds alpha-(2,6)- linked sialic acid to protein For example, dihydrofolate reductase (DHFR) deficient CHO cells are commonly used host cells for recombinant glycoprotein production. CHO cells do not endogenously express the enzyme beta-galactoside alpha-2,6 sialyltransferase, which is used to add sialic acid in the 2,6 linkage to galactose on the mannose alpha- 1,3 branch.
  • DHFR dihydrofolate reductase
  • the CHO cells may be transfected with a functional beta-galactosidase alpha-2,6 sialyltransferase gene to allow for incorporation of sialic acid in the 2,6 linkage to galactose as desired (see, e.g., Lee, et al., J. Biol. Chem. 264:13848- 13855, 1989).
  • GIcNAc bisecting N-acetylglucosamine
  • FIX a bisecting N-acetylglucosamine
  • GIcNAc bisecting N-acetylglucosamine
  • Systems have also been established to express proteins in plant cells that produce proteins with mammalian glycosylation patterns (see, e.g., bryophyte cells, WO 2004/057002).
  • N-linked glycosylation the final structures of the N-glycans are typically dependent upon the organism in which the polyp eptide is produced.
  • polypeptides produced in bacteria are completely unglycosylated.
  • Polypeptides expressed in insect cells contain high mannose and pauci-mannose N-linked oligosaccharide chains, among others.
  • Polypeptides produced in mammalian cell culture are usually glycosylated differently depending upon, for example, the species and cell culture conditions.
  • polypeptides produced in plant cells comprise glycan structures that differ significantly from those produced in animal cells.
  • the goal in the art of the production of recombinant polypeptides, particularly when the polypeptides are to be used as therapeutic agents, is to be able to generate polypeptides that are correctly glycosylated, that is, to be able to generate a polypeptide having a glycan structure that resembles, or is identical to that present on the naturally occurring form of the polypeptide.
  • O-glycans are linked primarily to serine and threonine residues and are formed by the stepwise addition of sugars from nucleotide sugars (Tanner, et al., Biochim. Biophys. Acta. 906:81-99, 1987); Hounsell, et al., Glycoconj. J. 13:19-26, 1996).
  • Polypeptide function can be affected by the structure of the O-linked glycans present.
  • Schachter and Brockhausen The Biosynthesis of Branched O-Linked Glycans, 1989, Society for Experimental Biology, pp. 1-26 (Great Britain).
  • Whether a FIX polypeptide has N-linked and/or O-linked glycosylation may be determined using standard techniques (see, e.g., Techniques in Glycobiology, R. Townsend and A. Hotchkiss, eds. (1997) Marcel Dekker; and Glycoanalysis Protocols (Methods in Molecular Biology, Vol. 76), E. Hounsell, ed. (1998) Humana Press).
  • the change in electrophoretic mobility of a protein before and after treatment with chemical or enzymatic deglycosylation e.g., using endoglycosidases and/or exoglycosidases
  • chemical or enzymatic deglycosylation e.g., using endoglycosidases and/or exoglycosidases
  • Enzymatic deglycosylation may be carried out using any of a variety of enzymes, including, but not limited to, peptide-N4-(N-acetyl-beta-D-glycosaminyl) asparagine amidase (PNGase F), endoglycosidase Fl, endoglycosidase F2, endoglycosidase F3, and the like.
  • PNGase F peptide-N4-(N-acetyl-beta-D-glycosaminyl) asparagine amidase
  • F endoglycosidase Fl
  • endoglycosidase F2 endoglycosidase F3
  • SDS-PAGE sodium docecyl sulfate-polyacrylamide gel electrophoresis
  • a marked decrease in band width and change in migration position after treatment with PNGaseF may be considered diagnostic of N-linked glycosylation.
  • the carbohydrate content of a glycosylated protein also can be detected using lectin analysis of protein blots (e.g., proteins separated by SDS-PAGE and transferred to a support, such as a nylon membrane).
  • Lectins carbohydrate binding proteins from various plant tissues, have both high affinity and narrow specificity for a wide range of defined sugar epitopes found on glycoprotein glycans (Cummings, Methods Enzymol. 230:66-86, 1994).
  • Lectins may be labeled (either directly or indirectly) allowing detection of binding of lectins to carbohydrates on glycosylated proteins.
  • a lectin bound to a glycosylated protein when conjugated with biotin or digoxigenin, a lectin bound to a glycosylated protein can be easily identified on membrane blots through a reaction utilizing avidin or anti-digoxigenin antibodies conjugated with an enzyme such as alkaline phosphatase, beta-galactosidase, luciferase, or horse radish peroxidase, to yield a detectable product. Screening with a panel of lectins with well- defined specificity provides considerable information about a glycoprotein's carbohydrate complement. The electrophoretic mobility of the modified FIX polypeptide may also be compared to the mobility of a reference protein.
  • the application provides, in part, FIX polypeptides with introduced glycosylation sites, wherein the carbohydrate chain attached to the glycosylation site may have a mammalian carbohydrate chain structure, that is, a mammalian glycosylation pattern.
  • the carbohydrate chain has a human glycosylation pattern.
  • a pattern of glycosylation refers to the representation of particular oligosaccharide structures within a given population of FIX polypeptides.
  • Non-limiting examples of such patterns include the relative proportion of oligosaccharide chains that (i) have at least one sialic acid residue; (ii) lack any sialic acid residues (i.e., are neutral in charge); (iii) have at least one terminal galactose residue; (iv) have at least one terminal N-acetylgalactosamine residue; (v) have at least one "uncapped” antenna, that is, have at least one terminal galactose or N-acetylgalactosamine residue; or (vi) have at least one fucose linked alphal->3 to an antennary N-acetylglucosamine residue.
  • the pattern of glycosylation may be determined using any method known in the art, including, without limitation: high-performance liquid chromatography (HPLC); capillary electrophoresis (CE); nuclear magnetic resonance (NMR); mass spectrometry (MS) using ionization techniques such as fast-atom bombardment, electrospray, or matrix-assisted laser desorption (MALDI); gas chromatography (GC); and treatment with exoglycosidases in conjunction with anion- exchange (AIE)-HPLC, size-exclusion chromatography (SEC), or MS (see, e.g., Weber, et al., Anal. Biochem. 225:135-142, 1995; Klausen, et al., J.
  • HPLC high-performance liquid chromatography
  • CE capillary electrophoresis
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • MALDI matrix-assisted laser desorption
  • GC gas chromatography
  • AIE anion- exchange
  • the effective dosage of the polypeptides of this invention may readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular polypeptide and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • compositions comprising FIX polypeptides with one or more introduced glycosylation sites as described herein.
  • the compositions may be suitable for in vivo administration and are pyrogen free.
  • the compositions may also comprise a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Supplementary active ingredients also may be incorporated into the compositions.
  • compositions of the present invention include classic pharmaceutical preparations. Administration of these compositions according to the present invention may be via any common route.
  • the pharmaceutical compositions may be introduced into the subject by any conventional method, for example, by intravenous, intradermal, intramuscular, subcutaneous, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary, oral, sublingual, nasal, anal, vaginal, or transdermal delivery, or by surgical implantation at a particular site.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • the active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water, suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions also may be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injectable use, include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like) sucrose, L-histidine, polysorbate 80, or suitable mixtures thereof, and vegetable oils.
  • the proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants.
  • the prevention of the action of microorganisms may be brought about by various antibacterial an antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • the injectable compositions may include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions may be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions may be prepared by incorporating the active compounds (e.g., FIX polypeptides) in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • active compounds e.g., FIX polypeptides
  • dispersions may be prepared by incorporating the various sterilized active ingredients into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • sterile powders for the preparation of sterile injectable solutions methods of preparation include, for example, vacuum-drying and freeze-drying techniques that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • solutions may be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • “Therapeutically effective amount” is used herein to refer to the amount of a polypeptide that is needed to provide a desired level of the polypeptide in the bloodstream or in the target tissue. The precise amount will depend upon numerous factors, for example, the particular FIX polypeptide, the components and physical characteristics of the therapeutic composition, intended patient population, mode of delivery, individual patient considerations, and the like, and can readily be determined by one skilled in the art, based upon the information provided herein.
  • the formulations may be easily administered in a variety of dosage forms, such as injectable solutions, and the like.
  • parenteral administration in an aqueous solution for example, the solution should be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • FIX Dosages of FIX are normally expressed in units. One unit of FIX per kg of body weight may raise plasma levels by 0.01 U/ml, that is, 1%. Otherwise healthy patients have one unit of FIX per ml of plasma, that is, 100%. Mild cases of hemophilia B are defined by FIX plasma concentrations between 6-60%, moderate cases between 1-5%, and severe cases, which account for about half of the hemophilia B cases, have less than 1% FIX. Prophylactic treatment or treatment of minor hemorrhaging usually requires raising FIX levels to between 15-30%. Treatment of moderate hemorrhaging usually requires raising levels to between 30-50%, while treatment of major trauma may require raising levels from 50 to 100%.
  • the total number of units needed to raise a patient's blood level can be determined as follows: 1.0 unit/kg x body weight (kg) x desired percentage increase (% of normal). Parenteral administration may be carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product. In some embodiments, between 15 to 150 units/kg of FIX polypeptide may be administered. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient.
  • the frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration.
  • the optimal pharmaceutical formulation may be determined by one of skill in the art depending on the route of administration and the desired dosage (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20 th edition, 2000, incorporated herein by reference). Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents.
  • a suitable dose may be calculated according to body weight, body surface area, or organ size.
  • exemplary dosing schedules include, without limitation, administration five times a day, four times a day, three times a day, twice daily, once daily, three times weekly, twice weekly, once weekly, twice monthly, once monthly, and any combination thereof.
  • Appropriate dosages may be ascertained through the use of established assays for determining blood clotting levels in conjunction with relevant dose response data.
  • the final dosage regimen may be determined by the attending physician, considering factors that modify the action of drugs, for example, the drug's specific activity, severity of the damage, and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration, and other clinical factors.
  • the composition may also include an antimicrobial agent for preventing or deterring microbial growth.
  • antimicrobial agents suitable for the present invention include benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate, thimersol, and combinations thereof.
  • An antioxidant may be present in the composition as well. Antioxidants may be used to prevent oxidation, thereby preventing the deterioration of the preparation. Suitable antioxidants for use in the present invention include, for example, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothioglycerol, propyl gallate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, and combinations thereof.
  • a surfactant may be present as an excipient.
  • exemplary surfactants include: polysorbates such as Tween®-20 (polyoxyethylenesorbitan monolaurate) and Tween®-80 (polyoxyethylenesorbitan monooleate) and pluronics such as F68 and F88 (both of which are available from BASF, Mount Olive, NJ.); sorbitan esters; lipids such as phospholipids such as lecithin and other phosphatidylcholines, phosphatidyl ethanolamines, fatty acids and fatty esters; steroids such as cholesterol; and chelating agents such as EDTA, zinc and other such suitable cations.
  • Acids or bases may be present as an excipient in the composition.
  • acids that may be used include hydrochloric acid, acetic acid, phosphoric acid, citric acid, malic acid, lactic acid, formic acid, trichloroacetic acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, fumaric acid, and combinations thereof.
  • suitable bases include, without limitation, sodium hydroxide, sodium acetate, ammonium hydroxide, potassium hydroxide, ammonium acetate, potassium acetate, sodium phosphate, potassium phosphate, sodium citrate, sodium formate, sodium sulfate, potassium sulfate, potassium fumerate, and combinations thereof.
  • the amount of any individual excipient in the composition may vary depending on the activity of the excipient and particular needs of the composition.
  • the optimal amount of any individual excipient may be determined through routine experimentation, that is, by preparing compositions containing varying amounts of the excipient (ranging from low to high), examining the stability and other parameters, and then determining the range at which optimal performance is attained with no significant adverse effects.
  • the excipient may be present in the composition in an amount of about 1% to about 99% by weight, from about 5% to about 98% by weight, from about 15 to about 95% by weight of the excipient, with concentrations less than 30% by weight.
  • compositions described herein may be used to treat any bleeding disorder associated with functional defects of FIX or deficiencies of FIX such as a shortened in vivo half-life of FIX, altered binding properties of FIX, genetic defects of FIX, and a reduced plasma concentration of FIX.
  • Genetic defects of FIX comprise, for example, deletions, additions, and/or substitution of bases in the nucleotide sequence encoding FIX.
  • the bleeding disorder may be hemophilia B.
  • Symptoms of such bleeding disorders include, for example, severe epistaxis, oral mucosal bleeding, hemarthrosis, hematoma, persistent hematuria, gastrointestinal bleeding, retroperitoneal bleeding, tongue/retropharyngeal bleeding, intracranial bleeding, and trauma- associated bleeding.
  • modified FIX polypeptides may be administered to a subject susceptible to or otherwise at risk of a disease state or injury to enhance the subject's own coagulative capability. Such an amount may be defined to be a "prophylactically effective dose.”
  • Administration of the modified FIX polypeptides for prophylaxis includes situations where a patient suffering from hemophilia B is about to undergo surgery and the polypeptide is administered between one to four hours prior to surgery.
  • the polypeptides are suited for use as a prophylactic against uncontrolled bleeding, optionally in patients not suffering from hemophilia.
  • the polypeptide may be administered to a patient at risk for uncontrolled bleeding prior to surgery.
  • polypeptides, materials, compositions, and methods described herein are intended to be representative examples of the invention, and it will be understood that the scope of the invention is not limited by the scope of the examples. Those skilled in the art will recognize that the invention may be practiced with variations on the disclosed polypeptides, materials, compositions and methods, and such variations are regarded as within the ambit of the invention.
  • N-linked glycosylation sites is Asn-X-Ser/Thr (where X is any amino acid except proline or perhaps aspartic acid which is also not favorable).
  • X is any amino acid except proline or perhaps aspartic acid which is also not favorable.
  • the amino acids within the activation peptide that are conserved between species was avoided as well as and consensus sequences for other N-linked sites and the consensus sequence for tyrosine sulfation so as not to disrupt these posttranslational modifications since these could be important for the pharmacokinetics of FIX.
  • three sites for adding new N-linked glycosylation sites within the activation peptide were identified as shown in Table 3.
  • the apparent site at which the additional amino acids in rat, mouse, and guinea pig are found can vary such that the site can be either between E160 and A161, between A161 and E162, between E162 and T163, or between T 163 and 1164 of human FIX.
  • N-S-T-Q-D-N-I-T-Q (SEQ ID NO: 2) were inserted in the activation peptide between Al 61 and El 62.
  • the sequences " N-S-T” and “N-I-T” are the N-linked consensus sequences from the natural FIX activation peptide. In both cases, these are followed by a glutamine (Q) and preceded by an aspartic acid (D) in the natural FIX sequence. Therefore, a glutamine as well as an aspartic acid were included in an attempt to emulate the natural site. It is envisioned that additional sequences containing between 1 and 3 consensus sequences for N-glycosylation site (N-X-S/T) may also be inserted between A161 and E162.
  • Table 5 shows the substitutions that were designed to convert O-linked glycosylation sites to N-linked glycosylation sites.
  • HKBl 1 is a human cell line generated by the fusion of HEK293 cells and a B cell lymphoma.
  • each of the glycosylation site substitutions were combined with a second substitution, R338A, as shown for HGl through HG8 in Figure 3.
  • the R338A substitution increases the specific activity of FIX by 3- to 4-fold as measured by the aPTT assay. Additional combinations of two glycosylation site muteins and R338A were also created and tested ( Figure 3).
  • HG4 exhibited increased mobility on the gel demonstrating that this substitution destroyed the O-linked site (thus reducing the molecular weight of the FIX protein), but failed to create a functional N-linked site.
  • HG5 and HG6 exhibited reduced mobility on the gel demonstrating that the substitutions in these clones did create functional N-linked glycosylation sites.
  • Mutein HG7 which also has an O-linked site mutated to an N-linked site exhibited no change in mobility on the gel compared to wild-type FIX or FIX-R338A. Given the fact that the substitution in HG7 would be expected to eliminate an O- linked site and thus increase the mobility on the gel, the finding that there was no change in mobility suggests that the introduced N-linked site may be functional.
  • a pair of PCR primers complementary to sequences at the 5' and 3' ends of the coding region of the human FIX cDNA were designed from the published cDNA sequence (NM_000133).
  • the 5' primer (FIXFl ; ATCATAAGCTTGCCACCATGCAGCGCGTGAACATG (SEQ ID NO: 8), start codon of FIX is in bold text) contained the first 18 nucleotides of the FIX coding region including the ATG start codon preceded by a consensus Kozak sequence (underlined) and a HindIII restriction site.
  • the 3' primer (FIXR3, ATCATAAGCTTGATTAGTTAGTGAGA GGCCCTG) (SEQ ID NO: 9) contained 22 nucleotides of FIX sequence that lies 45 nucleotides 3' of the end of the FIX coding region preceded by a HindIII site.
  • Amplification of first strand cDNA from normal human liver (Stratagene, San Diego CA) using these primers and high fidelity proofreading polymerase (Invitrogen, Carlsbad, CA) resulted in a single band of the expected size for human FIX cDNA (1464 bp). After digestion with HindIII, the PCR product was gel purified and then cloned into the HindIII site of the plasmid pAGE16.
  • underlined residues are those that were changed relative to wild type FIX to create a consensus sequence for N-linked glycosylation.
  • the resulting plasmid was digested with SnaBl and Xbal to remove the 27 bp fragment corresponding to residues Vl 56 to 1164 and then ligated to a double stranded fragment created by annealing of oligonucleotides 2PrimerF and 2PrimerR (Table 8).
  • the sequence of the resulting plasmid was determined by double strand DNA sequencing to have an insertion of 27 bp encoding nine amino acids with the sequence NSTQDNITQ (SEQ ID NO: 2) that contains two consensus sequences for N-linked glycosylation (NXT).
  • the bold/underline residues are those that were changed relative to wild type Factor IX to create either a new restriction site or the insertion of nine additional amino acids encoding two consensus sequences for N-linker glycosylation.
  • HKB 11 cells (a hybrid of HEK293 and a Burkitt B cell lymphoma line, 2B8) were grown in suspension culture on an orbital shaker (100-125 rpm) in a CO 2 (5%) incubator at 37 0 C in serum free media (RF#277) supplemented with 10 ng/mL soluble vitamin K 3 (Sigma- Aldrich, St. Louis, MO) and maintained at a density between 0.25 and 1.5 x 10 6 cells/mL.
  • 293fectinTM reagent (Invitrogen) was mixed gently with 0.2 mL Opti-MEM® I medium and incubated at room temperature for 5 minutes.
  • the diluted 293fectinTM was added to the diluted DNA solution, mixed gently, incubated at room temperature for 20-30 minutes and then added to each well that had been seeded with 5 x 10 6 (4.6 mL) HKBl 1 cells.
  • the cells were then incubated on an orbital rotator (125 rpm) in a CO 2 incubator at 37°C for 3 days after which the cells were pelleted by centrifugation at 1000 rpm for 5 minutes, and the supernatant was collected and stored at 4 0 C.
  • HEK293 cells were transfected with expression constructs for FIX variants in 384 well plates using standard lipofection protocols and commercial reagents. The cells were cultivated for 72 hours post-transfection at which time the supernatant was harvested for further analyses.
  • Example 11 Western Blot for FIX.
  • FIX antigen levels in cell culture supernatants were determined using a FIX ELISA kit (Hyphen Biomed/Aniara, Mason, OH). Cell culture supernatant was diluted in sample diluent buffer (supplied in the kit) to achieve a signal within the range of the standard curve.
  • FIX protein purified from human plasma Hyphen Biomed/Aniara, Catalog No. RK032A, specific activity 196 U/mg
  • sample diluent was used as to create a standard curve from 100 ng/mL to 0.2 ng/mL. Diluted samples and the standards were added to the ELISA plate that is pre-coated with a polyclonal anti-FIX capture antibody.
  • FIX expression levels were also quantitated using commercially available FIX ELISA reagents (Haemochrom Diagnostica GmbH, Essen, Germany) according to the manufacturer's instructions. Wheat germ agglutinin (Sigma- Aldrich, St.
  • FIX coagulation activity was determined using an aPTT assay in FIX deficient human plasma run on a ElectraTM 1800C automatic coagulation analyzer (Beckman Coulter, Fullerton, CA). Briefly, three dilutions of supernatant samples in coagulation diluent were created by the instrument, and 100 ⁇ L was then mixed with 100 ⁇ L FIX deficient plasma (Aniara, Mason, OH) and 100 ⁇ L automated aPTT reagent (rabbit brain phospholipid and micronized silica (bioMerieux, Inc., Durham, NC). After the addition of 100 ⁇ L 25 mM CaCl 2 solution, the time to clot formation was recorded.
  • a standard curve was generated for each run using serial dilutions of the same purified human FIX (Hyphen Biomed/Aniara) used as the standard in the ELISA assay.
  • the standard curve was routinely a straight line with a correlation coefficient of 0.95 or better and was used to determine the FIX activity of the unknown samples.
  • FIX polypeptides The circulating half-life of FIX polypeptides is measured using an in vitro assay. This assay is based on the ability of FIX in vivo and in vitro to mediate the accumulation of adenovirus (Ad) in hepatocytes. Briefly, it has been shown that FIX can bind the Ad fiber knob domain and provide a bridge for virus uptake through cell surface heparin sulfate proteoglycans (HSPG) (Shayakhmetov, et al., J. Virol 79:7478-7491, 2005). An Adenovirus vector mutant, Ad5mut, which contains mutations in the fiber knob domain, does not bind to FIX.
  • Ad5mut An Adenovirus vector mutant, Ad5mut, which contains mutations in the fiber knob domain, does not bind to FIX.
  • Ad5mut has significantly reduced ability to infect liver cells and liver toxicity in vivo, demonstrating that FIX plays a major role in targeting Ad vectors to hepatic cells (Shayakhmetov, et al., 2005).
  • the ability of FIX to target Ad vector to hepatic cells can be blocked by inhibitors of protein-HSPG interactions (Shayakhmetov, et al., 2005).
  • HSPG-mediated uptake of FIX contributes significantly to FIX clearance and consequently, interfering with the HSPG interaction is expected to increase the half-life of FIX. Therefore, in vitro uptake of FIX and/or FIX variants in hepatocytes is measured, and variants with reduced uptake are expected to have increased half-life in vivo.
  • FIX half-life in vitro mammalian cells are incubated with adenovirus in the presence or absence of FIX or FIX variants.
  • Viral uptake is mediated by wild-type FIX and measured by expression of the reporter gene encoded in viral genome, for example, green fluorescent protein (GFP) or luciferase expression.
  • GFP green fluorescent protein
  • luciferase expression Reduced uptake of adenovirus in the presence of FIX variants are measured as reduced reporter gene expression, for example, reduced GFP fluorescence or reduced luciferase enzymatic activity as compared to wild-type FIX.
  • FIX circulating half-life is measured in vivo using standard techniques well-known to those of ordinary skill in the art. Briefly, the respective dose of FIX or FIX variant is administered to a subject by intravenous injection. Blood samples are taken at a number of time points after injection and the FIX concentration is determined by an appropriate assay (e.g.. ELISA). To determine the half-life, that is the time at which the concentration of FIX is half of the concentration of FIX immediately after dosing, the FIX concentration at the various time points is compared to the FIX concentration expected or measured immediately after administering the dose of FIX. A correlation between reduced cellular uptake in the in vitro assay and increased half-life in the in vivo assay is expected.
  • an appropriate assay e.g. ELISA

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Disclosed are modified Factor IX polypeptides such as Factor IX polypeptides with one or more introduced glycosylation sites. The modified Factor IX polypeptides may exhibit increased in vitro or in vivo stability such as a longer plasma half-life. Also, methods are disclosed of making modified Factor IX polypeptides, and methods of using modified Factor IX polypeptides, for example, to treat patients afflicted with hemophilia B.

Description

MODIFIED FACTOR IX POLYPEPTIDES AND USES THEREOF
[001] This application claims benefit of U.S. Provisional Application Serial No. 61/124,567; filed on April 16, 2008, and U.S. Provisional Application Serial No. 61/045,961; filed on April 17, 2008, the contents of which are incorporated herein by reference in their entirety.
FIELD OF THE INVENTION
[002] The invention relates to modified Factor IX polypeptides such as Factor IX polypeptides with one or more introduced glycosylation sites. The modified Factor IX polypeptides may exhibit increased in vitro or in vivo stability such as a longer plasma half-life. The invention also relates to methods of making modified Factor IX polypeptides, and methods of using modified Factor IX polypeptides, for example, to treat patients afflicted with hemophilia B.
BACKGROUND OF THE INVENTION
[003] Hemophilia B effects one out of 34,500 males and is caused by various genetic defects in the gene encoding coagulation Factor IX (FIX) that result in either low or undetectable FIX protein in the blood (Kurachi, et al., Hematol. Oncol. Clin. North Am. 6:991-997, 1992; Lillicrap, Haemophilia 4:350-357, 1998). Insufficient levels of FIX lead to defective coagulation and symptoms that result from uncontrolled bleeding. Hemophilia B is treated effectively by the intravenous infusion of either plasma-derived or recombinant FIX protein either to stop bleeds that have already initiated or to prevent bleeding from occurring (prophylaxis) (Dargaud, et al., Expert Opin. Biol. Ther. 7:651-663; Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005). Effective prophylaxis requires maintaining a minimum trough level of FIX of about 1% of normal levels (Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005). Because of the approximately 18 to 24 hour half-life of native FIX (either plasma-derived or recombinant), FIX levels drop to less than 1% of normal levels within 3 to 4 days following bolus injection which necessitates repeat injection on average every three days to achieve effective prophylaxsis (Giangrande, Expert Opin. Pharmacother. 6:1517-1524, 2005). Such frequent intravenous injection is problematic for patients and is a hurdle for achieving effective prophylaxsis (Petrini, Haemophilia 13 Suppl 2:16-22, 2007), especially in children. A FIX protein with a longer half-life would enable less frequent administration and thus, be of significant medical benefit.
SUMMARY OF THE INVENTION
[004] The application provides FIX polypeptides (also referred to as modified FIX polypeptides, FIX muteins, or FIX variants) comprising amino acid sequences that have been modified by introducing one or more glycosylation sites. In some embodiments, the one or more glycosylation sites may be N-linked glycosylation sites. In some embodiments, the polypeptides have coagulation activity. In some embodiments, the modified polypeptides may comprise at least one substitution such as, but not limited to R338A and V86A. In some embodiments, the modified polypeptides may comprise both the R338A and V86A substitutions.
[005] The application also provides FIX polypeptides comprising amino acid sequences that have been modified by introducing one or more glycosylation sites. The FIX polypeptides may further comprise a carbohydrate chain attached to the one or more introduced glycosylation sites. In some embodiments, the carbohydrate chain may be an N-linked carbohydrate chain. In some embodiments, the carbohydrate chain may have a mammalian carbohydrate chain structure. In some embodiments, the carbohydrate chain may have a human carbohydrate chain structure. In some embodiments, the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites may increase serum half-life of the polypeptide by, for example, at least 30% relative to the polypeptide lacking the introduced glycosylation sites. In some embodiments, the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites does not reduce the amount of secreted polypeptide by, for example, more than 50% relative to the amount of the secreted polypeptide lacking the introduced glycosylation sites. In some embodiments, the attachment of a carbohydrate chain at one or more of the introduced glycosylation sites does not inhibit interaction of the polypeptide with at least one of Factor VIII (FVIII), Factor XI (FXI), or Factor X (FX) by, for example, more than 50% relative to interaction of the polypeptide lacking the carbohydrate chain at the introduced glycosylation sites with FVIII, FXI, or FX. In some embodiments, the modified polypeptide may have a specific activity of, for example, at least 100 units per mg of polypeptide.
[006] The one or more glycosylation sites may be introduced via one or more amino acid substitutions. The substitutions may be at surface exposed residues and/or be limited to substitutions that do not introduce a mutation known to be associated with hemophilia B. Exemplary embodiments include FIX polypeptides comprising one or more substitutions such as, but not limited to:
(a) G4T; E33N; E36T; E36N; R37N; F75N; F77T; E83T; D85N; V86A; K91T; A103T; V107T; K122N; K122T; S138N; A146N; T148N; F150T; P151N; T159N; A161T; A161N; T169N; Q170N; T172N; D177N; D177E; F178T; K201N; K201T; K214T; V223N; G226N; Y226T; K228N; K228T; E239N; E242N; I251T; A262T; E294N; R338A; R338N; K341N; F353N; H354V; H354I; E355T; V370N; T371V; T371I; E372T; E374N; M391N; K392V; G393T; E410N; K413N; L414I;
(b) YlN and S3T; S3N and K5T; G4N and L6T; K5N and E7T; L6N and E8T; E7N and F9T; F9N and Ql IT; VlON and G12T; QI lN and N13T; G12N and L14T; N13 and E15T; L14N and Rl 6T; E15N and E17T; M19N and E21T; E20N and K22T; S24N and E26T; F25N and E27T; E26N and A28T; E27N and R29T; A28N and E30T; R29N and V31T; E30N and F32T; V31N and E33T; F32N and N34T; T35N and R37T; T38N and E40T; T39N and F41T; E40N and W42T; F41N and K43T; W42N and Q44T; K43N and Y45T; Q44N and V46T; Y45N and D47T; V46N and G48T; E52N and N54T; S53N and P55T; G59N and S61T; K63N and D65T; I66N and S68T; S68N and E70T; G76N and E78T; E78N and K80T; E83N and D85T; L84N and V86T; I90N and N92T; KlOON and S102T; S102N and D104T; A103N and N105T; D104N and K106T; K106N and V108T; Rl 16N and Al 18T; El 19N and Q121T; Q121N and S123T; A127N and P129T; V135N and V137T; S136N and S138T; V137N and Q139T; Q139N and S141T; T140N and K142T; S141N and L143T; E147N and V149T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; Y155N and N157T; V156N and S158T; S158N and E160T; T159N and A161T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; L165N and N167T; D166N and I168T; I168N and Q170T; T169N and S171T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; G184N and D186T; E185N and A187T; D186N and K188T; A187N and P189T; P189N and Q191T; G200N and V202T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; V227N and I229T; H236N and I238T; I238N and E240T; E240N and E242T; T241N and H243T; H243N and E245T; K247N and N249T; V250N and R252T; 125 IN and I253T; I253N and P255T; A261N and I263T; A262N and N264T; D276N and P278T; V280N and N282T; F302N and S304T; S304N and Y306T; R312N and F314T; V313N and H315T; F314N and K316T; H315N and G317T; K316N and R318T; G317N and S319T; S319N and L321T; A320N and V322T; R327N and P329T; P329N and V331T; D332N and A334T; L337N and S339T; S339N and K341T; T340N and F342T; T343N and Y345T; G352N and H354T; F353N and E355T; H354N and G356T; E355N and G357T; G356N and R358T; G357N and D359T; E372N and E374T; W385N and E387T; G386N and E388T; A390N and K392T;
(c) D85N, K122T, and I251T; D85N, K122T, and E242N; E125N, P126A, and A127T; P126N, V128T, and P129A; T148N, F150T, and P151A; F150N, P151A, and D152T; P151N, V153T, and A161N; P151N, V153T, and T172N; V153N, Y155T, and E294N; T172N, G226N, and K228T; F353N, H354V, and E355T; F353N, H354I, and E355T; V370N, T371V, and E372T; V370N, T371I, and E372T; M391N, K392V, and G393T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; T148N, F150T, G226N, and K228T; P151N, V153T, T172N, and R338A; P151N, V153T, D177E, and F178T; P151N, V153T, G226N, and K228T; T172N, G226N, K228T, and R338A; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; K122T, P151N, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; T148N, F150T, G226N, K228T, and R338A; P151N, V153T, T172N, G226N, and K228T; P151N, V153T, D177E, F178T, and R338A; P151N, V153T, G226N, K228T, and R338A; and P151N, V153T, T172N, G226N, K228T, and R338A; and any combination thereof.
[007] The one or more glycosylation sites may be introduced into the catalytic domain of FIX or activation peptide. Exemplary embodiments include FIX polypeptides comprising one or more substitutions such as, but not limited to: R37N; D85N; K122T; S138N; A146N; A161N; Q170N; T172N; D177N; F178T; K201N; K228N; E239N; E242N; I251T; A262T; E294N; E374N; and E410N. Other embodiments may comprise the following FIX polypeptides comprising one or more substitutions such as: G59N and S61T; K63N and D65T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; E119N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; V156N and S158T; S158N and E160T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; D166N and I168T; I168N and Q170T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; T241N and H243T; 125 IN and I253T; I253N and P255T; A262N and N264T; V280N and N282T; T343N and Y345T; and E372N and E374T; Fl 5ON, Pl 5 IA, and D152T. In some embodiments, the modified polypeptides may further comprise at least one substitution such as, for example, R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
[008] The one or more glycosylation sites may be introduced by converting an O-linked glycosylation site to an N-linked glycosylation site. Exemplary embodiments include substitutions such as, but not limited to T169N; T172N; T148N and F150T; and T159N and A161T. In some embodiments, the substitutions may be T172N; T148N and F150T; and T159N and A161T. In some embodiments, the modified polypeptides may further comprise at least one substitution such as, but not limited to R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
[009] The one or more glycosylation sites may be introduced by inserting between 1 and 12 amino acid residues between amino acid residues 160-164. The glycosylation sites may be introduced between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163-164. In some embodiments, SEQ ID NO: 2 may be introduced between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163-164. In other embodiments, glycosylation sites may be introduced by adding between 1 and 12 amino acid residues to the C-terminus of the FIX polypeptide. In some embodiments, SEQ ID NOs: 4, 5, 6, or 7 may be introduced to the to the C-terminus of the FIX polypeptide. In some embodiments, the polypeptide may further comprise amino acid substitutions D177E and F178T. In some embodiments, the polypeptide may further comprise amino acid substitutions P151N and V153T. In some embodiments, the polypeptide may further comprise amino acid substitution T172N. In some embodiments, the polypeptide may further comprise amino acid substitutions Pl 5 IN, V153T, and T172N. In some embodiments, the polypeptide may further comprise amino acid substitutions T148N and Fl 5OT. In some embodiments, the polypeptide may further comprise amino acid substitutions G226N and K228T. In some embodiments, the modified polypeptides may further comprise at least one substitution such as R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
[010] In exemplary embodiments, modified FIX polypeptides are provided comprising one or more substitutions such as: D85N; K122T; S138N; T172N; K201N; K228N; E239N; E242N; I251T; A262T; E294N; G59N and S61T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; E119N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; S158N and E160T; E162N and I164T; T163N and L165T; T172N and S174T; Q173N and F175T; K201N and D203T; T225N and V227T; G226N and K228T; I253N and P255T; A262N and N264T; V280N and N282T; E372N and E374T; F150N, P151A, and D152T; an insertion of SEQ ID NO:2 between Al 61 and E 162; and G226N, K228T, and an insertion of SEQ ID NO:2 between A161 and E162. In some embodiments, the modified polypeptides may further comprise at least one substitution such as, for example, R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
[011] The application also provides FIX polypeptides comprising an R338A substitution and a V86A substitution. In some embodiments, the polypeptide may have a specific activity of at least 700 units per mg of polypeptide.
[012] The application also provides pharmaceutical preparations comprising modified FIX polypeptides and a pharmaceutically acceptable carrier.
[013] The application also provides methods for treating hemophilia B comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical preparations described herein.
[014] The application also provides DNA sequences encoding modified polypeptides, as well as eukaryotic host cells transfected with the DNA sequences. [015] The application also provides methods for producing modified FIX polypeptides comprising (i) modifying the amino acid sequence of the polypeptide by introducing one or more glycosylation sites; (ii) expressing the polypeptide in a manner which allows glycosylation at the one or more glycosylation sites; and (iii) purifying the polypeptide.
BRIEF DESCRIPTION OF THE DRAWINGS
[016] Figure 1 depicts a multiple sequence alignment of FIX sequences within the activation peptide from eight species
[017] Figure 2 depicts Western Blot analysis of media from HKBl 1 cells transfected with glycosylation site muteins of FIX. FIX protein was detected using an anti-FIX-HRP antibody.
[018] Figure 3 depicts a table of expression and activity of FIX glycosylation site muteins in HKBI l cells. Expression was determined by ELISA and activity by aPTT assay. Specific activity is calculated as international units per mg of FIX protein. Values were converted to a percentage of FIX-R338A run in the same transfection experiment, (none): no change in mobility, (+): decreased mobility with the number of + indicating degree, (-): increased mobility.
[019] Figure 4 depicts expression level, coagulation activity, and specific activity of FIX glycosylation site muteins in HKBl 1 cells. Values are expressed as a percentage of the FIX- R338A mutein. Constructs are described in Figure 3.
[020] Figure 5 depicts Western Blot analysis of media from HKBl 1 cells transfected with the G226N, K228T glycosylation site mutein of FIX. FIX protein was detected using an anti-FIX- HRP antibody.
DESCRIPTION OF THE INVENTION
[021] The present application provides FIX polypeptides that include one or more O- or N- linked glycosylation sites. Increased glycosylation of therapeutic proteins may be used to achieve 1) reduced immunogenicity; 2) less frequent administration of the protein; 3) increased protein stability such as increased serum half-life; and 4) reduction in adverse side effects such as inflammation.
[022] The application provides variants of human FIX with one or more additional glycosylation sites. The modified FIX polypeptides may also have an increased plasma half-life that would provide, for example, an extended time of protection against bleeding in hemophilia B patients. The modified FIX polypeptides would enable hemophilia B patients to achieve protection against bleeding with fewer injections of FIX than is possible with the currently available therapy of wild type FIX protein.
[023] The application provides a number of exemplary variants of FIX in which functional glycosylation sites were created in both the catalytic domain and in the activation peptide. Moreover, the application demonstrates that these variants may be expressed in mammalian cells, have increased apparent molecular weight indicative of increased glycosylation, and maintain between 50% and 100% activity in a coagulation assay. Finally, these modification sites may be combined with alterations that enhance the specific activity of FIX, including but not limited to the R338A substitution and/or the V86A substitution (Chang, et al., J. Biol. Chem. 273:12089-12094, 1998 and Chang, et al., J. Biol. Chem. 277:25393-25399, 2002). The combination with one or both of the R338A and V86A substitutions compensates for any reduction in activity resulting from the addition of glycosylation sites such that the specific activity of the modified polypeptides may be similar to or higher than that of wild type FIX.
[024] Once expressed, wild type FIX is a single chain glycoprotein of about 55,000 Daltons. It can structurally be considered as having four domains: the GIa or gamma carboxyglutamate-rich domain; the EGF-like regions; the activation peptide; and the catalytic domain containing the active site (Thomson, Blood 67:565-572, 1986). FIX is synthesized in the liver as a single chain polypeptide of 461 amino acids and undergoes extensive posttranslational modification during passage through the golgi and endoplasmic reticulum (Nemerson, et al., CRC Crit. Rev. Biochem. 9:45-48, 1980; Stenflo, et al., Annu. Rev. Biochem. 46:157-172, 1977). Both the signal sequence and the propeptide are removed resulting in a mature protein of 415 amino acids (SEQ ID NO: 1) (Choo, et al., Nature 299:178-180, 1982; Kurachi, et al., Proc. Natl. Acad. Sci. USA 79:6461- 6464, 1982). Efficient gamma carboxylation is essential for the coagulation activity of FIX and in humans 12 GIa residues are generated within the N terminal GIa domain, although gamma carboxylation on Gla36 and Gla40 are not required for function (DiScipio, et al., Biochemistry 18:899-904, 1979; Gillis, et al., Protein Sci. 6:185-196, 1997). In addition, FIX contains two N- linked glycosylation sites (N157, N167), six O-linked glycosylation sites (S53, S61, T159, T169, T172, T179), and one site each for Ser phosphorylation (S158), tyrosine sulfation (Y155) and β- hydroxylation (D64) (McMullen, et al., Biochem. Biophys. Res. Comm. 115:8-14, 1983).
[025] Activated Factor VII (FVII) initiates the normal hemostatic process by forming a complex with tissue factor (TF), exposed as a result of injury to the vessel wall. The complex subsequently activates FIX; the active form referred to as FIXa. The activation peptide of FIX is removed by proteolytic cleavage at two sites by either Factor XIa (FXIa) or the tissue factor (TF)/Factor Vila complex to generate the catalytically active molecule, Factor IXa (FIXa). FIXa and Factor Villa (FVIIIa) convert FX to Factor Xa (FXa), which in turn converts prothrombin to thrombin. Thrombin then converts fibrinogen to fibrin resulting in formation of a fibrin clot.
[026] As wild-type FIX has numerous post-translational modifications some of which have been suggested to play a role in the in vivo pharmacokinetic profile, an ectopic glycosylation site may be introduced at a position that does not affect these other modifications. Once produced, FIX should retain enzymatic activity and interact with FVIII, FXI, and FX in order to be an effective treatment for hemophilia B. The introduced glycosylation site should not perturb these interactions and function. The application provides, in part, modifications to FIX which are likely to result in an increased number of glycosylation sites with minimal perturbation of function and thus have utility for increasing the bioavailability of FIX. Finally, these modification sites may be combined with alterations that enhance the specific activity of FIX, including but not limited to the R338A substitution and/or the V86A substitution. Alterations that enhance the specific activity of FIX may compensate for potential loss of coagulation activity and also potentially prolong the efficacy of modified molecules by conferring efficacy at lower levels of protein.
Modified FIX Polypeptides
[027] The application provides FIX polypeptides comprising one or more introduced glycosylation sites, that is, modified FIX polypeptides. "Factor IX" as used herein refers to a human plasma FIX glycoprotein that is a member of the intrinsic coagulation pathway and is essential to blood coagulation. It is to be understood that this definition includes native as well as recombinant forms of the human plasma FIX glycoprotein. Unless otherwise specified or indicated, as used herein FIX means any functional human FIX protein molecule in its normal role in coagulation, including any fragment, analogue, variant, and derivative thereof. The terms "fragment," "derivative," "analogue," "mutein," and "variant," when referring to the polypeptides of the application, means fragments, derivatives, analogues, muteins, and variants of the polypeptides which retain substantially the same biological function or activity.
[028] Non-limiting examples of FIX polypeptides include FIX, FIXa, and truncated versions of FIX having FIX activity. Biologically active fragments, deletion variants, substitution variants, or addition variants of any of the foregoing that maintain at least some degree of FIX activity can also serve as a FIX polypeptide. In some embodiments, the FIX polypeptides may comprise an amino acid sequence at least about 70, 80, 90, or 95% identical to SEQ ID NO: 1. In some embodiments, the modified FIX polypeptides are biologically active. Biological activity can be determined, for example, by coagulation assays described herein.
[029] Modified FIX polypeptides may also contain conservative substitutions of amino acids. A conservative substitution is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties and include, for example, the changes of alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine. In some embodiments, the FIX polypeptides of SEQ ID NO: 1 comprise from 1-30, from 1 -20, or from 1-10 conservative amino acid substitutions in addition to the introduction of one or more glycosylation sites.
[030] The single letter abbreviation for a particular amino acid, its corresponding amino acid, and three letter abbreviation are as follows: A, alanine (Ala); C, cysteine (Cys); D, aspartic acid (Asp); E, glutamic acid (GIu); F, phenylalanine (Phe); G, glycine (GIy); H, histidine (His); I, isoleucine (He); K, lysine (Lys); L, leucine (Leu); M, methionine (Met); N, asparagine (Asn); P, proline (Pro); Q, glutamine (GIn); R, arginine (Arg); S, serine (Ser); T, threonine (Thr); V, valine (VaI); W, tryptophan (Trp); Y, tyrosine (Tyr); and norleucine (NIe).
[031] Glycosylation of polypeptides is typically either N-linked or O-linked. N-linked refers to the attachment of a carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences Asn-X-Ser and Asn-X-Thr, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the Asn side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential N- linked glycosylation site. An exemplary N-linked glycosylation site useful for the invention may also be represented as follows Xl-Asn-X2-X3-X4; where Xl is optionally Asp, VaI, GIu, GIy, or He; X2 is any amino acid except Pro; X3 is Ser or Thr; and X4 is optionally VaI, GIu, GIy, GIn, or He. In some embodiments, Xl is optionally Asp; X2 is Ser; X3 is Thr; and X4 is GIn. In some embodiments, Xl is Asp; X2 is He; X3 is Thr; and X4 is GIn. Addition of N-linked glycosylation sites to a FIX polypeptide is accomplished by altering the amino acid sequence such that one or more of the above-described tripeptide sequences is introduced.
[032] O-linked glycosylation refers to the attachment of one of the sugars N- aceytlgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly to serine or threonine, although attachment to 5-hydroxyproline or 5-hydroxylysine is also possible. Addition of O-linked glycosylation sites to a FIX polypeptide may be accomplished by altering the amino acid sequence such that one or more Ser or Thr residues are introduced.
[033] Glycosylation sites may be introduced, for example, by deleting one or more amino acid residues, substituting one or more endogenous FIX amino acid residues with another amino acid(s), or adding one or more amino acid residues. The addition of an amino acid residue may be either between two existing amino acid residues or at the N- or C-terminal end of the native FIX molecule.
[034] The terminology for amino acid substitutions used is as follows. The first letter represents the amino acid residue naturally present at a position of human FIX. The following number represents the position in the mature human FIX amino acid sequence (SEQ ID NO:1). The second letter represent the different amino acid substituting for (rφlacing/substituting) the natural amino acid. As an example, R338A denotes that the Arg residue at position 338 of SEQ ID NO: 1 has been replaced with an Ala residue. With respect to SEQ ID NO: 26 which includes an additional 5' amino acid sequence (46 amino acids) of human FIX polypeptide, position 338 of SEQ ID NO: 1 corresponds to position 384 of SEQ ID NO: 26.
[035] The FIX residue number system used herein refers to that of the mature human FIX protein in which residue 1 represents the first amino acid of the mature FIX polypeptide following removal of both the signal sequence and the propeptide. Native or wild type FIX is the full length mature human FIX molecule as shown in SEQ ID NO: 1.
[036] In some embodiments, the glycosylation sites are engineered in FIX at locations that will not abolish the function of the protein or its expression in cells. In order to design FIX polypeptides comprising one or more introduced glycosylation sites, several criteria may be applied. In some embodiments, the introduced glycosylation site is surface exposed. Surface exposure can be determined based on the solvent accessible surface area as determined in Autin, et al., (J. Thromb. Haemost. 3:2044-56, 2005). In some embodiments, the introduced glycosylation site does not introduce a mutation known to be associated with hemophilia B. Known mutations can be found on the world wide web at kcl.ac.uk/ip/petergreen/haemBdatabase.html and in Table 1.
TABLE 1
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
[037] It may be desirable to compare the properties of the modified FIX polypeptides having one or more introduced glycosylation sites to a control polypeptide. Properties for comparison include, for example, solubility, activity, plasma half-life, glycosylation state, and binding properties. In some embodiments, the modified FIX polypeptides may be glycosylated. It is within the purview of one skilled in the art to select the most appropriate control polypeptide for comparison. In some embodiments, the control polypeptide may be identical to the modified polypeptide except for the one or more introduced glycosylation sites. Exemplary polypeptides include wild-type FIX polypeptide and FIX polypeptides comprising one or more activating substitutions, such as R338A and/or V86A.
[038] One aspect of the application provides modified FIX polypeptides having increased in vitro or in vivo stability over a control polypeptide. Enhanced serum half-life and in vivo stability may be desirable to reduce the frequency of dosing that is required to achieve therapeutic effectiveness. Accordingly, in certain embodiments, the glycosylated FIX polypeptides have a serum half-life increased by about 20, 30, 40, 60, 80, 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, or 1000% relative to a control protein. In some embodiments, the modified FIX polypeptides may have a serum half-life of at least one, at least two, at least three, at least four, at least five, at least ten, or at least twenty days or more.
[039] The term "half-life," as used herein in the context of administering a polypeptide drug to a patient, is defined as the time required for plasma concentration of a drug in a patient to be reduced by one half. Methods for pharmacokinetic analysis and determination of half-life and in vivo stability will be familiar to those skilled in the art. Details may be found in Kenneth, et al., Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters, et al., Pharmacokinetc analysis: A Practical Approach (1996). Reference is also made to "Pharmacokinetics," M Gibaldi & D Perron, published by Marcel Dekker, 2nd Rev. edition (1982), which describes pharmacokinetic parameters such as t-alpha and t-beta half lives and area under the curve (AUC). [040] The activity of modified FIX polypeptides may be described either as an absolute value, such as in units, or as a percentage of the activity of a control polypeptide. In some embodiments, the modified FIX polypeptides may have a specific activity that is not reduced more than about 10, 20, 30, 40, 50, 60, 70, or 80% relative to a control protein. For example, a modified FIX polypeptide may have a specific activity that is not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide maintains at least about 20% of the specific activity as compared to the specific activity of the control. FIX specific activity may be defined as the ability to function in the coagulation cascade, induce the formation of FXa via interaction with FVIIIa on an activated platelet, or support the formation of a blood clot. The activity may be assessed in vitro by techniques such as clot analysis, as described in, for example, McCarthy, et al., (Thromb. Haemost. 87:824-830,2002), and other techniques known to those skilled in the art. The activity may also be assessed in vivo using one of the several animal lines that have been intentionally bred with a genetic mutation for hemophilia B such that an animal produced from such a line is deficient for FIX. Such lines are available from a variety of sources such as, without limitation, the Division of Laboratories and Research, New York Department of Public Health, Albany, N. Y. and the Department of Pathology, University of North Carolina, Chapel Hill, N.C. Both of these sources, for example, provide canines suffering from canine hemophilia B. Alternatively, mice deficient in FIX are also available (Sabatino, et al., Blood 104:2767-2774, 2005). In order to test for FIX activity, a test polypeptide is injected into the diseased animal, a small cut made and bleeding time compared to a healthy control.
[041 ] Human wild-type FIX has a specific activity of around 200 units per mg. One unit of FIX has been defined as the amount of FIX present in one millilitre of normal (pooled) human plasma (corresponding to a FIX level of 100%). In some embodiments, the modified FIX polypeptides may have a specific activity of at least about 100 units per mg of FIX polypeptide. In some embodiments, the modified FIX polypeptides may have a specific activity of at least about 120, 140, 160, 180, 200, 220, 240, 260 units, or more per mg of FIX polypeptide. In some embodiments, the specific activity of FIX may be measured using the APTT or activated partial thromboplastin time assay (described by, e.g., Proctor, et al., Am. J. Clin. Pathol. 36:212, 1961 and see Examples).
[042] When expressed in cells, such as liver or kidney cells, FIX polypeptide may be synthesized by the cellular machinery, undergoes posttranslational modification, and is then secreted by the cells into the extracellular milieu. The amount of FIX polypeptide secreted from cells is therefore dependent on both processes of protein translation and extracellular secretion. In some embodiments, the modified FIX polypeptides may be secreted in an amount that is not reduced more than about 10, 20, 30, 40, 50, 60, 70, or 80% relative to the amount secreted of a control protein. For example, a modified FIX polypeptide may be secreted in an amount that is not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide is secreted in an amount of at least about 20% as compared to the control. The amount of FIX polypeptide secreted may be measured, for example, by determining the protein levels in the extracellular medium using any art-known method. Traditional methodologies for protein quantification include 2-D gel electrophoresis, mass spectrometry, and antibody binding. Exemplary methods for assaying protein levels in a biological sample include antibody-based techniques, such as immunoblotting (western blotting), immunohistological assay, enzyme linked immunosorbent assay (ELISA), or radioimmunoassay (RIA).
[043] In some embodiments, the modified FIX polypeptides interact with at least one of FVIII, FXI, or FX at a level not reduced more than about 40, 50, 60, 70, or 80% relative to the interaction of a control protein with at least one of FVIII, FXI, or FX. For example, a modified FIX polypeptide may interact with at least one of FVIII, FXI, or FX at a level not reduced more than about 80% relative to a control FIX polypeptide, if the modified polypeptide interacts with at least one of FVIII, FXI, or FX at a level of at least about 20% as compared to the control. The binding of FIX to other members of the coagulation cascade can be determined by any method known to one skilled in the art, including for example, the methods described in Chang, et al., (J. Biol. Chem. 273:12089-12094, 1998).
[044] As previously described, FIX is composed of four structural domains with different biological functions. One aspect of the invention is to provide FIX polypeptides that are modified in particular domains, such as in the activation peptide and in the catalytic domain. The application provides, in part, FIX polypeptides comprising one more glycosylation sites introduced into the activation peptide of FIX. The natural glycosylation sites in FIX are located in the EGFl domain (two O-linked sites on Ser53 and Serόl) and in the activation peptide (four O-linked sites at Thrl59, Thrl69, Thrl72, and Thrl79 and 2 N-linked sites at Asnl57 and Asn 167). Thus, six of the eight natural glycosylation sites are located in the activation peptide. Introduced glycosylation sites may have less of a negative impact on activity or expression when introduced into regions of FIX that have natural glycosylation sites, such as the activation peptide. In addition, the activation peptide is cleaved when FIX is activated and so is not required for the catalytic activity of FIXa. However, the activation peptide is present in the zymogen, making the activation peptide an attractive region to introduce glycosylation sites to improve the circulating half-life of the zymogen.
[045] The application also provides, in part, FIX polypeptides comprising one or more glycosylation sites. In some embodiments, FIX polypeptides are provided comprising one or more substitutions selected from G4T; E33N; E36T; E36N; R37N; F75N; F77T; E83T; D85N; V86A; K91T; A103T; V107T; K122N; K122T; S138N; A146N; T148N; F150T; P151N; T159N; A161T; A161N; T169N; Q170N; T172N; D177N; D177E; F178T; K201N; K201T; K214T; V223N; G226N; Y226T; K228N; K228T; E239N; E242N; I251T; A262T; E294N; R338A; R338N; K341N; F353N; H354V; H354I; E355T; V370N; T371V; T371I; E372T; E374N; M391N; K392V; G393T; E410N; K413N; L414I; or any combination thereof.
[046] In some embodiments, FIX polypeptides are provided comprising one or more substitutions selected from YlN and S3T; S3N and K5T; G4N and L6T; K5N and E7T; L6N and E8T; E7N and F9T; F9N and Ql IT; VlON and G12T; QI lN and N13T; G12N and L14T; N13 and E15T; L14N and R16T; E15N and E17T; M19N and E21T; E20N and K22T; S24N and E26T; F25N and E27T; E26N and A28T; E27N and R29T; A28N and E30T; R29N and V3 IT; E30N and F32T; V3 IN and E33T; F32N and N34T; T35N and R37T; T38N and E40T; T39N and F41T; E40N and W42T; F41N and K43T; W42N and Q44T; K43N and Y45T; Q44N and V46T; Y45N and D47T; V46N and G48T; E52N and N54T; S53N and P55T; G59N and S61T; K63N and D65T; I66N and S68T; S68N and E70T; G76N and E78T; E78N and K80T; E83N and D85T; L84N and V86T; I90N and N92T; KlOON and S102T; S102N and D104T; A103N and N105T; D104N and K106T; K106N and V108T; Rl 16N and Al 18T; El 19N and Q121T; Q121N and S123T; A127N and P129T; V135N and V137T; S136N and S138T; V137N and Q139T; Q139N and S141T; T140N and K142T; S141N and L143T; E147N and V149T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; Y155N and N157T; V156N and S158T; S158N and E160T; T159N and A161T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; L165N and N167T; D166N and I168T; I168N and Q170T; T169N and S171T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; G184N and D186T; E185N and A187T; D186N and K188T; A187N and P189T; P189N and Q191T; G200N and V202T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; V227N and I229T; H236N and I238T; I238N and E240T; E240N and E242T; T241N and H243T; H243N and E245T; K247N and N249T; V250N and R252T; 125 IN and I253T; I253N and P255T; A261N and I263T; A262N and N264T; D276N and P278T; V280N and N282T; F302N and S304T; S304N and Y306T; R312N and F314T; V313N and H315T; F314N and K316T; H315N and G317T; K316N and R318T; G317N and S319T; S319N and L321T; A320N and V322T; R327N and P329T; P329N and V331T; D332N and A334T; L337N and S339T; S339N and K341T; T340N and F342T; T343N and Y345T; G352N and H354T; F353N and E355T; H354N and G356T; E355N and G357T; G356N and R358T; G357N and D359T; E372N and E374T; W385N and E387T; G386N and E388T; A390N and K392T; or any combination thereof
[047] In some embodiments, FIX polypeptides are provided comprising one or more substitutions selected from D85N, K122T, and I251T; D85N, K122T, and E242N; E125N, P126A, and A127T; P126N, V128T, and P129A; T148N, F150T, and P151A; F150N, P151A, and D152T; P151N, V153T, and A161N; P151N, V153T, and T172N; V153N, Y155T, and E294N; T172N, G226N, and K228T; F353N, H354V, and E355T; F353N, H354I, and E355T; V370N, T371 V, and E372T; V370N, T371I, and E372T; M391N, K392V, and G393T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; T148N, F150T, G226N, and K228T; P151N, V153T, T172N, and R338A; P151N, V153T, D177E, and F178T; P151N, V153T, G226N, and K228T; T172N, G226N, K228T, and R338A; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; K122T, Pl 5 IN, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; T148N, F150T, G226N, K228T, and R338A; P151N, V153T, T172N, G226N, and K228T; P151N, V153T, D177E, F178T, and R338A; P151N, V153T, G226N, K228T, and R338A; and P151N, V153T, T172N, G226N, K228T, and R338A; or any combination thereof
[048] In some embodiments, FIX polypeptides are provided comprising one or more substitutions selected from
(a) G4T; E33N; E36T; E36N; R37N; F75N; F77T; E83T; D85N; V86A; K91T; A103T; V107T; K122N; K122T; S138N; A146N; T148N; F150T; P151N; T159N; A161T; A161N; T169N; Q170N; T172N; D177N; D177E; F178T; K201N; K201T; K214T; V223N; G226N; Y226T; K228N; K228T; E239N; E242N; I251T; A262T; E294N; R338A; R338N; K341N; F353N; H354V; H354I; E355T; V370N; T371V; T371I; E372T; E374N; M391N; K392V; G393T; E410N; K413N; L414I;
(b) YlN and S3T; S3N and K5T; G4N and L6T; K5N and E7T; L6N and E8T; E7N and F9T; F9N and Ql IT; VlON and G12T; Ql IN and N13T; G12N and L14T; N13 and E15T; L14N and R16T; E15N and E17T; M19N and E21T; E20N and K22T; S24N and E26T; F25N and E27T; E26N and A28T; E27N and R29T; A28N and E30T; R29N and V31T; E30N and F32T; V31N and E33T; F32N and N34T; T35N and R37T; T38N and E40T; T39N and F41T; E40N and W42T; F41N and K43T; W42N and Q44T; K43N and Y45T; Q44N and V46T; Y45N and D47T; V46N and G48T; E52N and N54T; S53N and P55T; G59N and S61T; K63N and D65T; I66N and S68T; S68N and E70T; G76N and E78T; E78N and K80T; E83N and D85T; L84N and V86T; I90N and N92T; KlOON and S102T; S102N and D104T; A103N and N105T; D104N and K106T; K106N and V108T; R116N and A118T; E119N and Q121T; Q121N and S123T; A127N and P129T; V135N and V137T; S136N and S138T; V137N and Q139T; Q139N and S141T; T140N and K142T; S141N and L143T; E147N and V149T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; Y155N and N157T; V156N and S158T; S158N and E160T; T159N and A161T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; L165N and N167T; D166N and I168T; I168N and Q170T; T169N and S171T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; G184N and D186T; E185N and A187T; D186N and K188T; A187N and P189T; P189N and Q191T; G200N and V202T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; V227N and I229T; H236N and I238T; I238N and E240T; E240N and E242T; T241N and H243T; H243N and E245T; K247N and N249T; V250N and R252T; I251N and I253T; I253N and P255T; A261N and I263T; A262N and N264T; D276N and P278T; V280N and N282T; F302N and S304T; S304N and Y306T; R312N and F314T; V313N and H315T; F314N and K316T; H315N and G317T; K316N and R318T; G317N and S319T; S319N and L321T; A320N and V322T; R327N and P329T; P329N and V331T; D332N and A334T; L337N and S339T; S339N and K341T; T340N and F342T; T343N and Y345T; G352N and H354T; F353N and E355T; H354N and G356T; E355N and G357T; G356N and R358T; G357N and D359T; E372N and E374T; W385N and E387T; G386N and E388T; A390N and K392T;
(c) D85N, K122T, and I251T; D85N, K122T, and E242N; E125N, P126A, and A127T; P126N, V128T, and P129A; T148N, F150T, and P151A; F150N, P151A, and D152T; P151N, V153T, and A161N; P151N, V153T, and T172N; V153N, Y155T, and E294N; T172N, G226N, and K228T; F353N, H354V, and E355T; F353N, H354I, and E355T; V370N, T371V, and E372T; V370N, T371I, and E372T; M391N, K392V, and G393T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; T148N, F150T, G226N, and K228T; P151N, V153T, T172N, and R338A; P151N, V153T, D177E, and F178T; P151N, V153T, G226N, and K228T; T172N, G226N, K228T, and R338A; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; K122T, P151N, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; T148N, F150T, G226N, K228T, and R338A; P151N, V153T, T172N, G226N, and K228T; P151N, V153T, D177E, F178T, and R338A; P151N, V153T, G226N, K228T, and R338A; and P151N, V153T, T172N, G226N, K228T, and R338A; and any combination thereof.
[049] The application provides, in part, FIX polypeptides comprising one more glycosylation sites introduced by converting an endogenous O-linked glycosylation site to an N-linked glycosylation site. It has been reported that N-linked glycosylation sites are more likely to be sialylated than O-linked glycosylation sites and there is evidence that higher sialic acid content confers increased protein half-life. It is generally believed that the increased sialic acid content provided by additional N-linked glycosylation may be responsible for the increased half-life in blood (White, et al., Thromb. Haemost. 78:261-265, 1997). Exemplary embodiments of such modified FIX polypeptides are as follows. In some embodiments, FIX polypeptides are provided comprising a T169N substitution. In some embodiments, FIX polypeptides are provided comprising a T172N substitution. In some embodiments, FIX polypeptides are provided comprising a T148N substitution and an F 150T substitution. In some embodiments, FIX polypeptides are provided comprising a T159N substitution and an A161T substitution.
[050] Another aspect of the invention provides for the insertion of amino acids into the activation peptide in order to generate one or more glycosylation sites. The application provides, in part, FIX polypeptides comprising one more glycosylation sites introduced between amino acid residues 160 to 164 of human FIX. In some embodiments, the amino acid residues introduced include a glycosylation site. In some embodiments, the amino acid residues introduced form a glycosylation site in combination with wild-type FIX amino acid residues. The multiple sequence alignment of the FIX sequence from 8 species demonstrated that the mouse, rat, and guinea pig sequences all have additional amino acids (between 7 and 10 residues) in the activation peptide that are not found in other species (human, rhesus, dog, rabbit, pig) (Figure 1). These additional sequences are located between E 160 and E 162 of human FIX. This suggests that insertion of at least 10 amino acid residues is tolerated in the FIX structure at this site. This region was targeted for the introduction of additional amino acids and was found to be a good location as activity was not significantly reduced. In some embodiments, up to 30, 25, 20, 18, 16, 14, or 12 amino acid residues may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, up to 10 amino acids may be inserted. In some embodiments, up to 9 amino acid may be inserted.
[051] Depending upon the criteria used to perform the multiple sequence alignment between FIX from the eight species, the apparent site at which the additional amino acids in rat, mouse, and guinea pig are found can vary such that the site can be either between E160 and A161, between A161 and E162, between E162 and T163, or between T163 and 1164 of human FIX. In some embodiments, the amino acid sequence inserted between El 60 and Al 61 may be SEQ ID NO: 2. In some embodiments, the amino acid sequence inserted between Al 61 and E 162 may be SEQ ID NO: 2. In some embodiments, the amino acid sequence inserted between E162 and T163 may be SEQ ID NO: 2. In some embodiments, the amino acid sequence inserted between T163 and 1164 may be SEQ ID NO: 2. In some embodiments, one or more amino acids may be inserted between E160 and A161 and one or more amino acids may be inserted between A161 and E162. In some embodiments, one or more amino acids may be inserted between E160 and A161 and one or more amino acids may be inserted between E 162 and Tl 63. In some embodiments, one or more amino acids may be inserted between A161 and E162 and one or more amino acids may be inserted between E162 and T163. In some of the above described embodiments, one or more amino acids may be inserted between T163 and 1164. In some embodiments, up to about 30, 25, 20, 18, 16, 14, or 12 total amino acid residues may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, up to about 10 total amino acids may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, up to about 9 total amino acids may be inserted between amino acid residues 160 to 164 of human FIX. In some embodiments, one, two, three, or four glycosylation sites may be introduced.
[052] The application further provides modified FIX polypeptides comprising more than one of the introduced glycosylation sites disclosed herein. FIX polypeptides are provided that comprise at least one introduced glycosylation site in the catalytic domain and at least one introduced glycosylation site in the activation peptide. FIX polypeptides are provided that comprise at least two introduced glycosylation sites in the catalytic domain. FIX polypeptides are provided that comprise at least two introduced glycosylation sites in the activation peptide. In some embodiments, the FIX polypeptides may comprise one or more of the following substitutions: R37N; D85N; K122T; S138N; A146N; A161N; Q170N; T172N; D177N; F178T; K201N; K228N; E239N; E242N; 125 IT; A262T; E294N; E374N; and E410N. Other embodiments may comprise one or more of the following substitutions: G59N and S61T; K63N and D65T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; El 19N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; V156N and S158T; S158N and E160T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; D166N and I168T; I168N and Q170T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; T241N and H243T; 125 IN and I253T; I253N and P255T; A262N and N264T; V280N and N282T; T343N and Y345T; E372N and E374T; F 150N, P151A, and D152T; and insertion of SEQ ID NO:2 between A161 and E162. In some embodiments, the modified polypeptides may further comprise at least one substitution such as, for example, R338A and V86A. In some embodiments, the modified polypeptides may further comprise both the R338A and V86A substitutions.
[053] The application further provides modified FIX polypeptides that comprise at least one introduced glycosylation site at the C-terminus of the FIX polypeptide (i.e., following amino acid residue 415 of the FIX polypeptide). FIX polypeptides are provided that comprise at least two, at least three, at least four, or more glycosylation sites at the C-terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 4 at the C-terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 5 at the C- terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 6 at the C-terminus of the FIX polypeptide. In some embodiments, the FIX polypeptide may comprise the addition of the amino acid sequence of SEQ ID NO: 7 at the C-terminus of the FIX polypeptide. [054] One aspect of the application provides modified FIX polypeptides comprising at least one or more glycosylation sites and one or more substitutions that increase the activity of FIX. Examples of activating substitutions include the R338A and the V86A substitutions. In some embodiments, modified FIX polypeptides may comprise the R338A substitution. In some embodiments, modified FIX polypeptides may comprise the V86A substitution. In some embodiments, modified FIX polypeptides may comprise both the R338A and the V86A substitution.
[055] A further aspect of the application provides FIX polypeptides with increased specific activity. In some embodiments, FIX polypeptides may comprise an R338A substitution and a V86A substitution. In some embodiments, the polypeptides may have a specific activity of at least about 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1400, 1600, 1800, or 2000 units per mg of polypeptide. The specific activity can be determined as previously described, such as, for example, using the APTT assay. These polypeptides are useful as therapeutic agents, particularly in patients afflicted with hemophilia B. These polypeptides may comprise further substitutions or modifications, such as the glycosylation sites described herein.
[056] One aspect of the application provides modified Factor IX polypeptides comprising the following amino acid sequence:
YNSGKLEEFVQGNLERECMEEKCSFEEAREVFENTERTTEFWKQ YVDGDQCESNPC
LNGGSCKDDINSYECWCPFGFEGKNCELX85X86TCNIKNGRCEQFCKNSAX1O4NKW
CSCTEGYRLAENX121KSCEPAVPFPCGRVSVXI38QTSKLTRAEX148VXI5OXI5IXI52XI53D
YVNSX159EZ1X161Z2EZ3TZ4ILDNIX169QSX172QXI74FNX177X178TRWGGEDAKPGQFPW
QWLNGKVDAFCGGSIVNEKWIVTAAHCVETX226VX228ITWAGEHNIEETEHTEQK
RNVIRIIPHHNYNAAINKYNHDIALLELDEPLVLNSYVTPICIADKEYTNIFLKFGSGY
VSGWGRVFHKGRSALVLQYLRVPLVDRATCLX338STKFTIYNNMFCAGX353X354X355
GGRDSCQGDSGGPHX37OX371X372VEGTSFLTGIISWGEECAX391X392X393KYGIYTKVS
RYVNWIKE KTX413X414T (SEQ ID NO: 3);
wherein X85 is selected from D and E; wherein X86 is selected from A, E, P, S, and V; wherein X104 is selected from D, N, and T; wherein X121 is selected from N, Q, and T; wherein X138 is selected from N, S, and T; wherein X148 and X150 are selected from: (i) X148 is T and X150 is F; (ii) Xi48 is N and Xi50 is T; and (iii) Xi48 is N and Xi50 is S; wherein X151 is selected from A, P, and T; wherein X151 and Xi53 are selected from: (i) Xi5I is P and Xi53 is V; (ii) Xi5i is N and Xi53 is T; and (iii) Xi5i is N and Xi53 is S; wherein Zi, Z2, Z3, and Z4 are independently selected from (i) zero to twelve amino acid residues and (ii) SEQ ID NO: 2; wherein Xi52 is selected from D, N, and T; wherein Xi59 and Xi6I are selected from: (i) Xi59 is T and Xi6I is A; (ii) Xi59 is N and Xi6I is T; and (iii) Xi59 is N and Xi6i is S; wherein Xi69 is selected from T and N; wherein Xπ2 is selected from T and N; wherein Xi74 is selected from S and T; wherein Xi77 and Xi78 are selected from: (i) Xi77 is D and Xi78 is F; (ii) Xi77 is E and Xi78 is T; and (iii) Xi77 is E or D and Xi78 is S; wherein X226 and X228 are selected from: (i) X2215 is G and X228 is K; (ii) X226 is N and X228 is T; and (iii) X226 is N and X228 is S; wherein X338 is selected from R and A; wherein X353, X354, and X355 are selected from: (i) X353 is F, X354 is H, X355 is E; (ii) X353 is N, X354 is V, X355 is T; (iii) X353 is N, X354 is I, X355 is T; and (iv) X353 is N, X354 is H, V, or I, X355 is S; wherein X370, X37I, and X372 are selected from: (i) X370 is V, X37i is T, X372 is E; (ii) X370 is N, X37i is V, X372 is T; (iii) X370 is N, X371 is I, X372 is T; and (iv) X370 is N, X37i is T, V, or I, X372 is S; wherein X39I, X392, and X393 are selected from: (i) X39I is M, X392 is K, X393 is G; (ii) X39i is N, X392 is K, X393 is T; (iii) X39i is N, X392 is V, X393 is T; and (iv) X391 is N, X392 is V or K, X393 is S; wherein X4I3 and X414 are selected from: (i) X4I3 is K and X4I4 is L; (ii) X4I3 is N and X4I4 is L; and (iii) X4n is N and X4I4 is I; and wherein the FIX polypeptide comprises at least one introduced glycosylation site as compared to the FIX polypeptide having SEQ ID NO: 1.
[057] The introduction of at least one glycosylation site is the result of a substitution at least one of the X positions or an insertion in at least one of the Z positions. In some embodiments, the modified polypeptide additionally comprises between about 1-30, 1-20, or 1-10 conservative amino acid changes and maintains FIX activity. In some embodiments, the modified polypeptide is at least about 80, 85, 90, 95, or 99% identical to SEQ ID NO: 1 and maintains FIX activity.
Production of Modified FIX Polypeptides
[058] Amino acid residues may be inserted, deleted, or substituted in order to introduce a non- native glycosylation site. For example, glycosylation sites may be introduced by altering the amino acid sequence of FIX. Amino acid sequence alteration may be accomplished by a variety of techniques, such as, for example, by modifying the corresponding nucleic acid sequence by site- specific mutagenesis. Techniques for site-specific mutagenesis are well known in the art and are described in, for example, Zoller et al., (DNA 3:479-488, 1984) or Horton, et al., (Gene 77:61-68, 1989, pp. 61-68). Thus, using the nucleotide and amino acid sequences of FIX, one may introduce the alteration(s) of choice. Likewise, procedures for preparing a DNA construct using polymerase chain reaction using specific primers are well known to persons skilled in the art (see, e.g., PCR Protocols, 1990, Academic Press, San Diego, California, USA).
[059] The nucleic acid construct encoding the FIX polypeptide may also be prepared synthetically by established standard methods, for example, the phosphoramidite method described by Beaucage, et al., (Gene Amplif. Anal. 3:1-26, 1983). According to the phosphoamidite method, oligonucleotides are synthesized, for example, in an automatic DNA synthesizer, purified, annealed, ligated, and cloned in suitable vectors. The DNA sequences encoding the human FIX polypeptides may also be prepared by polymerase chain reaction using specific primers, for example, as described in US Patent No. 4,683,202; or Saiki, et al., (Science 239:487-491, 1988). Furthermore, the nucleic acid construct may be of mixed synthetic and genomic, mixed synthetic and cDNA, or mixed genomic and cDNA origin prepared by ligating fragments of synthetic, genomic, or cDNA origin (as appropriate), corresponding to various parts of the entire nucleic acid construct, in accordance with standard techniques.
[060] The DNA sequences encoding the FIX polypeptides may be inserted into a recombinant vector using recombinant DNA procedures. The choice of vector will often depend on the host cell into which the vector is to be introduced. The vector may be an autonomously replicating vector or an integrating vector. An autonomously replicating vector exists as an extrachromosomal entity and its replication is independent of chromosomal replication, for example, a plasmid. An integrating vector is a vector that integrates into the host cell genome and replicates together with the chromosome(s) into which it has been integrated.
[061] The vector may be an expression vector in which the DNA sequence encoding the modified FIX is operably linked to additional segments required for transcription, translation, or processing of the DNA, such as promoters, terminators, and polyadenylation sites. In general, the expression vector may be derived from plasmid or viral DNA, or may contain elements of both. The term "operably linked" indicates that the segments are arranged so that they function in concert for their intended purposes, for example, transcription initiates in a promoter and proceeds through the DNA sequence coding for the polypeptide.
[062] Expression vectors for use in expressing FIX polypeptides may comprise a promoter capable of directing the transcription of a cloned gene or cDNA. The promoter may be any DNA sequence that shows transcriptional activity in the host cell of choice and may be derived from genes encoding proteins either homologous or heterologous to the host cell.
[063] Examples of suitable promoters for directing the transcription of the DNA encoding the FIX polypeptides in mammalian cells are, for example, the SV40 promoter (Subramani, et al., MoI. Cell Biol. 1 :854-864, 1981), the MT-I (metallothionein gene) promoter (Palmiter, et al., Science 222:809-814, 1983), the CMV promoter (Boshart, et al., Cell 41 :521-530, 1985), or the adenovirus 2 major late promoter (Kaufman et al.,, MoI. Cell Biol, 2:1304-1319, 1982).
[064] The DNA sequences encoding the FIX polypeptide may also, if necessary, be operably connected to a suitable terminator, such as the human growth hormone terminator (Palmiter, et al.,
Science 222:809-814, 1983) or TPIl (Alber et al., J. MoI. Appl. Gen. 1 :419-434, 1982) or ADH3 (McKnight, et al., EMBO J. 4:2093-2099, 1985) terminators. The expression vectors may also contain a polyadenylation signal located downstream of the insertion site. Polyadenylation signals include the early or late polyadenylation signal from SV40, the polyadenylation signal from the adenovirus 5 EIb region, the human growth hormone gene terminator (DeNoto, et al., Nucl. Acids Res. 9:3719-3730, 1981), or the polyadenylation signal from the human FIX gene. The expression vectors may also include enhancer sequences, such as the SV40 enhancer.
[065] To direct the FIX polypeptides of the present invention into the secretory pathway of the host cells, the native FIX secretory signal sequence may be used. Alternatively, a secretory signal sequence (also known as a leader sequence, prepro sequence, or pre sequence) may be provided in the recombinant vector. The secretory signal sequence may be joined to the DNA sequences encoding the FIX analogues in the correct reading frame. Secretory signal sequences are commonly positioned 5' to the DNA sequence encoding the peptide. Exemplary signal sequences include, for example, the MPIF-I signal sequence and the stanniocalcin signal sequence.
[066] The procedures used to ligate the DNA sequences coding for the FIX polypeptides, the promoter, and optionally the terminator and/or secretory signal sequence and to insert them into suitable vectors containing the information necessary for replication, are well known to persons skilled in the art (see, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, New York, 1989).
[067] Methods of transfecting mammalian cells and expressing DNA sequences introduced into the cells are described in, for example, Kaufman, et al., (J. MoI. Biol. 159:601-621, 1982); Southern, et al., (J. MoI. Appl. Genet. 1 :327-341, 1982); Loyter, et al., (Proc. Natl. Acad. Sci. USA 79:422-426, 1982); Wigler, et al., (Cell 14:725-731, 1978); Corsaro, et al., (Somatic Cell Genetics 7:603-616, 1981), Graham, et al., (Virology 52:456-467, 1973); and Neumann, et al., (EMBO J. 1 :841-845, 1982). Cloned DNA sequences may be introduced into cultured mammalian cells by, for example, lipofection, DEAE-dextran-mediated transfection, microinj ection, protoplast fusion, calcium phosphate precipitation, retroviral delivery, electroporation, sonoporation, laser irradiation, magnetofection, natural transformation, and biolistic transformation (see, e.g., Mehier- Humbert, et al., Adv. Drug Deliv. Rev. 57:733-753, 2005). To identify and select cells that express the exogenous DNA, a gene that confers a selectable phenotype (a selectable marker) is generally introduced into cells along with the gene or cDNA of interest. Selectable markers include, for example, genes that confer resistance to drugs such as neomycin, puromycin, hygromycin, and methotrexate. The selectable marker may be an amplifiable selectable marker, which permits the amplification of the marker and the exogenous DNA when the sequences are linked. Exemplary amplifiable selectable markers include dihydrofolate reductase (DHFR) and adenosine deaminase. It is within the purview of one skilled in the art to choose suitable selectable markers (see, e.g., US Patent No. 5,238,820).
[068] After cells have been transfected with DNA, they are grown in an appropriate growth medium to express the gene of interest. As used herein the term "appropriate growth medium" means a medium containing nutrients and other components required for the growth of cells and the expression of the active FIX polypeptides.
[069] Media generally include, for example, a carbon source, a nitrogen source, essential amino acids, essential sugars, vitamins, salts, phospholipids, protein, and growth factors, and in the case of vitamin K dependent proteins such as FIX, vitamin K may also be provided. Drug selection is then applied to select for the growth of cells that are expressing the selectable marker in a stable fashion. For cells that have been transfected with an amplifiable selectable marker the drug concentration may be increased to select for an increased copy number of the cloned sequences, thereby increasing expression levels. Clones of stably transfected cells are then screened for expression of the FIX polypeptide.
[070] Examples of mammalian cell lines for use in the present invention are the COS-I (ATCC CRL 1650), baby hamster kidney (BHK), HKBl 1 cells (Cho, et al., J. Biomed. Sci, 9:631-638, 2002), and HEK-293 (ATCC CRL 1573; Graham, et al., J. Gen. Virol. 36:59-72, 1977) cell lines. In addition, a number of other cell lines may be used within the present invention, including rat Hep I (rat hepatoma; ATCC CRL 1600), rat Hep II (rat hepatoma; ATCC CRL 1548), TCMK-I (ATCC CCL 139), Hep-G2 (ATCC HB 8065), NCTC 1469 (ATCC CCL 9.1), CHO-Kl (ATCC CCL 61), and CHO-DUKX cells (Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77:4216-4220, 1980).
[071] FIX polypeptides may be recovered from cell culture medium and may then be purified by a variety of procedures known in the art including, but not limited to, chromatography (e.g., ion exchange, affinity, hydrophobic, chromatofocusing, and size exclusion), electrophoretic procedures (e.g., preparative isoelectric focusing (IEF), differential solubility (e.g., ammonium sulfate precipitation)), extraction (see, e.g., Protein Purification, Janson and Lars Ryden, editors, VCH Publishers, New York, 1989), or various combinations thereof. In an exemplary embodiment, the polypeptides may be purified by affinity chromatography on an anti-FIX antibody column. Additional purification may be achieved by conventional chemical purification means, such as high performance liquid chromatography. Other methods of purification are known in the art, and may be applied to the purification of the modified FIX polypeptides (see, e.g., Scopes, R., Protein Purification, Springer- Verlag, N. Y., 1982).
[072] Generally, "purified" shall refer to a protein or peptide composition that has been subjected to fractionation to remove various other components, and which substantially retains its expressed biological activity. Where the term "substantially purified" is used, this designation shall refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 99%, or more of the proteins in the composition.
[073] Various methods for quantifying the degree of purification of the polypeptide are known to those of skill in the art. These include, for example, determining the specific activity of an active fraction, or assessing the amount of polypeptides within a fraction by SDS/PAGE analysis. An exemplary method for assessing the purity of a fraction is to calculate the specific activity of the fraction, compare the activity to the specific activity of the initial extract, and to thus calculate the degree of purity, herein assessed by a "-fold purification number." The actual units used to represent the amount of activity will, of course, be dependent upon the particular assay technique.
[074] In some embodiments, FIX polypeptides are recombinantly expressed in tissue culture cells and glycosylation is the result of the normal post-translational cell functioning of the host cell, such as a mammalian cell.
[075] Alternatively, glycosylation may be achieved through chemical or enzymatic modification. FIX polypeptides may be glycosylated by using, for example, an enzyme that adds alpha-(2,6)- linked sialic acid to protein. For example, dihydrofolate reductase (DHFR) deficient CHO cells are commonly used host cells for recombinant glycoprotein production. CHO cells do not endogenously express the enzyme beta-galactoside alpha-2,6 sialyltransferase, which is used to add sialic acid in the 2,6 linkage to galactose on the mannose alpha- 1,3 branch. To add sialic acid at this linkage to a protein produced in CHO cells, the CHO cells may be transfected with a functional beta-galactosidase alpha-2,6 sialyltransferase gene to allow for incorporation of sialic acid in the 2,6 linkage to galactose as desired (see, e.g., Lee, et al., J. Biol. Chem. 264:13848- 13855, 1989).
[076] Similarly, a bisecting N-acetylglucosamine (GIcNAc) may be added to FIX by transfecting a host cell that does not endogenously produce this oligosaccharide linkage with the functional gene for the enzyme N-acetylglucosaminyltransferase, which has been reported to catalyze formation of a bisecting GIcNAc structure. Systems have also been established to express proteins in plant cells that produce proteins with mammalian glycosylation patterns (see, e.g., bryophyte cells, WO 2004/057002).
[077] For N-linked glycosylation, the final structures of the N-glycans are typically dependent upon the organism in which the polyp eptide is produced. Generally, polypeptides produced in bacteria are completely unglycosylated. Polypeptides expressed in insect cells contain high mannose and pauci-mannose N-linked oligosaccharide chains, among others. Polypeptides produced in mammalian cell culture are usually glycosylated differently depending upon, for example, the species and cell culture conditions. Further, polypeptides produced in plant cells comprise glycan structures that differ significantly from those produced in animal cells. The goal in the art of the production of recombinant polypeptides, particularly when the polypeptides are to be used as therapeutic agents, is to be able to generate polypeptides that are correctly glycosylated, that is, to be able to generate a polypeptide having a glycan structure that resembles, or is identical to that present on the naturally occurring form of the polypeptide.
[078] A variety of methods have been proposed in the art to customize the glycosylation pattern of a polypeptide (see, e.g., WO 99/22764; WO 98/58964; WO 99/54342; US Publication No. 2008/0050772; and US Patent No. 5,047,335). Essentially, many of the enzymes required for the in vitro glycosylation of polypeptides have been cloned and sequenced. In some instances, these enzymes have been used in vitro to add specific sugars to an incomplete glycan molecule on a polypeptide. In other instances, cells have been genetically engineered to express a combination of enzymes and desired polypeptides such that addition of a desired sugar moiety to an expressed polypeptide occurs within the cell.
[079] For O-linked glycosylation, O-glycans are linked primarily to serine and threonine residues and are formed by the stepwise addition of sugars from nucleotide sugars (Tanner, et al., Biochim. Biophys. Acta. 906:81-99, 1987); Hounsell, et al., Glycoconj. J. 13:19-26, 1996). Polypeptide function can be affected by the structure of the O-linked glycans present. For a review of O-linked glycan structures, see, for example, Schachter and Brockhausen, The Biosynthesis of Branched O-Linked Glycans, 1989, Society for Experimental Biology, pp. 1-26 (Great Britain).
[080] Whether a FIX polypeptide has N-linked and/or O-linked glycosylation may be determined using standard techniques (see, e.g., Techniques in Glycobiology, R. Townsend and A. Hotchkiss, eds. (1997) Marcel Dekker; and Glycoanalysis Protocols (Methods in Molecular Biology, Vol. 76), E. Hounsell, ed. (1998) Humana Press). The change in electrophoretic mobility of a protein before and after treatment with chemical or enzymatic deglycosylation (e.g., using endoglycosidases and/or exoglycosidases) is routinely used to determine the glycosylation status of a protein. Enzymatic deglycosylation may be carried out using any of a variety of enzymes, including, but not limited to, peptide-N4-(N-acetyl-beta-D-glycosaminyl) asparagine amidase (PNGase F), endoglycosidase Fl, endoglycosidase F2, endoglycosidase F3, and the like. For example, sodium docecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analysis of the protein, either pre-treated with PNGaseF or untreated with PNGaseF, may be conducted. A marked decrease in band width and change in migration position after treatment with PNGaseF may be considered diagnostic of N-linked glycosylation. The carbohydrate content of a glycosylated protein also can be detected using lectin analysis of protein blots (e.g., proteins separated by SDS-PAGE and transferred to a support, such as a nylon membrane). Lectins, carbohydrate binding proteins from various plant tissues, have both high affinity and narrow specificity for a wide range of defined sugar epitopes found on glycoprotein glycans (Cummings, Methods Enzymol. 230:66-86, 1994). Lectins may be labeled (either directly or indirectly) allowing detection of binding of lectins to carbohydrates on glycosylated proteins. For example, when conjugated with biotin or digoxigenin, a lectin bound to a glycosylated protein can be easily identified on membrane blots through a reaction utilizing avidin or anti-digoxigenin antibodies conjugated with an enzyme such as alkaline phosphatase, beta-galactosidase, luciferase, or horse radish peroxidase, to yield a detectable product. Screening with a panel of lectins with well- defined specificity provides considerable information about a glycoprotein's carbohydrate complement. The electrophoretic mobility of the modified FIX polypeptide may also be compared to the mobility of a reference protein.
[081] The application provides, in part, FIX polypeptides with introduced glycosylation sites, wherein the carbohydrate chain attached to the glycosylation site may have a mammalian carbohydrate chain structure, that is, a mammalian glycosylation pattern. In some embodiments, the carbohydrate chain has a human glycosylation pattern. As used herein, a pattern of glycosylation refers to the representation of particular oligosaccharide structures within a given population of FIX polypeptides. Non-limiting examples of such patterns include the relative proportion of oligosaccharide chains that (i) have at least one sialic acid residue; (ii) lack any sialic acid residues (i.e., are neutral in charge); (iii) have at least one terminal galactose residue; (iv) have at least one terminal N-acetylgalactosamine residue; (v) have at least one "uncapped" antenna, that is, have at least one terminal galactose or N-acetylgalactosamine residue; or (vi) have at least one fucose linked alphal->3 to an antennary N-acetylglucosamine residue.
[082] The pattern of glycosylation may be determined using any method known in the art, including, without limitation: high-performance liquid chromatography (HPLC); capillary electrophoresis (CE); nuclear magnetic resonance (NMR); mass spectrometry (MS) using ionization techniques such as fast-atom bombardment, electrospray, or matrix-assisted laser desorption (MALDI); gas chromatography (GC); and treatment with exoglycosidases in conjunction with anion- exchange (AIE)-HPLC, size-exclusion chromatography (SEC), or MS (see, e.g., Weber, et al., Anal. Biochem. 225:135-142, 1995; Klausen, et al., J. Chromatog. 718:195-202, 1995; Morris, et al., in Mass Spectrometry of Biological Materials, McEwen et al., eds., Marcel Dekker, (1990), pp 137-167; Conboy et al., Biol. Mass Spectrom. 21 :397-407, 1992; Hellerqvist, Meth. Enzymol. 193:554-573, 1990; Sutton, et al., Anal. Biochem. 218:34-46, 1994; Harvey, et al., Organic Mass Spectrometry 29:753-766, 1994). Pharmaceutical Compositions
[083] Based on well known assays used to determine the efficacy for treatment of conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the polypeptides of this invention may readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular polypeptide and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
[084] The application provides, in part, compositions comprising FIX polypeptides with one or more introduced glycosylation sites as described herein. The compositions may be suitable for in vivo administration and are pyrogen free. The compositions may also comprise a pharmaceutically acceptable carrier. The phrase "pharmaceutically or pharmacologically acceptable" refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Supplementary active ingredients also may be incorporated into the compositions.
[085] The compositions of the present invention include classic pharmaceutical preparations. Administration of these compositions according to the present invention may be via any common route. The pharmaceutical compositions may be introduced into the subject by any conventional method, for example, by intravenous, intradermal, intramuscular, subcutaneous, intramammary, intraperitoneal, intrathecal, retrobulbar, intrapulmonary, oral, sublingual, nasal, anal, vaginal, or transdermal delivery, or by surgical implantation at a particular site. The treatment may consist of a single dose or a plurality of doses over a period of time.
[086] The active compounds may be prepared for administration as solutions of free base or pharmacologically acceptable salts in water, suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions also may be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
[087] The pharmaceutical forms, suitable for injectable use, include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. The form should be sterile and should be fluid to the extent that easy syringability exists. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like) sucrose, L-histidine, polysorbate 80, or suitable mixtures thereof, and vegetable oils. The proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion, and by the use of surfactants. The prevention of the action of microorganisms may be brought about by various antibacterial an antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. The injectable compositions may include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions may be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
[088] Sterile injectable solutions may be prepared by incorporating the active compounds (e.g., FIX polypeptides) in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
[089] Generally, dispersions may be prepared by incorporating the various sterilized active ingredients into a sterile vehicle that contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation include, for example, vacuum-drying and freeze-drying techniques that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[090] Upon formulation, solutions may be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. "Therapeutically effective amount" is used herein to refer to the amount of a polypeptide that is needed to provide a desired level of the polypeptide in the bloodstream or in the target tissue. The precise amount will depend upon numerous factors, for example, the particular FIX polypeptide, the components and physical characteristics of the therapeutic composition, intended patient population, mode of delivery, individual patient considerations, and the like, and can readily be determined by one skilled in the art, based upon the information provided herein.
[091] The formulations may be easily administered in a variety of dosage forms, such as injectable solutions, and the like. For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
[092] Dosages of FIX are normally expressed in units. One unit of FIX per kg of body weight may raise plasma levels by 0.01 U/ml, that is, 1%. Otherwise healthy patients have one unit of FIX per ml of plasma, that is, 100%. Mild cases of hemophilia B are defined by FIX plasma concentrations between 6-60%, moderate cases between 1-5%, and severe cases, which account for about half of the hemophilia B cases, have less than 1% FIX. Prophylactic treatment or treatment of minor hemorrhaging usually requires raising FIX levels to between 15-30%. Treatment of moderate hemorrhaging usually requires raising levels to between 30-50%, while treatment of major trauma may require raising levels from 50 to 100%. The total number of units needed to raise a patient's blood level can be determined as follows: 1.0 unit/kg x body weight (kg) x desired percentage increase (% of normal). Parenteral administration may be carried out with an initial bolus followed by continuous infusion to maintain therapeutic circulating levels of drug product. In some embodiments, between 15 to 150 units/kg of FIX polypeptide may be administered. Those of ordinary skill in the art will readily optimize effective dosages and administration regimens as determined by good medical practice and the clinical condition of the individual patient.
[093] The frequency of dosing will depend on the pharmacokinetic parameters of the agents and the routes of administration. The optimal pharmaceutical formulation may be determined by one of skill in the art depending on the route of administration and the desired dosage (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 20th edition, 2000, incorporated herein by reference). Such formulations may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the administered agents. Depending on the route of administration, a suitable dose may be calculated according to body weight, body surface area, or organ size. Further refinement of the calculations necessary to determine the appropriate treatment dose is routinely made by those of ordinary skill in the art without undue experimentation, especially in light of the dosage information and assays disclosed herein, as well as the pharmacokinetic data observed in animals or human clinical trials. Exemplary dosing schedules include, without limitation, administration five times a day, four times a day, three times a day, twice daily, once daily, three times weekly, twice weekly, once weekly, twice monthly, once monthly, and any combination thereof.
[094] Appropriate dosages may be ascertained through the use of established assays for determining blood clotting levels in conjunction with relevant dose response data. The final dosage regimen may be determined by the attending physician, considering factors that modify the action of drugs, for example, the drug's specific activity, severity of the damage, and the responsiveness of the patient, the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration, and other clinical factors.
[095] The composition may also include an antimicrobial agent for preventing or deterring microbial growth. Non-limiting examples of antimicrobial agents suitable for the present invention include benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate, thimersol, and combinations thereof.
[096] An antioxidant may be present in the composition as well. Antioxidants may be used to prevent oxidation, thereby preventing the deterioration of the preparation. Suitable antioxidants for use in the present invention include, for example, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothioglycerol, propyl gallate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, and combinations thereof.
[097] A surfactant may be present as an excipient. Exemplary surfactants include: polysorbates such as Tween®-20 (polyoxyethylenesorbitan monolaurate) and Tween®-80 (polyoxyethylenesorbitan monooleate) and pluronics such as F68 and F88 (both of which are available from BASF, Mount Olive, NJ.); sorbitan esters; lipids such as phospholipids such as lecithin and other phosphatidylcholines, phosphatidyl ethanolamines, fatty acids and fatty esters; steroids such as cholesterol; and chelating agents such as EDTA, zinc and other such suitable cations.
[098] Acids or bases may be present as an excipient in the composition. Non-limiting examples of acids that may be used include hydrochloric acid, acetic acid, phosphoric acid, citric acid, malic acid, lactic acid, formic acid, trichloroacetic acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, fumaric acid, and combinations thereof. Examples of suitable bases include, without limitation, sodium hydroxide, sodium acetate, ammonium hydroxide, potassium hydroxide, ammonium acetate, potassium acetate, sodium phosphate, potassium phosphate, sodium citrate, sodium formate, sodium sulfate, potassium sulfate, potassium fumerate, and combinations thereof.
[099] The amount of any individual excipient in the composition may vary depending on the activity of the excipient and particular needs of the composition. Typically, the optimal amount of any individual excipient may be determined through routine experimentation, that is, by preparing compositions containing varying amounts of the excipient (ranging from low to high), examining the stability and other parameters, and then determining the range at which optimal performance is attained with no significant adverse effects. Generally, the excipient may be present in the composition in an amount of about 1% to about 99% by weight, from about 5% to about 98% by weight, from about 15 to about 95% by weight of the excipient, with concentrations less than 30% by weight. These foregoing pharmaceutical excipients along with other excipients are described in "Remington: The Science & Practice of Pharmacy," 19 ed., Williams & Williams, (1995); the "Physician's Desk Reference," 52 ed., Medical Economics, Montvale, NJ. (1998); and Kibbe, A. H., Handbook of Pharmaceutical Excipients, 3 Edition, American Pharmaceutical Association, Washington, D.C., 2000. Exemplary Uses
[100] The compositions described herein may be used to treat any bleeding disorder associated with functional defects of FIX or deficiencies of FIX such as a shortened in vivo half-life of FIX, altered binding properties of FIX, genetic defects of FIX, and a reduced plasma concentration of FIX. Genetic defects of FIX comprise, for example, deletions, additions, and/or substitution of bases in the nucleotide sequence encoding FIX. In one embodiment, the bleeding disorder may be hemophilia B. Symptoms of such bleeding disorders include, for example, severe epistaxis, oral mucosal bleeding, hemarthrosis, hematoma, persistent hematuria, gastrointestinal bleeding, retroperitoneal bleeding, tongue/retropharyngeal bleeding, intracranial bleeding, and trauma- associated bleeding.
[101] The compositions of the present invention maybe used for prophylactic applications. In some embodiments, modified FIX polypeptides may be administered to a subject susceptible to or otherwise at risk of a disease state or injury to enhance the subject's own coagulative capability. Such an amount may be defined to be a "prophylactically effective dose." Administration of the modified FIX polypeptides for prophylaxis includes situations where a patient suffering from hemophilia B is about to undergo surgery and the polypeptide is administered between one to four hours prior to surgery. In addition, the polypeptides are suited for use as a prophylactic against uncontrolled bleeding, optionally in patients not suffering from hemophilia. Thus, for example, the polypeptide may be administered to a patient at risk for uncontrolled bleeding prior to surgery.
[102] The polypeptides, materials, compositions, and methods described herein are intended to be representative examples of the invention, and it will be understood that the scope of the invention is not limited by the scope of the examples. Those skilled in the art will recognize that the invention may be practiced with variations on the disclosed polypeptides, materials, compositions and methods, and such variations are regarded as within the ambit of the invention.
[103] The following examples are presented to illustrate the invention described herein, but should not be construed as limiting the scope of the invention in any way.
EXAMPLES
[104] In order that this invention may be better understood, the following examples are set forth. These examples are for the purpose of illustration only, and are not to be construed as limiting the scope of the invention in any manner. All publications mentioned herein are incorporated by reference in their entirety.
Example 1: In Silico Analysis
[105] An in silico analysis of the FIX sequence for solvent accessibility, known secondary structure, location of known hemophilia B mutations, proximity to predicted sites of interaction with both FVIII and FX, and predicted effect of a given mutation upon FIX protein stability was performed in order to identify potential sites for the addition of new N-glycosylation site consensus sequences.
[106] Based upon this analysis, five sites were identified within the catalytic domain for creation of new N-linked glycosylation sites. The selected sites and the altered amino acid sequences that were designed are shown in Table 2.
TABLE 2
Figure imgf000037_0001
Example 2: Sequence Alignment of Activation Peptide
[107] Conserved and non-conserved residues in the activation peptide were identified by a multiple species sequence alignment as shown in Figure 1.
[108] The consensus sequence of N-linked glycosylation sites is Asn-X-Ser/Thr (where X is any amino acid except proline or perhaps aspartic acid which is also not favorable). To design new N- linked glycosylation sites in the activation peptide of FIX, the amino acids within the activation peptide that are conserved between species was avoided as well as and consensus sequences for other N-linked sites and the consensus sequence for tyrosine sulfation so as not to disrupt these posttranslational modifications since these could be important for the pharmacokinetics of FIX. Using these criteria, three sites for adding new N-linked glycosylation sites within the activation peptide were identified as shown in Table 3.
TABLE 3
Figure imgf000038_0001
Example 3: Insertion of Amino Acids to Generate N-linked Glycosylation Sites
[109] The multiple sequence alignment (Figure 1) also revealed that mouse, rat, and guinea pig have additional amino acids between residues Al 61 and E 162 relative to human, rhesus monkey, pig, dog, and rabbit FIX. These additional amino acids vary in size from 7 to 10 between the three species and contain an over representation of Asp and to some extent He residues. This observation demonstrates that between 7 and 10 additional residues are tolerated at this site in rat, mouse, and guinea pig without significant effects on FIX activity. Depending upon the criteria used to perform the multiple sequence alignment between FIX from the eight species, the apparent site at which the additional amino acids in rat, mouse, and guinea pig are found can vary such that the site can be either between E160 and A161, between A161 and E162, between E162 and T163, or between T 163 and 1164 of human FIX.
[110] Nine extra amino acids with the sequence N-S-T-Q-D-N-I-T-Q (SEQ ID NO: 2) were inserted in the activation peptide between Al 61 and El 62. The sequences " N-S-T" and "N-I-T" are the N-linked consensus sequences from the natural FIX activation peptide. In both cases, these are followed by a glutamine (Q) and preceded by an aspartic acid (D) in the natural FIX sequence. Therefore, a glutamine as well as an aspartic acid were included in an attempt to emulate the natural site. It is envisioned that additional sequences containing between 1 and 3 consensus sequences for N-glycosylation site (N-X-S/T) may also be inserted between A161 and E162.
[111] Additional examples of modified FIX polypeptides are described in Tables 4a, 4b, and 4c. TABLE 4a
Figure imgf000039_0001
Specific activity was calculated by dividing the activity measured in the cell culture supernatant by the FIX antigen concentration in the same supernatant and is expressed as a percentage of the control FIX molecule lacking the new glycosylation sites. NT: not tested. TABLE 4b
Figure imgf000040_0001
TABLE 4c
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Expression and activity relative to the wild type control are expressed on a scale of 1 to 5 where 1 = <10%; 2 = 10-50%; 3 = 50-100%; 4 = 100-200%; 5 = >200%.
Example 4: Substitutions to Convert O-linked to N-linked Glycosylation Sites
[112] Table 5 shows the substitutions that were designed to convert O-linked glycosylation sites to N-linked glycosylation sites.
TABLE 5
Figure imgf000048_0002
Example 5: Expression of FIX Variants in HKBIl Cells
[113] In order to determine if the FIX genes with altered protein sequences could be expressed and secreted from mammalian cells and to determine the effect of these substitutions upon FIX coagulation activity, expression plasmids encoding these FIX variants were transfected into HKBl 1 cells. HKBl 1 is a human cell line generated by the fusion of HEK293 cells and a B cell lymphoma. In this example, each of the glycosylation site substitutions were combined with a second substitution, R338A, as shown for HGl through HG8 in Figure 3. The R338A substitution increases the specific activity of FIX by 3- to 4-fold as measured by the aPTT assay. Additional combinations of two glycosylation site muteins and R338A were also created and tested (Figure 3).
[114] Three days after transfection, the media from the cells was collected and analyzed for FIX expression by Western blot using an antibody specific to FIX. The results demonstrated that the FIX muteins tested were expressed and secreted into the media at levels similar to that of wild type FIX or FIX with only the R338A substitution (Figure 2).
[115] The activity of the FIX muteins was also tested using the aPPT assay. The results demonstrated that the Factor IX muteins have activity similar to or increased compared to that of wild type FIX (Figures 3 and 4). Compared to R338A, the activity of the muteins varied between 14% and 109%. In particular, muteins HGl, HG3, HG4, HG5, HG6, HG7, HG8, and HG3 plus HG8 had coagulation activity that was at least 55% of that of R338A, and at least 300% of wild type FIX.
Example 6: Glycosylation of Factor IX variants in HKBIl cells
[116] An initial analysis of muteins HGl through HG8 (Figure 3) and HG9 (Figure 5) demonstrated that HG2, HG3, HG5, HG6, HG8, and HG9 had increased glycosylation as evidenced by an increase in apparent molecular weight on SDS-PAGE gels. Additional FIX polypeptides were created containing various combinations of the substitutions present in HG2, HG3, HG5, HG6, and HG8. For example, the following combinations were created: HG2/HG3, HG8/HG2, HG8/HG3, HG8/HG2/HG3. Other possible combinations include, for example, HG3/HG5, HG5/HG8, HG3/HG5/HG8. HG3/HG6, HG5/HG6, HG8/HG6, HG3/HG5/HG6, HG3/HG6/HG8, HG5/HG6/HG8, and HG3/HG5/HG6/HG8. Combinations with HG9 may also be generated.
[117] Glycosylation increases the molecular weight of a protein and this can be visualized as reduced mobility in SDS-PAGE gels. As demonstrated in Figures 2 and 3, the substitutions made in HG2, HG3, HG5, HG6, HG8, and HG9 resulted in reduced mobility on a SDS-PAGE gel. Among these eight single muteins, HG8 which has two additional N-linked consensus sequences inserted between A161 and E162, exhibited the greatest increase in apparent molecular weight suggesting that both N-linked sites were functional. Of the four muteins in which an O-linked site was mutated to an N-linked site (HG4 to HG7), HG4 exhibited increased mobility on the gel demonstrating that this substitution destroyed the O-linked site (thus reducing the molecular weight of the FIX protein), but failed to create a functional N-linked site. In contrast, HG5 and HG6 exhibited reduced mobility on the gel demonstrating that the substitutions in these clones did create functional N-linked glycosylation sites. Mutein HG7 which also has an O-linked site mutated to an N-linked site exhibited no change in mobility on the gel compared to wild-type FIX or FIX-R338A. Given the fact that the substitution in HG7 would be expected to eliminate an O- linked site and thus increase the mobility on the gel, the finding that there was no change in mobility suggests that the introduced N-linked site may be functional.
[118] The combinations of substitutions present in HG2/HG3, HG8/HG2, HG8/HG3, and HG8/HG2/HG3 resulted in more significant reductions in mobility compared to the individual muteins, indicating that the various new glycosylation sites present in these combinations were functional when combined in a single FIX molecule. The HG8/HG2/HG3 mutein that contains a total of 4 new N-linked sites exhibited the largest decrease in mobility.
Example 7: Combination ofR338A and V86A Substitutions
[119] Amino acid V86 of FIX was changed to alanine by site directed mutagenesis either in the context of wild type FIX (WT-FIX) or FIX-R338A. Expression vectors containing these constructs were transfected into HKBl 1 cells and media was collected 3 days later and assayed for FIX protein level by ELISA and for FIX coagulation activity by aPTT assay. Both muteins were expressed at similar levels to WT-FIX and FIX-R338A. The data from a single experiment is summarized in Table 6a. Table 6b summarizes the average of three experiments.
TABLE 6a
Figure imgf000050_0001
TABLE 6b
Figure imgf000051_0001
[120] The results demonstrate that the V86A substitution alone results in about a 1.8-fold increase in specific activity, while R338A alone resulted in a 4.5-fold increase in specific activity. The combination of R338A and V86A resulted in a 8.1-fold increase in specific activity as compared to wild type FIX. These results show that the positive effects of the R338A and V86A substitutions are additive and result in a Factor IX mutein with an 8-fold increased specific activity compared to WT-FIX. The R338A-V86A mutein would have improved therapeutic benefit for hemophilia B patients as it would allow a 8-fold lower dose of protein to achieve the same therapeutic effect as the currently available recombinant WT-IX. In addition, the increased specific activity of R338A-V86A is beneficial when creating glycosylated forms of FIX in which a reduction in activity may result from glycosylation.
Example 8: Cloning of Human FIXcDNA
[121] A pair of PCR primers complementary to sequences at the 5' and 3' ends of the coding region of the human FIX cDNA were designed from the published cDNA sequence (NM_000133). The 5' primer (FIXFl ; ATCATAAGCTTGCCACCATGCAGCGCGTGAACATG (SEQ ID NO: 8), start codon of FIX is in bold text) contained the first 18 nucleotides of the FIX coding region including the ATG start codon preceded by a consensus Kozak sequence (underlined) and a HindIII restriction site. The 3' primer (FIXR3, ATCATAAGCTTGATTAGTTAGTGAGA GGCCCTG) (SEQ ID NO: 9) contained 22 nucleotides of FIX sequence that lies 45 nucleotides 3' of the end of the FIX coding region preceded by a HindIII site. Amplification of first strand cDNA from normal human liver (Stratagene, San Diego CA) using these primers and high fidelity proofreading polymerase (Invitrogen, Carlsbad, CA) resulted in a single band of the expected size for human FIX cDNA (1464 bp). After digestion with HindIII, the PCR product was gel purified and then cloned into the HindIII site of the plasmid pAGE16. Clones in which the FIX cDNA was inserted in the forward orientation relative to the CMV promoter in the vector were identified by restriction digest. Double stranded DNA sequencing was performed for the insert of several clones and alignment of the derived sequence to the published FIX sequence demonstrated that the cDNA encodes human FIX with threonine at amino acid 148 of the mature protein. This plasmid was designated as pAGE16-Factor IX (pAGE16-FIX). Example 9: Generation of Modified FIX Polypeptides
[122] To change various amino acids within the human FIX sequence, a pair of primers were designed using the Quickchange™ primer design program available from Stratagene. These primers were used to generate mutations in the pAGE16-FIX plasmid employing the Quickchange™ II XL site directed mutagenesis kit (Stratagene, San Diego, CA) according to the manufacturer's instructions. Clones containing the desired substitution were identified by DNA sequencing of the entire FIX coding region. Table 7 below shows the sequence of the sense strand oligonucleotide used to create the substitutions.
TABLE 7
Figure imgf000052_0001
Figure imgf000053_0001
The underlined residues are those that were changed relative to wild type FIX to create a consensus sequence for N-linked glycosylation.
[123] In addition to the substitutions described above, a sequence of nine amino acids containing 2 consensus sequences for N-glycosylation was inserted into the activation peptide between residues A161 and E162. To generate this variant of FIX, unique restriction sites for SnaBl and Xbal at Yl 55 and 1164 were created without altering the amino acid sequence by site directed mutagenesis with primers t8216c_g8218a and t8188c (Table 8). The resulting plasmid was digested with SnaBl and Xbal to remove the 27 bp fragment corresponding to residues Vl 56 to 1164 and then ligated to a double stranded fragment created by annealing of oligonucleotides 2PrimerF and 2PrimerR (Table 8). The sequence of the resulting plasmid was determined by double strand DNA sequencing to have an insertion of 27 bp encoding nine amino acids with the sequence NSTQDNITQ (SEQ ID NO: 2) that contains two consensus sequences for N-linked glycosylation (NXT).
TABLE 8
Figure imgf000053_0002
The bold/underline residues are those that were changed relative to wild type Factor IX to create either a new restriction site or the insertion of nine additional amino acids encoding two consensus sequences for N-linker glycosylation.
Example 10: Cell Culture and Transient Transfection
[124] HKB 11 cells (a hybrid of HEK293 and a Burkitt B cell lymphoma line, 2B8) were grown in suspension culture on an orbital shaker (100-125 rpm) in a CO2 (5%) incubator at 370C in serum free media (RF#277) supplemented with 10 ng/mL soluble vitamin K3 (Sigma- Aldrich, St. Louis, MO) and maintained at a density between 0.25 and 1.5 x 106 cells/mL.
[125] Cells for transfection were collected by centrifugation at 1000 rpm for 5 minutes then resuspended in FreeStyle™ 293 Expression Medium (Invitrogen, Carlsbad, CA) at 1.1 x 106 cells/mL. The cells were seeded in 6 well plates (4.6 mL/well) and incubated on an orbital rotator (125 rpm) in a 370C CO2 incubator. For each well, 5 μg plasmid DNA was mixed with 0.2 mL Opti-MEM® I medium (Invitrogen). For each well, 7 μL 293fectin™ reagent (Invitrogen) was mixed gently with 0.2 mL Opti-MEM® I medium and incubated at room temperature for 5 minutes. The diluted 293fectin™ was added to the diluted DNA solution, mixed gently, incubated at room temperature for 20-30 minutes and then added to each well that had been seeded with 5 x 106 (4.6 mL) HKBl 1 cells. The cells were then incubated on an orbital rotator (125 rpm) in a CO2 incubator at 37°C for 3 days after which the cells were pelleted by centrifugation at 1000 rpm for 5 minutes, and the supernatant was collected and stored at 40C.
[126] In some instances, HEK293 cells were transfected with expression constructs for FIX variants in 384 well plates using standard lipofection protocols and commercial reagents. The cells were cultivated for 72 hours post-transfection at which time the supernatant was harvested for further analyses.
Example 11: Western Blot for FIX.
[127] Cell culture supernatant (50 μL) was mixed with 20 μL 4x SDS-PAGE loading dye, heated at 950C for 5 minutes, loaded on NuP AGE® 4-12% SDS PAGE gels and then transferred to nitrocellulose membranes. After blocking with 5% milk powder for 30 minutes, the membranes were incubated with a HRP-labeled goat polyclonal antibody against human FIX (US Biological, Swarapscott, Massachusetts, Catalog No. F0017-07B) for 60 minutes at room temperature. After washing with phosphate-buffered saline with 0.1% Tween®-20 buffer, the signal from HRP was detected using SuperSignal® Pico (Pierce, Rockford, IL) and exposure to x-ray film.
Example 12: FIXELISA
[128] FIX antigen levels in cell culture supernatants were determined using a FIX ELISA kit (Hyphen Biomed/Aniara, Mason, OH). Cell culture supernatant was diluted in sample diluent buffer (supplied in the kit) to achieve a signal within the range of the standard curve. FIX protein purified from human plasma (Hyphen Biomed/Aniara, Catalog No. RK032A, specific activity 196 U/mg) diluted in sample diluent was used as to create a standard curve from 100 ng/mL to 0.2 ng/mL. Diluted samples and the standards were added to the ELISA plate that is pre-coated with a polyclonal anti-FIX capture antibody. After adding the polyclonal detection antibody, the plate was incubated at room temperature for 1 hour, washed extensively, then developed using TMB substrate (3,3',5,5'-tetramethylbenzidine) as described by the kit manufacturer and the signal is measured at 450 nM using a SpectraMax® plate reader (Molecular Devices, Sunnyvale, CA). The standard curve was fitted to a 2-component plot and the values of the unknowns extrapolated from the curve. [129] FIX expression levels were also quantitated using commercially available FIX ELISA reagents (Haemochrom Diagnostica GmbH, Essen, Germany) according to the manufacturer's instructions. Wheat germ agglutinin (Sigma- Aldrich, St. Louis, MO) was coated on 384 well MaxiSorp™ plates (Nunc™, Rochester, NY). The wells were blocked, washed, and then supernatant was added. After further washing, detection was carried out using HRP-coupled polyclonal anti-FIX antibody (Haemochrom Diagnostica GmbH, Essen, Germany).
Example 13: FIX Coagulation Assay
[130] FIX coagulation activity was determined using an aPTT assay in FIX deficient human plasma run on a Electra™ 1800C automatic coagulation analyzer (Beckman Coulter, Fullerton, CA). Briefly, three dilutions of supernatant samples in coagulation diluent were created by the instrument, and 100 μL was then mixed with 100 μL FIX deficient plasma (Aniara, Mason, OH) and 100 μL automated aPTT reagent (rabbit brain phospholipid and micronized silica (bioMerieux, Inc., Durham, NC). After the addition of 100 μL 25 mM CaCl2 solution, the time to clot formation was recorded. A standard curve was generated for each run using serial dilutions of the same purified human FIX (Hyphen Biomed/Aniara) used as the standard in the ELISA assay. The standard curve was routinely a straight line with a correlation coefficient of 0.95 or better and was used to determine the FIX activity of the unknown samples.
Example 14: Measurement of Circulating FIX
[131] The circulating half-life of FIX polypeptides is measured using an in vitro assay. This assay is based on the ability of FIX in vivo and in vitro to mediate the accumulation of adenovirus (Ad) in hepatocytes. Briefly, it has been shown that FIX can bind the Ad fiber knob domain and provide a bridge for virus uptake through cell surface heparin sulfate proteoglycans (HSPG) (Shayakhmetov, et al., J. Virol 79:7478-7491, 2005). An Adenovirus vector mutant, Ad5mut, which contains mutations in the fiber knob domain, does not bind to FIX. Ad5mut has significantly reduced ability to infect liver cells and liver toxicity in vivo, demonstrating that FIX plays a major role in targeting Ad vectors to hepatic cells (Shayakhmetov, et al., 2005). The ability of FIX to target Ad vector to hepatic cells can be blocked by inhibitors of protein-HSPG interactions (Shayakhmetov, et al., 2005).
[132] Furthermore, HSPG-mediated uptake of FIX contributes significantly to FIX clearance and consequently, interfering with the HSPG interaction is expected to increase the half-life of FIX. Therefore, in vitro uptake of FIX and/or FIX variants in hepatocytes is measured, and variants with reduced uptake are expected to have increased half-life in vivo.
[133] To measure FIX half-life in vitro, mammalian cells are incubated with adenovirus in the presence or absence of FIX or FIX variants. Viral uptake is mediated by wild-type FIX and measured by expression of the reporter gene encoded in viral genome, for example, green fluorescent protein (GFP) or luciferase expression. Reduced uptake of adenovirus in the presence of FIX variants are measured as reduced reporter gene expression, for example, reduced GFP fluorescence or reduced luciferase enzymatic activity as compared to wild-type FIX.
[134] FIX circulating half-life is measured in vivo using standard techniques well-known to those of ordinary skill in the art. Briefly, the respective dose of FIX or FIX variant is administered to a subject by intravenous injection. Blood samples are taken at a number of time points after injection and the FIX concentration is determined by an appropriate assay (e.g.. ELISA). To determine the half-life, that is the time at which the concentration of FIX is half of the concentration of FIX immediately after dosing, the FIX concentration at the various time points is compared to the FIX concentration expected or measured immediately after administering the dose of FIX. A correlation between reduced cellular uptake in the in vitro assay and increased half-life in the in vivo assay is expected.
[135] All publications and patents mentioned in the above specification are incorporated herein by reference. Various modifications and variations of the described methods of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention.
[136] Although the invention has been described in connection with specific embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the above-described modes for carrying out the invention which are obvious to those skilled in the field of biochemistry or related fields are intended to be within the scope of the following claims. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

CLAIMS:
1. A Factor IX polypeptide comprising an amino acid sequence that has been modified by introducing one or more amino acid substitutions or insertions.
2. The polypeptide according to claim 1, wherein the amino acid sequence has been modified by introducing one or more glycosylation sites.
3. The polypeptide according to claim 2, wherein the one or more glycosylation sites are selected from N-linked glycosylation sites and O-linked glycosylation sites.
4. The polypeptide according to claims 2 and 3, further comprising a carbohydrate chain attached to the one or more introduced glycosylation sites.
5. The polypeptide according to any one of claims 1 to 4, wherein the one or more amino acid substitutions are selected from G4T; E33N; E36T; E36N; R37N; F75N; F77T; E83T; D85N; V86A; K91T; A103T; V107T; K122N; K122T; S138N; A146N; T148N; F150T; P151N; T159N; A161T; A161N; T169N; Q170N; T172N; D177N; D177E; F178T; K201N; K201T; K214T; V223N; G226N; Y226T; K228N; K228T; E239N; E242N; I251T; A262T; E294N; R338A; R338N; K341N; F353N; H354V; H354I; E355T; V370N; T371V; T371I; E372T; E374N; M391N; K392V; G393T; E410N; K413N; L414I; YlN and S3T; S3N and K5T; G4N and L6T; K5N and E7T; L6N and E8T; E7N and F9T; F9N and QI lT; VlON and G12T; Ql IN and N13T; G12N and L14T; N13 and E15T; L14N and R16T; E15N and E17T; M19N and E21T; E20N and K22T; S24N and E26T; F25N and E27T; E26N and A28T; E27N and R29T; A28N and E30T; R29N and V3 IT; E30N and F32T; V31N and E33T; F32N and N34T; T35N and R37T; T38N and E40T; T39N and F41T; E40N and W42T; F41N and K43T; W42N and Q44T; K43N and Y45T; Q44N and V46T; Y45N and D47T; V46N and G48T; E52N and N54T; S53N and P55T; G59N and S61T; K63N and D65T; I66N and S68T; S68N and E70T; G76N and E78T; E78N and K80T; E83N and D85T; L84N and V86T; I90N and N92T; KlOON and S102T; S102N and D104T; A103N and N105T; D104N and K106T; K106N and V108T; Rl 16N and Al 18T; E119N and Q121T; Q121N and S123T; A127N and P129T; V135N and V137T; S136N and S138T; V137N and Q139T; Q139N and S141T; T140N and K142T; S141N and L143T; E147N and V149T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; Y155N and N157T; V156N and S158T; S158N and E160T; T159N and A161T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; L165N and N167T; D166N and I168T; I168N and Q170T; T169N and S171T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; G184N and D186T; E185N and A187T; D186N and K188T; A187N and P189T; P189N and Q191T; G200N and V202T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; V227N and I229T; H236N and I238T; I238N and E240T; E240N and E242T; T241N and H243T; H243N and E245T; K247N and N249T; V250N and R252T; 125 IN and I253T; I253N and P255T; A261N and I263T; A262N and N264T; D276N and P278T; V280N and N282T; F302N and S304T; S304N and Y306T; R312N and F314T; V313N and H315T; F314N and K316T; H315N and G317T; K316N and R318T; G317N and S319T; S319N and L321T; A320N and V322T; R327N and P329T; P329N and V331T; D332N and A334T; L337N and S339T; S339N and K341T; T340N and F342T; T343N and Y345T; G352N and H354T; F353N and E355T; H354N and G356T; E355N and G357T; G356N and R358T; G357N and D359T; E372N and E374T; W385N and E387T; G386N and E388T; A390N and K392T; D85N, K122T, and 125 IT; D85N, K122T, and E242N; E125N, P126A, and A127T; P126N, V128T, and P129A; T148N, F150T, and P151A; F150N, P151A, and D152T; P151N, V153T, and A161N; P151N, V153T, and T172N; V153N, Y155T, and E294N; T172N, G226N, and K228T; F353N, H354V, and E355T; F353N, H354I, and E355T; V370N, T371V, and E372T; V370N, T371I, and E372T; M391N, K392V, and G393T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; T148N, F150T, G226N, and K228T; P151N, V153T, T172N, and R338A; P151N, V153T, D177E, and F178T; P151N, V153T, G226N, and K228T; T172N, G226N, K228T, and R338A; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; K122T, P151N, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; T148N, F150T, G226N, K228T, and R338A; P151N, V153T, T172N, G226N, and K228T; P151N, V153T, D177E, F178T, and R338A; P151N, V153T, G226N, K228T, and R338A; and P151N, V153T, T172N, G226N, K228T, and R338A; and any combination thereof.
6. The polypeptide according to any one of claims 1 to 4, wherein the one or more amino acid substitutions are selected from R37N; D85N; K122T; S138N; A146N; A161N; Q170N; T172N; D177N; F178T; K201N; K228N; E239N; E242N; I251T; A262T; E294N; E374N; E410N; G59N and S61T; K63N and D65T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; El 19N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; V153N and Y155T; D154N and V156T; V156N and S158T; S158N and E160T; E160N and E162T; E162N and I164T; T163N and L165T; I164N and D166T; D166N and I168T; I168N and Q170T; S171N and Q173T; T172N and S174T; Q173N and F175T; S174N and N176T; K201N and D203T; V202N and A204T; E224N and G226T; T225N and V227T; G226N and K228T; T241N and H243T; 125 IN and I253T; I253N and P255T; A262N and N264T; V280N and N282T; T343N and Y345T; E372N and E374T; D85N, K122T, and E242N; D85N, K122T, and I251T; F150N, P151A, and D152T; T172N, G226N, and K228T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; P151, V153T, G226N, and K228T; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; P151N, V153T, T172N, G226N, and K228T.
7. The polypeptide according to any one of claims 1 to 4, wherein the one or more amino acid substitutions are selected from D85N; K122T; S138N; T172N; K201N; K228N; E239N; E242N; I251T; A262T; E294N; G59N and S61T; G76N and E78T; S102N and D104T; A103N and N105T; D104N and K106T; El 19N and Q121T; Q121N and S123T; S136N and S138T; Q139N and S141T; T140N and K142T; T148N and F150T; V149N and P151T; P151N and V153T; D152N and D154T; S158N and E160T; E162N and I164T; T163N and L165T; T172N and S174T; Q173N and F175T; K201N and D203T; T225N and V227T; G226N and K228T; I253N and P255T; A262N and N264T; V280N and N282T; E372N and E374T; and D85N, K122T, and E242N; D85N, K122T, and I251T; F150N, P151A, and D152T; T172N, G226N, and K228T; D85N, P151N, V153T, and K228N; D85N, P151N, V153T, and E242N; K122T, P151N, V153T, and K228N; K122T, P151N, V153T, and E242N; K122T, P151N, V153T, and I251T; P151, V153T, G226N, and K228T; D85N, K122T, P151N, V153T, and E242N; D85N, P151N, V153T, G226N, and K228T; S138N, P151N, V153T, G226N, and K228T; P151N, V153T, T172N, G226N, and K228T.
8. A Factor IX polypeptide comprising the amino acid sequence
YNSGKLEEFVQGNLERECMEEKCSFEEAREVFENTERTTEFWKQYVDGDQCE
SNPCLNGGSCKDDINSYECWCPFGFEGKNCELX85X86TCNIKNGRCEQFCKNSA
XIO4NKWCSCTEGYRLAENXI2 IKSCEPAVPFPCGRVSVXI38QTSKLTRAEXI48V
XI5OXI5IXI52XI53DYVNSXI59EZ1X161Z2EZ3TZ4ILDNIX169QSXI72QXI74FNX177X178TR
WGGEDAKPGQFPWQWLNGKVDAFCGGSIVNEKWIVTAAHCVETX226VX228
ITVVAGEHNIEETEHTEQKRNVIRIIPHHNYNAAINKYNHDIALLELDEPLVLNS
YVTPICIADKEYTNIFLKFGSGYVSGWGRVFHKGRSALVLQYLRVPLVDRATC
LX338STKFTIYNNMFCAGX353X354X355GGRDSCQGDSGGPHX37OX371X372VEGTSF
LTGIISWGEECAX391X392X393KYGIYTKVSRYVNWIKE KTX413X414T
(SEQ ID NO: 3); wherein X85 is selected from D and E; wherein X86 is selected from A, E, P, S, and V; wherein X104 is selected from D, N, and T; wherein X12I is selected from N, Q, and T; wherein Xi38 is selected from N, S, and T; wherein X[48 and X150 are selected from: (i) Xi48 is T and Xi50 is F; (ii) Xi48 is N and Xi50 is T; and (iii) Xi48 is N and Xi50 is S; wherein X151 is selected from A, P, and T; wherein X151 and Xi53 are selected from: (i) Xi5i is P and Xi53 is V; (ii) Xi5i is N and Xi53 is T; and (iii) Xi5i is N and Xi53 is S; wherein Z1, Z2, Z3, and Z4 are independently selected from (i) zero to twelve amino acid residues and (ii) SEQ ID NO: 2; wherein X[52 is selected from D, N, and T; wherein Xi59 and X^1 are selected from: (i) Xi59 is T and Xi6i is A; (ii) Xi59 is N and Xi61 is T; and (iii) Xi59 is N and Xi6 I is S; wherein Xi69 is selected from T and N; wherein Xi72 is selected from T and N; wherein Xn4 is selected from S and T; wherein X177 and Xn8 are selected from: (i) X177 is D and Xi78 is F; (ii) X177 is E and X178 is T; and (iii) Xi77 is E or D and Xi78 is S; wherein X226 and X228 are selected from: (i) X226 is G and X228 is K; (ii) X226 is N and X228 is T; and (iii) X226 is N and X228 is S; wherein X338 is selected from R and A; wherein X353, X354, and X355 are selected from: (i) X353 is F, X354 is H, X355 is E; (ii) X353 is N, X354 is V, X355 is T; (iii) X353 is N, X354 is I, X355 is T; and (iv) X353 is N, X354 is H, V, or I, X355 is S; wherein X370, X371, and X372 are selected from: (i) X370 is V, X37i is T, X372 is E; (ii) X370 is N, X37i is V, X372 is T; (iii) X370 is N, X371 is I, X372 is T; and (iv) X370 is N, X37i is T, V, or I, X372 is S; wherein X39i, X392, and X393 are selected from: (i) X39i is M, X392 is K, X393 is G; (ii) X391 is N, X392 is K, X393 is T; (iii) X39i is N, X392 is V, X393 is T; and (iv) X391 is N, X392 is V or K, X393 is S; wherein X4I3 and X4I4 are selected from: (i) X4I3 is K and X4I4 is L; (ii) X4I3 is N and X4I4 is L; and (iii) X4I3 is N and X4I4 is I; and wherein the FIX polypeptide comprises at least one introduced glycosylation site as compared to the FIX polyp eptide having SEQ ID NO: 1.
9. The polypeptide according to claims 6 or 7, further comprising amino acid substitutions selected from R338A and V86A.
10. The polypeptide according to any one of claims 1 -9, wherein the amino acid sequence has been modified by introducing between 1 and 10 amino acid residues between amino acid residues 160-164 of human Factor IX resulting in the introduction of one or more glycosylation sites.
11. The polypeptide according to claim 10, wherein the amino acid residues are inserted between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163-164.
12. The polypeptide according to claim 11, wherein a polypeptide comprising the amino acid sequence of SEQ ID NO: 2 is introduced between amino acid residues 160-161, between amino acid residues 161-162, between amino acid residues 162-163, or between amino acid residues 163 - 164.
13. The polypeptide according to claim 11 or 12 further comprising amino acid substitutions selected from V86A; R338A; V86A and R338A; T148N and F150T; D177E and F178T; P151N and V153T; P151N, V153T, and T172N; G226N and K228T.
14. The polypeptide according to any one of claims 1-7, wherein the amino acid sequence has been modified by introducing between 1 and 10 amino acid residues at the C-terminus of Factor IX polypeptide resulting in the introduction of one or more glycosylation sites.
15. The polypeptide according to claim 14, wherein a polypeptide comprising the amino acid sequence selected from SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: 6, and SEQ ID NO: 7 is introduced at the C-terminus of Factor IX polypeptide.
16. A Factor IX polypeptide comprising an R338A substitution and a V86A substitution.
17. The polypeptide according to any one of claims 1 to 15, wherein attachment of a carbohydrate chain at one or more of the introduced glycosylation sites increases serum half-life of the polypeptide by at least 30% relative to the polypeptide lacking the introduced glycosylation sites.
18. The polypeptide according to any one of claims 1 to 17, wherein the polypeptide has a specific activity of at least 100 units per mg of polypeptide.
19. A pharmaceutical preparation comprising the Factor IX polypeptide of any one of claims 1-18 and a pharmaceutically acceptable carrier.
20. A method of treating hemophilia B comprising administering to a subject in need thereof a therapeutically effective amount of the pharmaceutical preparation of claim 19.
21. A DNA sequence encoding the polypeptide of any one of claims 1-18.
22. A eukaryotic host cell transfected with the DNA sequence according to claim 20 in a manner allowing the host cell to express a Factor IX polypeptide.
23. A method for producing a Factor IX polypeptide comprising (i) modifying the amino acid sequence of the polypeptide by introducing one or more glycosylation sites; (ii) expressing the polypeptide in a manner which allows glycosylation at the one or more glycosylation sites; and (iii) purifying the polypeptide.
PCT/US2009/040813 2008-04-16 2009-04-16 Modified factor ix polypeptides and uses thereof WO2009137254A2 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA2721683A CA2721683A1 (en) 2008-04-16 2009-04-16 Modified factor ix polypeptides and uses thereof
MX2010011345A MX2010011345A (en) 2008-04-16 2009-04-16 Modified factor ix polypeptides and uses thereof.
JP2011505204A JP2011517951A (en) 2008-04-16 2009-04-16 Modified factor IX polypeptides and their use
BRPI0910702A BRPI0910702A2 (en) 2008-04-16 2009-04-16 modified factor ix polypeptides and uses thereof
CN2009801227842A CN102083856A (en) 2008-04-16 2009-04-16 Modified factor IX polypeptides and uses thereof
EP09743241A EP2288622A4 (en) 2008-04-16 2009-04-16 Modified factor ix polypeptides and uses thereof
AU2009244633A AU2009244633A1 (en) 2008-04-16 2009-04-16 Modified Factor IX polypeptides and uses thereof
IL208718A IL208718A0 (en) 2008-04-16 2010-10-14 Modified factor ix polypeptides and uses thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US12456708P 2008-04-16 2008-04-16
US61/124,567 2008-04-16
US4596108P 2008-04-17 2008-04-17
US61/045,961 2008-04-17

Publications (2)

Publication Number Publication Date
WO2009137254A2 true WO2009137254A2 (en) 2009-11-12
WO2009137254A3 WO2009137254A3 (en) 2010-01-14

Family

ID=41265284

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/040813 WO2009137254A2 (en) 2008-04-16 2009-04-16 Modified factor ix polypeptides and uses thereof

Country Status (17)

Country Link
EP (1) EP2288622A4 (en)
JP (1) JP2011517951A (en)
KR (1) KR20110005862A (en)
CN (1) CN102083856A (en)
AU (1) AU2009244633A1 (en)
BR (1) BRPI0910702A2 (en)
CA (1) CA2721683A1 (en)
CO (1) CO6311000A2 (en)
CR (1) CR11737A (en)
DO (1) DOP2010000311A (en)
EC (1) ECSP10010551A (en)
IL (1) IL208718A0 (en)
MX (1) MX2010011345A (en)
RU (1) RU2010146387A (en)
SG (1) SG189790A1 (en)
SV (1) SV2010003704A (en)
WO (1) WO2009137254A2 (en)

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2209487A2 (en) * 2007-10-15 2010-07-28 The University of North Carolina At Chapel Hill Human factor ix variants with an extended half life
US20110244550A1 (en) * 2008-09-15 2011-10-06 Paolo Simioni Factor ix polypeptide mutant, its uses and a method for its production
WO2012061654A1 (en) 2010-11-03 2012-05-10 Catalyst Biosciences, Inc. Modified factor ix polypeptides and uses thereof
US8383388B2 (en) * 2006-06-19 2013-02-26 Catalyst Biosciences, Inc. Modified coagulation factor IX polypeptides and use thereof for treatment
WO2013162078A1 (en) * 2012-04-27 2013-10-31 学校法人日本大学 Therapeutic agent for injury in epithelium and endothelium
WO2014063108A1 (en) 2012-10-18 2014-04-24 Biogen Idec Ma Inc. Methods of using a fixed dose of a clotting factor
WO2015085276A1 (en) 2013-12-06 2015-06-11 Biogen Idec Ma Inc. Population pharmacokinetics tools and uses thereof
WO2015095809A1 (en) 2013-12-20 2015-06-25 Biogen Idec Ma Inc. Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality
WO2015148444A1 (en) 2014-03-24 2015-10-01 Biogen Ma Inc. Lyophilized factor ix formulations
US9233145B2 (en) 2010-07-09 2016-01-12 Biogen Hemophila Inc. Factor IX polypeptides and methods of use thereof
AU2013204511B2 (en) * 2010-11-03 2016-03-17 Gc Biopharma Corp. Modified factor ix polypeptides and uses thereof
US9486507B2 (en) 2011-06-10 2016-11-08 Biogen Ma Inc. Pro-coagulant compounds and methods of use thereof
WO2018102743A1 (en) 2016-12-02 2018-06-07 Bioverativ Therapeutics Inc. Methods of treating hemophilic arthropathy using chimeric clotting factors
US10001495B2 (en) 2012-07-25 2018-06-19 Bioverativ Therapeutics Inc. Blood factor monitoring assay and uses thereof
WO2018144623A1 (en) 2017-01-31 2018-08-09 Bioverativ Therapeutics Inc. Factor ix fusion proteins and methods of making and using same
WO2019020966A1 (en) 2017-07-28 2019-01-31 Universite Claude Bernard Lyon 1 Modified protein with improved half-life
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
WO2019067766A1 (en) 2017-09-27 2019-04-04 Sigilon Therapeutics, Inc. Methods, compositions, and implantable elements comprising active cells
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
EP3542861A1 (en) 2012-09-25 2019-09-25 Bioverativ Therapeutics Inc. Methods of using fix polypeptides
WO2019195055A1 (en) 2018-04-04 2019-10-10 Sigilon Therapeutics, Inc. Implantable particles and related methods
WO2019195056A1 (en) 2018-04-04 2019-10-10 Sigilon Therapeutics, Inc. Methods, compositions, and implantable elements comprising stem cells
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
US10584147B2 (en) 2013-11-08 2020-03-10 Biovertiv Therapeutics Inc. Procoagulant fusion compound
US10588949B2 (en) 2013-03-15 2020-03-17 Bioverativ Therapeutics Inc. Factor IX polypeptide formulations
WO2020069429A1 (en) 2018-09-27 2020-04-02 Sigilon Therapeutics, Inc. Implantable devices for cell therapy and related methods
US10611794B2 (en) 2013-09-25 2020-04-07 Bioverativ Therapeutics Inc. On-column viral inactivation methods
WO2020086408A1 (en) 2018-10-26 2020-04-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A high-yield perfusion-based transient gene expression bioprocess
US10656167B2 (en) 2011-07-25 2020-05-19 Bioverativ Therapeutics Inc. Assays to monitor bleeding disorders
US10717965B2 (en) 2013-01-10 2020-07-21 Gloriana Therapeutics, Inc. Mammalian cell culture-produced neublastin antibodies
US10745680B2 (en) 2015-08-03 2020-08-18 Bioverativ Therapeutics Inc. Factor IX fusion proteins and methods of making and using same
WO2020215010A1 (en) 2019-04-17 2020-10-22 Codiak Biosciences, Inc. Compositions of exosomes and aav
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
WO2021154414A2 (en) 2020-01-29 2021-08-05 Catalyst Biosciences, Inc. Gene therapy for hemophilia b with a chimeric aav capsid vector encoding modified factor ix polypeptides
US20220268871A1 (en) * 2021-02-24 2022-08-25 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11491212B1 (en) 2017-09-27 2022-11-08 Catalyst Biosciences, Inc. Subcutaneous administration of modified factor IX polypeptides and treatment of hemophilia B
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
US11787851B2 (en) 2013-02-15 2023-10-17 Bioverativ Therapeutics Inc. Optimized factor VIII gene
WO2024081310A1 (en) 2022-10-11 2024-04-18 Sigilon Therapeutics, Inc. Engineered cells and implantable elements for treatment of disease
WO2024081309A1 (en) 2022-10-11 2024-04-18 Sigilon Therapeutics, Inc. Engineered cells and implantable elements for treatment of disease

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105695616A (en) * 2016-04-22 2016-06-22 王冬国 Analysis marker for diagnosing thyroid cancer and application thereof

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2638643B1 (en) * 1988-11-09 1991-04-12 Transgene Sa DNA SEQUENCE ENCODING HUMAN FACTOR IX OR AN ANALOGUE PROTEIN, EXPRESSION VECTOR, TRANSFORMED CELLS, PROCESS FOR PREPARING FACTOR IX, AND PRODUCTS OBTAINED THEREFROM
WO1999003496A1 (en) * 1997-07-21 1999-01-28 The University Of North Carolina At Chapel Hill Factor ix antihemophilic factor with increased clotting activity
RU2004110239A (en) * 2001-09-04 2005-10-20 Мерк Патент ГмбХ (DE) MODIFIED FACTOR IX
CA2520970A1 (en) * 2003-04-09 2004-10-28 Robert G. Schaub Hemophilia treatment by inhalation of coagulation factors
EP1694315A4 (en) * 2003-12-03 2009-10-28 Novo Nordisk As Glycopegylated factor ix
US20060040856A1 (en) * 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
EP2423307A1 (en) * 2006-06-19 2012-02-29 Catalyst Biosciences, Inc. Modified coagulation factor IV polypeptides and use thereof for treatment
US7700734B2 (en) * 2007-01-09 2010-04-20 Shu-Wha Lin Recombinant human factor IX and use thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2288622A4 *

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8383388B2 (en) * 2006-06-19 2013-02-26 Catalyst Biosciences, Inc. Modified coagulation factor IX polypeptides and use thereof for treatment
EP2209487A4 (en) * 2007-10-15 2012-06-20 Univ North Carolina Human factor ix variants with an extended half life
EP2209487A2 (en) * 2007-10-15 2010-07-28 The University of North Carolina At Chapel Hill Human factor ix variants with an extended half life
US9249405B2 (en) * 2008-09-15 2016-02-02 Paolo Simioni Factor IX polypeptide mutant, its uses and a method for its production
US20110244550A1 (en) * 2008-09-15 2011-10-06 Paolo Simioni Factor ix polypeptide mutant, its uses and a method for its production
US10465180B2 (en) 2008-09-15 2019-11-05 Uniqure Biopharma B.V. Factor IX polypeptide mutant, its uses and method for its production
US9982248B2 (en) 2008-09-15 2018-05-29 Uniqure Biopharma B.V. Factor IX polypeptide mutant, its uses and method for its production
EP3552619A1 (en) 2010-07-09 2019-10-16 Bioverativ Therapeutics Inc. Factor ix polypeptides and methods of use thereof
US9623091B2 (en) 2010-07-09 2017-04-18 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US10548954B2 (en) 2010-07-09 2020-02-04 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US9629903B2 (en) 2010-07-09 2017-04-25 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US10898554B1 (en) 2010-07-09 2021-01-26 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US10561714B2 (en) 2010-07-09 2020-02-18 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US9867873B2 (en) 2010-07-09 2018-01-16 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US9675676B2 (en) 2010-07-09 2017-06-13 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US9233145B2 (en) 2010-07-09 2016-01-12 Biogen Hemophila Inc. Factor IX polypeptides and methods of use thereof
US9670475B2 (en) 2010-07-09 2017-06-06 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
US10568943B2 (en) 2010-07-09 2020-02-25 Bioverativ Therapeutics Inc. Factor IX polypeptides and methods of use thereof
AU2013204511B2 (en) * 2010-11-03 2016-03-17 Gc Biopharma Corp. Modified factor ix polypeptides and uses thereof
KR101841906B1 (en) 2010-11-03 2018-03-26 주식회사 이수앱지스 Modified factor ix polypeptides and uses thereof
CN103282491B (en) * 2010-11-03 2016-08-24 催化剂生物科学公司 Factor IX polypeptide modified and application thereof
WO2012061654A1 (en) 2010-11-03 2012-05-10 Catalyst Biosciences, Inc. Modified factor ix polypeptides and uses thereof
TWI557135B (en) * 2010-11-03 2016-11-11 介控生化科技公司 Modified factor ix polypeptides and uses thereof
US9328339B2 (en) 2010-11-03 2016-05-03 Catalyst Biosciences, Inc. Modified factor IX polypeptides and uses thereof
CN103282491A (en) * 2010-11-03 2013-09-04 催化剂生物科学公司 Modified factor ix polypeptides and uses thereof
JP2013545457A (en) * 2010-11-03 2013-12-26 カタリスト・バイオサイエンシーズ・インコーポレイテッド Modified factor IX polypeptides and uses thereof
US8778870B2 (en) 2010-11-03 2014-07-15 Catalyst Biosciences, Inc. Modified factor IX polypeptides and uses thereof
TWI595004B (en) * 2010-11-03 2017-08-11 介控生化科技公司 Modified factor ix polypeptides and uses thereof
AU2011323295B2 (en) * 2010-11-03 2015-05-28 Gc Biopharma Corp. Modified factor IX polypeptides and uses thereof
KR101790589B1 (en) 2010-11-03 2017-10-27 주식회사 이수앱지스 Modified factor ix polypeptides and uses thereof
EP3663399A1 (en) 2010-11-03 2020-06-10 Catalyst Biosciences, Inc. Modified factor ix polypeptides and uses thereof
JP2016116519A (en) * 2010-11-03 2016-06-30 カタリスト・バイオサイエンシーズ・インコーポレイテッドCatalyst Biosciences, Inc. Modified Factor IX polypeptides and uses thereof
US10982203B2 (en) 2010-11-03 2021-04-20 Catalyst Biosciences, Inc. Modified factor IX polypeptides and uses thereof
US9486507B2 (en) 2011-06-10 2016-11-08 Biogen Ma Inc. Pro-coagulant compounds and methods of use thereof
EP3527218A1 (en) 2011-06-10 2019-08-21 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
US10656167B2 (en) 2011-07-25 2020-05-19 Bioverativ Therapeutics Inc. Assays to monitor bleeding disorders
US11747351B2 (en) 2011-07-25 2023-09-05 Bioverativ Therapeutics Inc. Assays to monitor bleeding disorders
US9738884B2 (en) 2012-04-27 2017-08-22 Nihon University Therapeutic agent for epithelial and endothelial injury
WO2013162078A1 (en) * 2012-04-27 2013-10-31 学校法人日本大学 Therapeutic agent for injury in epithelium and endothelium
JPWO2013162078A1 (en) * 2012-04-27 2015-12-24 学校法人日本大学 Treatment for epithelial and endothelial damage
US11372008B2 (en) 2012-07-25 2022-06-28 Bioverativ Therapeutics Inc. Blood factor monitoring assay and uses thereof
US10001495B2 (en) 2012-07-25 2018-06-19 Bioverativ Therapeutics Inc. Blood factor monitoring assay and uses thereof
US11225650B2 (en) 2012-09-25 2022-01-18 Bioverativ Therapeutics Inc. Methods of using FIX polypeptides
EP3542861A1 (en) 2012-09-25 2019-09-25 Bioverativ Therapeutics Inc. Methods of using fix polypeptides
US10391152B2 (en) 2012-10-18 2019-08-27 Bioverativ Therapeutics Inc. Methods of using a fixed dose of a clotting factor
WO2014063108A1 (en) 2012-10-18 2014-04-24 Biogen Idec Ma Inc. Methods of using a fixed dose of a clotting factor
US10717965B2 (en) 2013-01-10 2020-07-21 Gloriana Therapeutics, Inc. Mammalian cell culture-produced neublastin antibodies
US11787851B2 (en) 2013-02-15 2023-10-17 Bioverativ Therapeutics Inc. Optimized factor VIII gene
US11642398B2 (en) 2013-03-15 2023-05-09 Bioverativ Therapeutics Inc. Factor IX polypeptide formulations
US10588949B2 (en) 2013-03-15 2020-03-17 Bioverativ Therapeutics Inc. Factor IX polypeptide formulations
US11578098B2 (en) 2013-09-25 2023-02-14 Bioverativ Therapeutics Inc. On-column viral inactivation methods
EP3903599A1 (en) 2013-09-25 2021-11-03 Bioverativ Therapeutics Inc. On-column viral inactivation methods
US10611794B2 (en) 2013-09-25 2020-04-07 Bioverativ Therapeutics Inc. On-column viral inactivation methods
US10584147B2 (en) 2013-11-08 2020-03-10 Biovertiv Therapeutics Inc. Procoagulant fusion compound
WO2015085276A1 (en) 2013-12-06 2015-06-11 Biogen Idec Ma Inc. Population pharmacokinetics tools and uses thereof
US10325687B2 (en) 2013-12-06 2019-06-18 Bioverativ Therapeutics Inc. Population pharmacokinetics tools and uses thereof
EP4332839A2 (en) 2013-12-06 2024-03-06 Bioverativ Therapeutics Inc. Population pharmacokinetics tools and uses thereof
WO2015095809A1 (en) 2013-12-20 2015-06-25 Biogen Idec Ma Inc. Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality
US10772942B2 (en) 2014-03-24 2020-09-15 Bioverativ Therapeutics Inc. Lyophilized factor IX formulations
WO2015148444A1 (en) 2014-03-24 2015-10-01 Biogen Ma Inc. Lyophilized factor ix formulations
US11008561B2 (en) 2014-06-30 2021-05-18 Bioverativ Therapeutics Inc. Optimized factor IX gene
US10413598B2 (en) 2014-11-12 2019-09-17 Ucl Business Plc Factor IX gene therapy
US11344608B2 (en) 2014-11-12 2022-05-31 Ucl Business Ltd Factor IX gene therapy
US10745680B2 (en) 2015-08-03 2020-08-18 Bioverativ Therapeutics Inc. Factor IX fusion proteins and methods of making and using same
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes
WO2018102743A1 (en) 2016-12-02 2018-06-07 Bioverativ Therapeutics Inc. Methods of treating hemophilic arthropathy using chimeric clotting factors
WO2018144623A1 (en) 2017-01-31 2018-08-09 Bioverativ Therapeutics Inc. Factor ix fusion proteins and methods of making and using same
WO2019020966A1 (en) 2017-07-28 2019-01-31 Universite Claude Bernard Lyon 1 Modified protein with improved half-life
WO2019032898A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
US11491212B1 (en) 2017-09-27 2022-11-08 Catalyst Biosciences, Inc. Subcutaneous administration of modified factor IX polypeptides and treatment of hemophilia B
WO2019067766A1 (en) 2017-09-27 2019-04-04 Sigilon Therapeutics, Inc. Methods, compositions, and implantable elements comprising active cells
WO2019195055A1 (en) 2018-04-04 2019-10-10 Sigilon Therapeutics, Inc. Implantable particles and related methods
WO2019195056A1 (en) 2018-04-04 2019-10-10 Sigilon Therapeutics, Inc. Methods, compositions, and implantable elements comprising stem cells
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
US11517631B2 (en) 2018-08-20 2022-12-06 Ucl Business Ltd Factor IX encoding nucleotides
WO2020069429A1 (en) 2018-09-27 2020-04-02 Sigilon Therapeutics, Inc. Implantable devices for cell therapy and related methods
WO2020086408A1 (en) 2018-10-26 2020-04-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A high-yield perfusion-based transient gene expression bioprocess
WO2020215010A1 (en) 2019-04-17 2020-10-22 Codiak Biosciences, Inc. Compositions of exosomes and aav
WO2021154414A2 (en) 2020-01-29 2021-08-05 Catalyst Biosciences, Inc. Gene therapy for hemophilia b with a chimeric aav capsid vector encoding modified factor ix polypeptides
US11742578B2 (en) 2021-02-24 2023-08-29 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11824280B2 (en) 2021-02-24 2023-11-21 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11695209B2 (en) 2021-02-24 2023-07-04 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11670855B2 (en) 2021-02-24 2023-06-06 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11777215B2 (en) 2021-02-24 2023-10-03 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11664594B2 (en) 2021-02-24 2023-05-30 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11817636B2 (en) * 2021-02-24 2023-11-14 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11742579B2 (en) 2021-02-24 2023-08-29 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11843188B2 (en) 2021-02-24 2023-12-12 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11870159B2 (en) 2021-02-24 2024-01-09 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US20220268871A1 (en) * 2021-02-24 2022-08-25 Bluehalo, Llc System and method for a digitally beamformed phased array feed
US11955727B2 (en) 2021-02-24 2024-04-09 Bluehalo, Llc System and method for a digitally beamformed phased array feed
WO2024081310A1 (en) 2022-10-11 2024-04-18 Sigilon Therapeutics, Inc. Engineered cells and implantable elements for treatment of disease
WO2024081309A1 (en) 2022-10-11 2024-04-18 Sigilon Therapeutics, Inc. Engineered cells and implantable elements for treatment of disease

Also Published As

Publication number Publication date
CA2721683A1 (en) 2009-11-12
RU2010146387A (en) 2012-05-27
ECSP10010551A (en) 2010-11-30
DOP2010000311A (en) 2011-02-28
KR20110005862A (en) 2011-01-19
SV2010003704A (en) 2011-02-21
CR11737A (en) 2011-02-07
EP2288622A2 (en) 2011-03-02
CO6311000A2 (en) 2011-08-22
IL208718A0 (en) 2010-12-30
CN102083856A (en) 2011-06-01
BRPI0910702A2 (en) 2016-07-05
WO2009137254A3 (en) 2010-01-14
EP2288622A4 (en) 2012-04-18
MX2010011345A (en) 2011-02-23
AU2009244633A1 (en) 2009-11-12
SG189790A1 (en) 2013-05-31
JP2011517951A (en) 2011-06-23

Similar Documents

Publication Publication Date Title
EP2288622A2 (en) Modified factor ix polypeptides and uses thereof
TWI595004B (en) Modified factor ix polypeptides and uses thereof
ES2704083T3 (en) Modified factor x polypeptides and uses thereof
DK1891091T3 (en) Coagulation factor-X polypeptides with modified activation properties
AU2010290131C1 (en) Coagulation factor VII compositions and methods of making and using same
EP2282767A2 (en) Site-directed modification of factor ix
EP1991255B1 (en) Coagulation factor x polypeptides with modified activation properties
US20120164130A1 (en) Modified Factor IX Polypeptides and Uses Thereof
EP2108046B9 (en) Fviii-independent fix-mutant proteins for hemophilia a treatment
US20120178693A1 (en) Cofactors for Thrombin Activation of Factor VII and Uses Thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980122784.2

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09743241

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2721683

Country of ref document: CA

Ref document number: 588611

Country of ref document: NZ

Ref document number: 2009244633

Country of ref document: AU

Ref document number: 10128682

Country of ref document: CO

Ref document number: 201011737

Country of ref document: CR

Ref document number: D2010201

Country of ref document: CU

Ref document number: 7309/DELNP/2010

Country of ref document: IN

Ref document number: 12010502349

Country of ref document: PH

Ref document number: MX/A/2010/011345

Country of ref document: MX

Ref document number: CR2010-011737

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 2011505204

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2009743241

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009244633

Country of ref document: AU

Date of ref document: 20090416

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20107025567

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010146387

Country of ref document: RU

Ref document number: A20101635

Country of ref document: BY

WWE Wipo information: entry into national phase

Ref document number: 224411

Country of ref document: IL

ENP Entry into the national phase

Ref document number: PI0910702

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20101015