WO2007126808A1 - Pegylated factor viii - Google Patents

Pegylated factor viii Download PDF

Info

Publication number
WO2007126808A1
WO2007126808A1 PCT/US2007/007560 US2007007560W WO2007126808A1 WO 2007126808 A1 WO2007126808 A1 WO 2007126808A1 US 2007007560 W US2007007560 W US 2007007560W WO 2007126808 A1 WO2007126808 A1 WO 2007126808A1
Authority
WO
WIPO (PCT)
Prior art keywords
fviii
construct
factor viii
pegylated
molecule
Prior art date
Application number
PCT/US2007/007560
Other languages
French (fr)
Inventor
Juergen Siekmann
Katalin Varadi
Herbert Gritsch
Peter Turecek
Original Assignee
Baxter International Inc
Baxter Healthcare S.A.
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 Baxter International Inc, Baxter Healthcare S.A. filed Critical Baxter International Inc
Priority to BRPI0708832-9A priority Critical patent/BRPI0708832A2/en
Priority to NZ572050A priority patent/NZ572050A/en
Priority to JP2009502944A priority patent/JP2009532351A/en
Priority to MX2008012600A priority patent/MX2008012600A/en
Priority to EP07754130A priority patent/EP2010222A1/en
Priority to AU2007245190A priority patent/AU2007245190B2/en
Priority to CA002647314A priority patent/CA2647314A1/en
Publication of WO2007126808A1 publication Critical patent/WO2007126808A1/en
Priority to NO20084608A priority patent/NO20084608L/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • 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
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/745Blood coagulation or fibrinolysis factors
    • C07K14/755Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)

Definitions

  • the present invention relates to a proteinaceous construct comprising coagulation factor VIII (FVIII) being bound to at least one soluble polymer, such as a poly(alkylene oxide) such as polyethylene glycol. Further the present invention relates to methods for prolonging the in vivo-half-life of FVIII in the blood of a mammal having a bleeding disorder associated with functional defects or deficiencies of FVIII.
  • FVIII coagulation factor VIII
  • Coagulation factor VIII (FVIII) circulates, in plasma at a very low concentration and is bound non-covalently to von Willebrand factor (VWF). During hemostasis, FVIII is separated from VWF and acts as a cofactor for activated factor IX (FIXa)- mediated factor X (FX) activation by enhancing the rate of activation in the presence of calcium and phospholipids or cellular membranes.
  • VWF von Willebrand factor
  • FVIII is synthesized as a single-chain precursor of approximately 270-330 kD with the domain structure A1-A2-B-A3-C1-C2.
  • FVIII is composed of a heavy chain (A1-A2-B) and a light chain (A3-C1-C2).
  • the molecular mass of the light chain is 80 kD whereas, due to proteolysis within the B domain, the heavy chain is in the range of 90-220 kD.
  • FVIII is also synthesized as a recombinant protein for therapeutic use in bleeding disorders.
  • Various in vitro assays have been devised to determine the potential efficacy of recombinant FVIII (rFVIII) as a therapeutic medicine. These assays mimic the in vivo effects of endogenous FVIII.
  • in vitro thrombin treatment of FVlII results in a rapid increase and subsequent decrease in its procoagulant activity, as measured by in vitro assay. This activation and inactivation coincides with specific limited proteolysis both in the heavy and the light chains, which alter the availability of different binding epitopes in FVIII, e.g. allowing FVIII to dissociate from VWF and bind to a phospholipid surface or altering the binding ability to certain monoclonal antibodies.
  • the lack or dysfunction of FVIII is associated with the most frequent bleeding disorder, hemophilia A.
  • the treatment of choice for the management of hemophilia A is replacement therapy with plasma derived or rFVIII concentrates. Patients with severe haemophilia A with FVIII levels below 1%, are generally on prophylactic therapy with the aim of keeping FVIII above 1% between doses. Taking into account the average half-lives of the various FVIII products in the circulation, this can usually be achieved by giving FVIII two to three times a week.
  • Therapeutic polypeptide drugs are rapidly degraded by proteolytic enzymes and neutralized by antibodies. This reduces their half-life and circulation time, thereby limiting their therapeutic effectiveness.
  • the addition of a soluble polymer or carbohydrate to a polypeptide has been shown to prevent degradation and increase the polypeptides half-life.
  • PEGylation of polypeptide drugs protects them and improves their pharmacodynamic and pharmacokinetic profiles (Harris JM et Chess RB, Nat Rev Drug Discov 2003;2:214-21).
  • the PEGylation process attaches repeating units of polyethylene glycol (PEG) to a polypeptide drug.
  • PEGylation of molecules can lead to increased resistance of drugs to enzymatic degradation, increased half-life in vivo, reduced dosing frequency, decreased immunoge ⁇ icity, increased physical and thermal stability, increased solubility, increased liquid stability, and reduced aggregation.
  • US6037452 describes a poly(alkylene oxide)-FVIII or FIX conjugate, where the protein is covalently bound to a poly(alkylene oxide) through carbonyl- groups of satd FVIII.
  • EP1258497B1 describes a method to prepare conjugates of FVIII and a biocompatible polymer. This patent was supplemented by a publication of R ⁇ stin et al. (Bioconj Chem 2000; 11 :387-96).
  • the conjugates comprise a B-domain deleted recombinant FVIII modified with monomethoxy polyethylene glycol. The conjugate had reduced FVIII function and the coagulant activity decreased rapidly with the degree of modification.
  • WO04075923A3 describes polymer-FVIII molecular conjugate comprising a plurality of conjugates wherein each conjugate has one to three water soluble polymers covalently attached to an FVIII molecule.
  • the FVIII molecule is B-domain- deleted.
  • US4970300 describes a modified FVIII, wherein an infusible conjugate comprising a protein having FVIII activity was covalently linked to a nonantigenic ligand.
  • US6048720 describes conjugates of a polypeptide and a biocompatible polymer.
  • WO94/15625 describes FVIII bound to polyethylene glycol having a preferred molecular weight of no greater than 5,000 Daltons.
  • an FVIII having an attached soluble polymer to extend the half-life of the FVIII in vivo, for example, a PEGylated FVIlI, such as full- length FVIII having PEG greater than 10,000 Daltons conjugated thereto, which retains functional activity while providing an extended half-life in vivo, as compared to non-PEGylated FVIII.
  • a PEGylated FVIlI such as full- length FVIII having PEG greater than 10,000 Daltons conjugated thereto, which retains functional activity while providing an extended half-life in vivo, as compared to non-PEGylated FVIII.
  • Figure 1 shows the broadening and mass increase of rFVIII after conjugation with PEG measured by SDS-PAGE with subsequent immunoblotting.
  • Figure 2 shows the pharmacokinetics of PEG-rFVIII conjugate compared to non-conjugated FVIII in hemophilic mice. Open squares: PEGrFVIII, dose 200 IU FVIII/kg. Closed diamonds: native rFVIII, dose 200 IU FVHI/kg.
  • FIG. 3 shows the detailed analysis of PEGylation sites by SDS-PAGE using various anti FVIII antibodies.
  • Figure 4 shows the thrombin-induced activation and inactivation of native and PEGylated rFVIII.
  • Figure 5 shows the bands demonstrating the domains of native and PEGylated rFVIII.
  • Figure 6 shows the extent of PEGylation of various domains of native and PEGylated rFVIII.
  • Figure 7 shows the thrombin inactivation rate of native and PEGylated rFVIII.
  • the invention is a proteinaceous construct comprising an FVIII molecule having at least a portion of the B domain intact, bound to a water-soluble polymer which is a polyalkylene oxide, polyvinyl pyrrolidone, polyvinyl alcohol, polyoxazoline, a poly acryloylmorpholine or a carbohydrate, such as polysialic acid (PSA).
  • a water-soluble polymer which is a polyalkylene oxide, polyvinyl pyrrolidone, polyvinyl alcohol, polyoxazoline, a poly acryloylmorpholine or a carbohydrate, such as polysialic acid (PSA).
  • PSA polysialic acid
  • the water soluble polymer is a polyethylene glycol molecule having a molecular weight of greater than 10,000 Daltons.
  • the construct retains the full functional activity of standard therapeutic FVIII products, and provides an extended half-life in vivo, as compared to standard therapeutic FVIII products
  • the starting material of the present invention is FVIII, which can be derived from human plasma, or produced by recombinant engineering techniques, as described in patents US4757006; US5733873; US5198349; US5250421; US5919766; EP 306 968.
  • the term "Factor VIII” or “FVIII” refers to any FVIII molecule which has at least a portion of the B domain intact, and which exhibits biological activity that is associated with native FVIII.
  • the FVIII molecule is full-length Factor VIII.
  • the FVIII molecule is a protein which is encoded for by DNA sequences capable of hybridizing to DNA encoding Factor VIIhC. Such a protein may contain amino acid deletions at various sites between or within the domains A1-A2-B-A3-C1-C2 (US4868112).
  • the FVIII molecule may also be an analog of native FVIII wherein one or more amino acid residues have been replaced by site-directed mutagenesis.
  • an FVIII molecule can be PEGylated by a variety of chemical methods (Roberts JM et al., Advan Drug Delivery Rev 2002;54:459-76).
  • FVIII can be PEGylated by the conjugation of PEG to free SH groups using maleimide chemistry or the coupling of PEG hydrazides or PEG amines to carbohydrate moieties of the FVIII after prior oxidation.
  • FVIII was modified via lysine residues by use of polyethylene glycol derivatives containing an active N-hydroxysuccinimide ester (NHS) such as Succinimidyl succinate, Succinimidyl glutarate or Succinimidyl propionate. These derivatives react with the lysine residues of FVIII under mild conditions by forming a stable amide bond.
  • the chain length of the PEG derivative is 5,000 Da.
  • Other PEG derivatives with chain lengths of 500 to 2,000 Da, 2,000 to 5,000 Da, greater than 5,000 up to 10,000 Da or greater than 10,000 up to 20,000 Da, or greater than 20,000 up to 150,000 Da can be used, including linear and branched structures.
  • an FVIII molecule is chemically modified using PEG derivatives that are commercially available. These PEG derivatives can have a linear or branched structures. Examples of PEG-derivatives containing NHS groups are listed below. [031] The following PEG derivatives are examples of those commercially available from Nektar Therapeutics (Huntsville, Al; see www.nektar.com/PEG reagent catalog; Nektar Advanced PEGylation, price list 2005-2006):
  • PEG derivatives are commercially available from NOF Corporation (Tokyo, Japan; see www.nof.co.jp/english: Catalogue 2005)
  • PEG derivatives with degradable for example, hydrolysable linkers
  • degradable linkers for example, hydrolysable linkers
  • the PEGylated FVIII of this invention exhibits full functional activity, combined with an extended FVIII half-life in vivo.
  • the PEGylated rFVIII seems to be more resistant against thrombin inactivation. This was shown by a variety of in vitro and in vivo methods, and is illustrated by the following examples.
  • reaction mixture was applied onto an equilibrated ion- exchange chromatography resin (Fractogel EMD TMAE 650M / Pharmacia XK-10 column, bed height: 15.0 cm) in 20 mM Hepes buffer, 150 mM NaCI, pH 7.4, containing 0.5% sucrose and 0.1% Polysorbate 80.
  • the eluate was concentrated by ultrafiltration/diafiltration with a membrane consisting of regenerated cellulose and with a molecular weight cut-off of 30 kD using a buffer system consisting of 20 mM Hepes, 150 mM NaCI, 0.5% sucrose, pH 7.4.
  • RFVIIl derived from the Advate manufacturing process was PEGylated according to Example 1 and the PEGylated FVIII product was biochemically characterized.
  • the functional activity of the PEG-rFVIII was determined by use of the FVIII chromogenic assay (Rosen S, Scand J Haematol 1984;33 (Suppl 40):139-45). The method is based on Ph. Eur. 5th edition (5.05) 2.7.4 Assay of Blood Coagulation Factor VIII.
  • a sample, containing factor VIII (FVIIhC) is mixed with thrombin, activated factor IX (FIXa), phospholipids and factor X (FX) in a buffer containing calcium.
  • FVIII is activated by thrombin and subsequently forms a complex with phospholipids, FIXa and calcium ions.
  • This complex activates factor X to factor Xa, which in turn cleaves the chromogenic substrate FXa-1 (AcOH*CH3OCO-D-CHA-Gly-Arg-pNA).
  • the time course of para-nitroaniline (pNA) released is measured with a micro plate reader at 405 nm. The slope of the reaction is proportional to the factor VIII concentration in the sample.
  • the FVIII antigen value was measured by use of an ELISA system commercially available (Cedarlane, Hornby, Ontario, Canada) with minor modifications. From these values the ratios FVIII chromogen/FVIII antigen were calculated.
  • the protein content in the preparations was determined by measuring the optical density at 280nm. From these data the protein content was calculated (Hoyer LW in: Human Protein Data. Installments 1-6; Heberli Ed.; Wiley VCH, Weinheim, Germany, 1998) and expressed in mg/ml.
  • Native rFVIII was characterized by SDS PAGE under reducing conditions by using a 4-12% polyacrylamide gradient gel obtained from Invitrogen (Carlsbad, California, USA) according to the instructions of the manufacturer. As molecular weight markers (MW) Precision Plus markers (10 kD - 250 kD) obtained from Bio- Rad (Hercules, CA, USA) were used. Then the proteins were transferred on a PVDF membrane obtained from Bio-Rad (Hercules, CA, USA) by electroblotting and subsequently incubated with a polyclonal sheep anti human FVIII:C antibody obtained from Cedarlane (Hornby, Ontario, Canada).
  • MW molecular weight markers
  • Precision Plus markers 10 kD - 250 kD obtained from Bio- Rad (Hercules, CA, USA) were used. Then the proteins were transferred on a PVDF membrane obtained from Bio-Rad (Hercules, CA, USA) by electroblotting and subsequently incubated with a polyclonal sheep anti human
  • the last steps of the immunostaining procedure were the incubation with an alkaline phosphatase (ALP) conjugated anti-sheep antibody obtained from Accurate (Westbury, NY, USA) followed by the final visualization by use of an ALP substrate kit (Bio-Rad, Hercules, CA, USA).
  • ALP alkaline phosphatase
  • the results are summarized in Figure 1.
  • the blot demonstrates the domain structure of native and PEGylated rFVIII. It is shown that the PEGylated rFVII! has broader bands and high molecular masses than the native recombinant protein.
  • FVIII deficient mice described in detail by Bi et al. were used as a model of severe human hemophilia A.
  • Groups of 5 mice received a bolus injection (10 ml/kg) via the tail vein with either PEG-rFVHI (PEG-SS, 5K) prepared according to Example 1 or native rFVIII in a dose of 200 IU FVIII/kg bodyweight.
  • Citrate plasma by heart puncture after anesthesia was prepared from the respective groups, 5 minutes, 3, 6, 9 and 24 hours after injection. FVIII activity levels were measured in plasma samples. The results of this experiment are summarized in Figure 2.
  • FVIII was incubated at 37°C with 0.5 or 1 nM thrombin. Subsamples were withdrawn at time intervals between 0.5 to 40 minutes and added to a mixture of FIXa, FX, PL-vesicles and CaCb also containing a specific thrombin inhibitor to stop the further thrombin-mediated reactions and incubated for 3 minutes. A subsample was added to a chromogenic substrate, which is selectively cleaved by Fxa and contained EDTA to stop further Xa activation. After a 15 min incubation, the reaction was terminated by acetic acid.
  • the absorbance (A405) values which are proportional to the Fxa concentrations, were measured in an ELISA reader and converted to Fxa concentrations using a purified Fxa reference curve. The generated Fxa concentrations were plotted against the incubation time with thrombin. [048] Pseudo-first order inactivation rate of FVIII was determined by fitting the declining part of the curves with a single exponential fit.
  • PEG-SG PEG succinimidyl glutarate
  • reaction mixture was applied onto an equilibrated ion- exchange chromatography resin (Fractogel EMD TMAE 650M / Pharmacia XK-50 column, bed height: 14.5 cm) in 20 mM Hepes buffer, 150 mM NaCI, pH 7.4, containing 0.5% sucrose and 0.1% Polysorbate 80 using a linear flow rate of 1 cm/min.
  • an equilibrated ion- exchange chromatography resin Frazier EMD TMAE 650M / Pharmacia XK-50 column, bed height: 14.5 cm
  • the column was washed with 25 CV equilibration buffer to remove excess reagent (linear flow rate: 2 cm/min) and the PEGylated rFVIII was eluted with elution buffer (20 mM Hepes, 1.0 M NaCI, 0.5% sucrose, 0.1% Polysorbate 80, pH 7.4) at a linear flow rate of 0.5 cm/min. Then the eluate was concentrated by ultrafiltration/diafiltration with a membrane consisting of regenerated cellulose and with a molecular weight cut-off of 30 kD using a buffer system consisting of 20 mM Hepes, 150 mM NaCI, 0.5% sucrose, pH 7.4.
  • RFVIII derived from the Advate manufacturing process was PEGylated via lysine residues using a branched PEG-SG reagent according to Example 7 and the PEGylated rFVIII product was biochemically characterized as described in Example 2.
  • the PEGylated rFVIIl was characterized by SDS-PAGE and immunoblotting techniques under reducing conditions using a 4-12% polyacrylamide gradient gel as described in Example 3. The results are summarized in Figure 5. The blot demonstrates the domain structure of native and PEGylated rFVIIl. It is shown that the PEGylated rFVIIl has broader bands and high molecular masses than the native recombinant protein.

Abstract

The invention is a proteinaceous construct comprising a Factor VIII molecule having at least a portion of the B domain intact, which is conjugated to a water-soluble polymer such as polyethylene glycol having a molecular weight of greater than 10,000 Daltons. The construct has a biological activity of at least 80% of the biological activity of native Factor VIII, and the in vivo half-life of the construct is increased by at least 1.5 fold as compared to the in vivo half-life of native factor FVIII.

Description

PEGYLATED FACTOR VIII
FIELD OF THE INVENTION
[001] The present invention relates to a proteinaceous construct comprising coagulation factor VIII (FVIII) being bound to at least one soluble polymer, such as a poly(alkylene oxide) such as polyethylene glycol. Further the present invention relates to methods for prolonging the in vivo-half-life of FVIII in the blood of a mammal having a bleeding disorder associated with functional defects or deficiencies of FVIII.
BACKGROUND OF THE INVENTION
[002] Coagulation factor VIII (FVIII) circulates, in plasma at a very low concentration and is bound non-covalently to von Willebrand factor (VWF). During hemostasis, FVIII is separated from VWF and acts as a cofactor for activated factor IX (FIXa)- mediated factor X (FX) activation by enhancing the rate of activation in the presence of calcium and phospholipids or cellular membranes.
[003] FVIII is synthesized as a single-chain precursor of approximately 270-330 kD with the domain structure A1-A2-B-A3-C1-C2. When purified from plasma, FVIII is composed of a heavy chain (A1-A2-B) and a light chain (A3-C1-C2). The molecular mass of the light chain is 80 kD whereas, due to proteolysis within the B domain, the heavy chain is in the range of 90-220 kD.
[004] FVIII is also synthesized as a recombinant protein for therapeutic use in bleeding disorders. Various in vitro assays have been devised to determine the potential efficacy of recombinant FVIII (rFVIII) as a therapeutic medicine. These assays mimic the in vivo effects of endogenous FVIII. in vitro thrombin treatment of FVlII results in a rapid increase and subsequent decrease in its procoagulant activity, as measured by in vitro assay. This activation and inactivation coincides with specific limited proteolysis both in the heavy and the light chains, which alter the availability of different binding epitopes in FVIII, e.g. allowing FVIII to dissociate from VWF and bind to a phospholipid surface or altering the binding ability to certain monoclonal antibodies.
[005] The lack or dysfunction of FVIII is associated with the most frequent bleeding disorder, hemophilia A. The treatment of choice for the management of hemophilia A is replacement therapy with plasma derived or rFVIII concentrates. Patients with severe haemophilia A with FVIII levels below 1%, are generally on prophylactic therapy with the aim of keeping FVIII above 1% between doses. Taking into account the average half-lives of the various FVIII products in the circulation, this can usually be achieved by giving FVIII two to three times a week.
[006] There are many concentrates on the market for the treatment of hemophilia A. One of these concentrates is the recombinant product Advate®, which is produced in CHO-cells and manufactured by Baxter Healthcare Corporation. No human or animal plasma proteins or albumin are added in the cell culture process, purification, or final formulation of this product.
[007] The aim of many manufacturers of FVIII concentrates and therapeutic polypeptide drugs is to develop a next generation product with enhanced pharmacodynamic and pharmacokinetic properties, while maintaining all other product characteristics.
[008] Therapeutic polypeptide drugs are rapidly degraded by proteolytic enzymes and neutralized by antibodies. This reduces their half-life and circulation time, thereby limiting their therapeutic effectiveness. The addition of a soluble polymer or carbohydrate to a polypeptide has been shown to prevent degradation and increase the polypeptides half-life. For instance, PEGylation of polypeptide drugs protects them and improves their pharmacodynamic and pharmacokinetic profiles (Harris JM et Chess RB, Nat Rev Drug Discov 2003;2:214-21). The PEGylation process attaches repeating units of polyethylene glycol (PEG) to a polypeptide drug. PEGylation of molecules can lead to increased resistance of drugs to enzymatic degradation, increased half-life in vivo, reduced dosing frequency, decreased immunogeπicity, increased physical and thermal stability, increased solubility, increased liquid stability, and reduced aggregation.
[009] Thus, the addition of a soluble polymer, such as through PEGylation is one approach to improve the properties of a FVJII product. The state of the art is documented by different patents and patent applications:
[010] US6037452 describes a poly(alkylene oxide)-FVIII or FIX conjugate, where the protein is covalently bound to a poly(alkylene oxide) through carbonyl- groups of satd FVIII.
[011] EP1258497B1 describes a method to prepare conjugates of FVIII and a biocompatible polymer. This patent was supplemented by a publication of Rόstin et al. (Bioconj Chem 2000; 11 :387-96). The conjugates comprise a B-domain deleted recombinant FVIII modified with monomethoxy polyethylene glycol. The conjugate had reduced FVIII function and the coagulant activity decreased rapidly with the degree of modification.
[012] WO04075923A3 describes polymer-FVIII molecular conjugate comprising a plurality of conjugates wherein each conjugate has one to three water soluble polymers covalently attached to an FVIII molecule. The FVIII molecule is B-domain- deleted.
[013] US4970300 describes a modified FVIII, wherein an infusible conjugate comprising a protein having FVIII activity was covalently linked to a nonantigenic ligand. [014] US6048720 describes conjugates of a polypeptide and a biocompatible polymer.
[015] WO94/15625 describes FVIII bound to polyethylene glycol having a preferred molecular weight of no greater than 5,000 Daltons.
[016] There remains a need for an FVIII having an attached soluble polymer to extend the half-life of the FVIII in vivo, for example, a PEGylated FVIlI, such as full- length FVIII having PEG greater than 10,000 Daltons conjugated thereto, which retains functional activity while providing an extended half-life in vivo, as compared to non-PEGylated FVIII.
FIGURES
[017] Figure 1 shows the broadening and mass increase of rFVIII after conjugation with PEG measured by SDS-PAGE with subsequent immunoblotting.
[018] Figure 2 shows the pharmacokinetics of PEG-rFVIII conjugate compared to non-conjugated FVIII in hemophilic mice. Open squares: PEGrFVIII, dose 200 IU FVIII/kg. Closed diamonds: native rFVIII, dose 200 IU FVHI/kg.
[019] Figure 3 shows the detailed analysis of PEGylation sites by SDS-PAGE using various anti FVIII antibodies.
[020] Figure 4 shows the thrombin-induced activation and inactivation of native and PEGylated rFVIII.
[021] Figure 5 shows the bands demonstrating the domains of native and PEGylated rFVIII. [022] Figure 6 shows the extent of PEGylation of various domains of native and PEGylated rFVIII.
[023] Figure 7 shows the thrombin inactivation rate of native and PEGylated rFVIII.
DETAILED DESCRIPTION OF THE INVENTION
[024] The invention is a proteinaceous construct comprising an FVIII molecule having at least a portion of the B domain intact, bound to a water-soluble polymer which is a polyalkylene oxide, polyvinyl pyrrolidone, polyvinyl alcohol, polyoxazoline, a poly acryloylmorpholine or a carbohydrate, such as polysialic acid (PSA). In one embodiment of the invention, the water soluble polymer is a polyethylene glycol molecule having a molecular weight of greater than 10,000 Daltons. The construct retains the full functional activity of standard therapeutic FVIII products, and provides an extended half-life in vivo, as compared to standard therapeutic FVIII products.
[025] The starting material of the present invention is FVIII, which can be derived from human plasma, or produced by recombinant engineering techniques, as described in patents US4757006; US5733873; US5198349; US5250421; US5919766; EP 306 968.
[026] Herein, the term "Factor VIII" or "FVIII" refers to any FVIII molecule which has at least a portion of the B domain intact, and which exhibits biological activity that is associated with native FVIII. In one embodiment of the invention, the FVIII molecule is full-length Factor VIII. The FVIII molecule is a protein which is encoded for by DNA sequences capable of hybridizing to DNA encoding Factor VIIhC. Such a protein may contain amino acid deletions at various sites between or within the domains A1-A2-B-A3-C1-C2 (US4868112). The FVIII molecule may also be an analog of native FVIII wherein one or more amino acid residues have been replaced by site-directed mutagenesis.
[027] By example, an FVIII molecule can be PEGylated by a variety of chemical methods (Roberts JM et al., Advan Drug Delivery Rev 2002;54:459-76). For example, FVIII can be PEGylated by the conjugation of PEG to free SH groups using maleimide chemistry or the coupling of PEG hydrazides or PEG amines to carbohydrate moieties of the FVIII after prior oxidation.
[028] In one embodiment of the invention, FVIII was modified via lysine residues by use of polyethylene glycol derivatives containing an active N-hydroxysuccinimide ester (NHS) such as Succinimidyl succinate, Succinimidyl glutarate or Succinimidyl propionate. These derivatives react with the lysine residues of FVIII under mild conditions by forming a stable amide bond. In one embodiment of the invention, the chain length of the PEG derivative is 5,000 Da. Other PEG derivatives with chain lengths of 500 to 2,000 Da, 2,000 to 5,000 Da, greater than 5,000 up to 10,000 Da or greater than 10,000 up to 20,000 Da, or greater than 20,000 up to 150,000 Da can be used, including linear and branched structures.
[029] Alternative methods for the PEGylation of amino groups are the chemical conjugation with PEG carbonates by forming urethane bonds, or the reaction with aldehydes or ketones by reductive amination forming secondary amide bonds.
[030] In the present invention an FVIII molecule is chemically modified using PEG derivatives that are commercially available. These PEG derivatives can have a linear or branched structures. Examples of PEG-derivatives containing NHS groups are listed below. [031] The following PEG derivatives are examples of those commercially available from Nektar Therapeutics (Huntsville, Al; see www.nektar.com/PEG reagent catalog; Nektar Advanced PEGylation, price list 2005-2006):
mPEG-Succlnlmidyl propionate (mPEG-SPA)
Figure imgf000009_0001
mPEG-Succinimldyl α-methylbutanoate (mPEG-SMB)
Figure imgf000009_0002
mPEG-CM-HBA-NHS (CM = carboxymethyl; HBA = Hydroxy butyric acid)
Figure imgf000009_0003
Structure of a branched PEG-derivative (Nektar Therapeutics):
Branched PEG N-Hydroxysucclnimidθ (mPEG2-NHS)
Figure imgf000009_0004
[032] This reagent with branched structure is described in more detail by Kozlowski et al. (BioDrugs 2001 ;5:419-29).
[033] Other examples of PEG derivatives are commercially available from NOF Corporation (Tokyo, Japan; see www.nof.co.jp/english: Catalogue 2005)
General Structure of linear PEG - derivatives (NOF Corp.):
Figure imgf000010_0001
X = carboxymethyt
Figure imgf000010_0002
X = carboxypentyl
Figure imgf000010_0003
x = succinate
Figure imgf000011_0001
Succinimidyl succinate
x = glutarate
Figure imgf000011_0002
mPEG Succinimidyl glutarate
Structures of branched PEG-derivatives (NOF Corp.):
2,3-Bis(methylpolyoxyethylene-oxy)-1-(1 ,5-dioxo-5-succinimidyloxy, pentyloxy) propane
Figure imgf000011_0003
2,3-Bis(methylpolyoxyethylene-oxy)-1 -(succinimidyl carboxypentyloxy) propane
Figure imgf000011_0004
[034} These propane derivatives show a glycerol backbone with a 1 ,2 substitution pattern. In the present invention branched PEG derivatives based on glycerol structures with 1 ,3 substitution or other branched structures described in US2003/0143596A1 can also be used.
[035] PEG derivatives with degradable (for example, hydrolysable linkers) as described by Tsubery et al. (J Biol Chem 2004;279:38118-24) and Shechter et al. (WO04089280A3) can also be used in the present invention.
[036] Surprisingly, the PEGylated FVIII of this invention exhibits full functional activity, combined with an extended FVIII half-life in vivo. In addition the PEGylated rFVIII seems to be more resistant against thrombin inactivation. This was shown by a variety of in vitro and in vivo methods, and is illustrated by the following examples.
EXAMPLES
Example 1:
PEGylation of lysine residues in rFVIII with mPEG Succinimidyl succinate
[037] A solution of a rFVIII bulk derived from the Advate manufacturing process (3,400 U/ml) was gel filtrated by use of Econo-Pac 10DG columns (Bio-Rad) using 20 mM Hepes buffer, 150 mM NaCI, pH 7.4, containing 0.5% sucrose and 0.1% Polysorbate 80. Then mPEG Succinimidyl succinate (Abuchowski et al. Cancer Biochim Biophys 1984;7: 175-86) with a chain length of 5,000 Da (PEG-SS 5000) was added to this solution under gentle stirring (5 mg PEG-SS / mg protein) and the pH value was adjusted to 7.4 by drop wise addition of 0.5 M NaOH. Then the PEGylation was carried out under gentle stirring for 1 hour at room temperature.
[038] Subsequently the reaction mixture was applied onto an equilibrated ion- exchange chromatography resin (Fractogel EMD TMAE 650M / Pharmacia XK-10 column, bed height: 15.0 cm) in 20 mM Hepes buffer, 150 mM NaCI, pH 7.4, containing 0.5% sucrose and 0.1% Polysorbate 80. Thep the column was washed with 20 CV equilibration buffer to remove excess reagent and the PEGylated rFVIIl was eluted with elution buffer (20 mM Hepes, 1.0 M NaCI, 0.5% sucrose, 0.1% Polysorbate 80, pH 7.4). The eluate was concentrated by ultrafiltration/diafiltration with a membrane consisting of regenerated cellulose and with a molecular weight cut-off of 30 kD using a buffer system consisting of 20 mM Hepes, 150 mM NaCI, 0.5% sucrose, pH 7.4.
Example 2:
Biochemical characterization of PEGylated rFVIIl in vitro
[039] RFVIIl derived from the Advate manufacturing process was PEGylated according to Example 1 and the PEGylated FVIII product was biochemically characterized. The functional activity of the PEG-rFVIII was determined by use of the FVIII chromogenic assay (Rosen S, Scand J Haematol 1984;33 (Suppl 40):139-45). The method is based on Ph. Eur. 5th edition (5.05) 2.7.4 Assay of Blood Coagulation Factor VIII.
[040] A sample, containing factor VIII (FVIIhC) is mixed with thrombin, activated factor IX (FIXa), phospholipids and factor X (FX) in a buffer containing calcium. FVIII is activated by thrombin and subsequently forms a complex with phospholipids, FIXa and calcium ions. This complex activates factor X to factor Xa, which in turn cleaves the chromogenic substrate FXa-1 (AcOH*CH3OCO-D-CHA-Gly-Arg-pNA). The time course of para-nitroaniline (pNA) released is measured with a micro plate reader at 405 nm. The slope of the reaction is proportional to the factor VIII concentration in the sample. The FVIII antigen value was measured by use of an ELISA system commercially available (Cedarlane, Hornby, Ontario, Canada) with minor modifications. From these values the ratios FVIII chromogen/FVIII antigen were calculated. The protein content in the preparations was determined by measuring the optical density at 280nm. From these data the protein content was calculated (Hoyer LW in: Human Protein Data. Installments 1-6; Heberli Ed.; Wiley VCH, Weinheim, Germany, 1998) and expressed in mg/ml.
Table 1
Figure imgf000014_0001
[041] The data in Table 1 shows that in the PEGylated rFVIII preparation, the biological activity (expressed by the ratio FVIII chromogenic activity to FVIII antigen) is recovered to more than 90% in comparison to the biological activity of the native rFVIII (100%).
Example 3:
Characterization of PEGylated rFVIII by SDS-PAGE and immunoblottinq techniques
[042] Native rFVIII was characterized by SDS PAGE under reducing conditions by using a 4-12% polyacrylamide gradient gel obtained from Invitrogen (Carlsbad, California, USA) according to the instructions of the manufacturer. As molecular weight markers (MW) Precision Plus markers (10 kD - 250 kD) obtained from Bio- Rad (Hercules, CA, USA) were used. Then the proteins were transferred on a PVDF membrane obtained from Bio-Rad (Hercules, CA, USA) by electroblotting and subsequently incubated with a polyclonal sheep anti human FVIII:C antibody obtained from Cedarlane (Hornby, Ontario, Canada). The last steps of the immunostaining procedure were the incubation with an alkaline phosphatase (ALP) conjugated anti-sheep antibody obtained from Accurate (Westbury, NY, USA) followed by the final visualization by use of an ALP substrate kit (Bio-Rad, Hercules, CA, USA). The results are summarized in Figure 1. The blot demonstrates the domain structure of native and PEGylated rFVIII. It is shown that the PEGylated rFVII! has broader bands and high molecular masses than the native recombinant protein.
Example 4:
Pharmacokinetics of PEGylated rFVIII in a FVIII deficient knock out mouse model
[043] FVIII deficient mice described in detail by Bi et al. (Nat Genet 1995;10:119-21 ) were used as a model of severe human hemophilia A. Groups of 5 mice received a bolus injection (10 ml/kg) via the tail vein with either PEG-rFVHI (PEG-SS, 5K) prepared according to Example 1 or native rFVIII in a dose of 200 IU FVIII/kg bodyweight. Citrate plasma by heart puncture after anesthesia was prepared from the respective groups, 5 minutes, 3, 6, 9 and 24 hours after injection. FVIII activity levels were measured in plasma samples. The results of this experiment are summarized in Figure 2. Mean half life increased from 1.9 hours (for native rFVIII) to 4.9 hours (for PEGylated rFVIII), area under curve (AUC) increased from 13.0 to 25.2 hours*IU/ml. Half-life calculation was performed with MicroMath Scientist, model 1 from pharmacokinetic library (MicroMath, Saint Louis, MO, USA).
Example 5:
Detailed analysis of PEGylation of rFViH by SDS-PAGE and immunoblotting techniques
[044] Native and PEGylated rFVIII was digested with 1 nM thrombin for 60 minutes at 600C, which resulted in specific cleavage of the FVIII molecule with well defined degradation products. These heavy- and light chain fragments were separated by SDS-PAGE followed by electroblotting, as described in Example 3. To visualize the cleaved fragments, a polyclonal antibody and monoclonal antibodies against the heavy chain A1 and A2 domains, the B domain and the light chain N-terminal A3 domain were applied.
[045] As seen in Figure 3 all domains were PEGylated, albeit to a different extent. The B domain was strongly PEGylated. Both the A1 and A2 domains of the heavy chain were partially PEGylated. Various PEGylation-degrees (mono-, di-, tri-...) could be observed in the light chain A3-domain. In agreement with Example 6, the PEGylated FVIII seemed to be more resistant to thrombin.
Example 6:
Thrombin-resistancv of PEGylated rFVIII
[046] In vitro thrombin treatment of FVIII results in a rapid increase and subsequent decrease in its procoagulant activity. The rate of activation and inactivation, which depends on the thrombin concentration and on the integrity of FVIII, was monitored by a FIXa cofactor assay, as follows:
[047] FVIII was incubated at 37°C with 0.5 or 1 nM thrombin. Subsamples were withdrawn at time intervals between 0.5 to 40 minutes and added to a mixture of FIXa, FX, PL-vesicles and CaCb also containing a specific thrombin inhibitor to stop the further thrombin-mediated reactions and incubated for 3 minutes. A subsample was added to a chromogenic substrate, which is selectively cleaved by Fxa and contained EDTA to stop further Xa activation. After a 15 min incubation, the reaction was terminated by acetic acid. The absorbance (A405) values, which are proportional to the Fxa concentrations, were measured in an ELISA reader and converted to Fxa concentrations using a purified Fxa reference curve. The generated Fxa concentrations were plotted against the incubation time with thrombin. [048] Pseudo-first order inactivation rate of FVIII was determined by fitting the declining part of the curves with a single exponential fit.
Table 2
Figure imgf000017_0001
[049] As shown in Figure 4 and Table 2, PEGylated rFVIII showed a slower inactivation rate at both applied thrombin concentrations.
Example 7:
PEGylation of lysine residues in rFVIII with branched 2,3-Bis(methylpolvoxyethylene- oxy)-1-(1.5-dioxo-5-succinimidyloxy. pentyloxy) propane
[050] A solution of rFVIII in 20 mM Hepes buffer pH 7.4 containing 150 mM NaCI, 0.5% sucrose and 0.1% Polysorbate 80 was prepared from bulk material derived from the Advate manufacturing process containing 489 IU FVIII / ml. A branched PEG succinimidyl glutarate (PEG-SG) reagent (2,3-Bis(methylpolyoxyethylene-oxy)- 1-(1,5-dioxo-5-succinimidyloxy, pentyloxy) propane) obtained from NOF Corporation (Tokyo, Japan) with a molecular weight of 20 kD was added to 153 ml of this solution under gentle stirring (5 mg reagent / mg protein) and the pH value was adjusted to 7.4 by drop wise addition of 0.5 M NaOH after 10 minutes. Then the PEGylation of rFVIII was performed under gentle stirring for 1 hour at room temperature. [051] Subsequently the reaction mixture was applied onto an equilibrated ion- exchange chromatography resin (Fractogel EMD TMAE 650M / Pharmacia XK-50 column, bed height: 14.5 cm) in 20 mM Hepes buffer, 150 mM NaCI, pH 7.4, containing 0.5% sucrose and 0.1% Polysorbate 80 using a linear flow rate of 1 cm/min. The column was washed with 25 CV equilibration buffer to remove excess reagent (linear flow rate: 2 cm/min) and the PEGylated rFVIII was eluted with elution buffer (20 mM Hepes, 1.0 M NaCI, 0.5% sucrose, 0.1% Polysorbate 80, pH 7.4) at a linear flow rate of 0.5 cm/min. Then the eluate was concentrated by ultrafiltration/diafiltration with a membrane consisting of regenerated cellulose and with a molecular weight cut-off of 30 kD using a buffer system consisting of 20 mM Hepes, 150 mM NaCI, 0.5% sucrose, pH 7.4.
Example 8:
In-vitro characterization of rFVIII PEGylated with branched PEG-SG 2OkD
[052] RFVIII derived from the Advate manufacturing process was PEGylated via lysine residues using a branched PEG-SG reagent according to Example 7 and the PEGylated rFVIII product was biochemically characterized as described in Example 2.
Table 3
Figure imgf000018_0001
[053] The data in Table 3 show that in the PEGylated rFVIIl preparation the biological activity (expressed by the ratio FVIII chromogenic activity to FVIII antigen) completely recovered in comparison to the biological activity of the native rFVIIl (100%).
[054] The PEGylated rFVIIl was characterized by SDS-PAGE and immunoblotting techniques under reducing conditions using a 4-12% polyacrylamide gradient gel as described in Example 3. The results are summarized in Figure 5. The blot demonstrates the domain structure of native and PEGylated rFVIIl. It is shown that the PEGylated rFVIIl has broader bands and high molecular masses than the native recombinant protein.
[055] For more detailed analysis of PEGylation of the rFVIIl preparation by SDS- PAGE and immunoblotting techniques, the native and PEGylated rFVIIl was digested with 1 nM thrombin for 60 minutes at 60°, which resulted in specific cleavage of the FVIII molecule with well defined degradation products, as described in Example 5. The fragments were separated by SDS-PAGE followed by electroblotting and visualized by different anti-FVIII antibodies. As seen in Figure 6, all domains were PEGylated, albeit to a different extent. The B domain was strongly PEGylated. Various PEGylation-degrees (mono-, di-, tri-PEGylation) could be observed in the light chain A3-domain. The results indicate that the PEGylated rFVIIl seemed to be more resistant to thrombin.
[056] The rate of activation and inactivation by thrombin was monitored by a FIXa cofactor assay as described in Example 6. Pseudo-first order inactivation rate of FVIII was determined by fitting the declining part of the curves with a single exponential fit. Table 4
Figure imgf000020_0001
[057] As shown in Figure 7 and Table 4, the PEGylated rFVlll showed a slower inactivation rate at both applied thrombin concentrations.

Claims

1. A proteinaceous construct comprising,
(a) a Factor VlII molecule having at least a portion of the B domain intact; and
(b) at least one polyethylene glycol molecule bound to said Factor VIII molecule, said polyethylene glycol molecule having a molecular weight of greater than 10,000 Daltons; said construct having a biological activity of at least 80% of the biological activity of native Factor VlII1 wherein the biological activities of the construct and of native Factor VIII are determined by the ratios of chromogenic activity to FVIII antigen value (FVIII:Chr/FVIH:Ag), and wherein the in vivo half-life of said construct is increased by at least 1.5-fold as compared to the in vivo half- life of native Factor VIII.
2. The proteinaceous construct of claim 1 wherein said construct has a biological activity of at least 90% of the biological activity of native Factor VIII.
3. The proteinaceous construct of claim 1 wherein said Factor VIII molecule is a recombinant Factor VIII.
4. The proteinaceous construct of claim 1 wherein said Factor VIII molecule is full-length Factor VIII.
5. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a molecular weight of greater than 10,000 Da to about 125,000 Da.
6. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a molecular weight of about 15,000 to about 20,000 Da.
7. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a molecular weight of about 18,000 to about 25,000 Da.
8. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a molecular weight of about 20,000 Da.
9. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a molecular weight of about 20,000 to about 150,000 Da.
10. The proteinaceous construct of claim 1 wherein said polyethylene glycol molecule has a linear or branched structure.
PCT/US2007/007560 2006-03-31 2007-03-29 Pegylated factor viii WO2007126808A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
BRPI0708832-9A BRPI0708832A2 (en) 2006-03-31 2007-03-29 proteinacean construction
NZ572050A NZ572050A (en) 2006-03-31 2007-03-29 Factor VIII conjugated to polyethylene glycol
JP2009502944A JP2009532351A (en) 2006-03-31 2007-03-29 PEGylated factor VIII
MX2008012600A MX2008012600A (en) 2006-03-31 2007-03-29 Pegylated factor viii.
EP07754130A EP2010222A1 (en) 2006-03-31 2007-03-29 Pegylated factor viii
AU2007245190A AU2007245190B2 (en) 2006-03-31 2007-03-29 Pegylated factor VIII
CA002647314A CA2647314A1 (en) 2006-03-31 2007-03-29 Pegylated factor viii
NO20084608A NO20084608L (en) 2006-03-31 2008-10-30 Pegylated Factor VIII

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US78796806P 2006-03-31 2006-03-31
US60/787,968 2006-03-31
US79023906P 2006-04-06 2006-04-06
US60/790,239 2006-04-06

Publications (1)

Publication Number Publication Date
WO2007126808A1 true WO2007126808A1 (en) 2007-11-08

Family

ID=38330191

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/007560 WO2007126808A1 (en) 2006-03-31 2007-03-29 Pegylated factor viii

Country Status (11)

Country Link
US (2) US7683158B2 (en)
EP (1) EP2010222A1 (en)
JP (1) JP2009532351A (en)
KR (1) KR20080108147A (en)
AU (1) AU2007245190B2 (en)
BR (1) BRPI0708832A2 (en)
CA (1) CA2647314A1 (en)
MX (1) MX2008012600A (en)
NO (1) NO20084608L (en)
NZ (1) NZ572050A (en)
WO (1) WO2007126808A1 (en)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008116913A2 (en) * 2007-03-28 2008-10-02 Novo Nordisk A/S Peptide compounds with transient biodegradable pegylation
WO2009108806A1 (en) * 2008-02-27 2009-09-03 Novo Nordisk A/S Conjugated factor viii molecules
WO2010102886A1 (en) * 2009-02-19 2010-09-16 Novo Nordisk A/S Modification of factor viii
JP2011503101A (en) * 2007-11-09 2011-01-27 バクスター・インターナショナル・インコーポレイテッド Modified recombinant factor VIII and von Willebrand factor and methods of use thereof
JP2011519833A (en) * 2008-04-24 2011-07-14 セルティック ファーマ ピーイージー リミテッド Factor IX conjugates with extended half-life
EP2416157A2 (en) 2007-12-27 2012-02-08 Baxter International Inc. Method and compositions for specifically detecting physiologically acceptable polymer molecules
JP2012505901A (en) * 2008-10-15 2012-03-08 バクスター・インターナショナル・インコーポレイテッド PEGylation of recombinant blood clotting factors in the presence of conjugated antibodies
WO2012079979A1 (en) * 2010-12-16 2012-06-21 Novo Nordisk A/S Aqueous factor viii solution
JP2013500375A (en) * 2009-07-27 2013-01-07 リポクセン テクノロジーズ リミテッド Glycopolysial oxidation of non-blood clotting proteins
JP2013500238A (en) * 2008-08-01 2013-01-07 バクスター・インターナショナル・インコーポレイテッド Factor FVIII polymer conjugate
US20130183280A1 (en) * 2010-07-15 2013-07-18 Novo Nordisk A/S Stabilized factor viii variants
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
WO2013156488A3 (en) * 2012-04-16 2014-01-16 Leverton Licence Holdings Limited Optimised subcutaneous therapeutic agents
WO2014173873A1 (en) 2013-04-22 2014-10-30 Csl Behring Gmbh Complex
US9018166B2 (en) 2010-02-16 2015-04-28 Novo Nordisk A/S Conjugated FVIII variants
JP2015155469A (en) * 2015-06-01 2015-08-27 バクスター・インターナショナル・インコーポレイテッドBaxter International Incorp0Rated Fviii-polymer conjugates
EP2387413A4 (en) * 2009-01-19 2015-12-23 Bayer Healthcare Llc Protein conjugate having an endopeptidase-cleavable bioprotective moiety
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US9309507B2 (en) 2010-04-30 2016-04-12 Polytherics Limited Conjugated blood coagulation factor VIIa
JP2016113626A (en) * 2009-07-27 2016-06-23 リポクセン テクノロジーズ リミテッド Glycopolysialylation of non-blood coagulation protein
WO2016142288A1 (en) 2015-03-06 2016-09-15 Csl Behring Recombinant Facility Ag Modified von willebrand factor having improved half-life
US9474790B2 (en) 2013-04-18 2016-10-25 Novo Nordisk A/S Stable, protracted GLP-1/glucagon receptor co-agonists for medical use
US9486506B2 (en) 2011-09-23 2016-11-08 Novo Nordisk A/S Glucagon analogues
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
JP2018115170A (en) * 2018-03-02 2018-07-26 バクスアルタ ゲーエムベーハー Factor fviii-polymer conjugates
EP3437661A1 (en) 2008-10-17 2019-02-06 Baxalta GmbH Modified blood factors comprising a low degree of water soluble polymer
US10238718B2 (en) 2014-08-04 2019-03-26 Csl Limited Factor VIII formulation
US10570184B2 (en) 2014-06-04 2020-02-25 Novo Nordisk A/S GLP-1/glucagon receptor co-agonists for medical use
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii
US11078518B2 (en) 2012-09-28 2021-08-03 Chugai Seiyaku Kabushiki Kaisha Method for evaluating blood coagulation reaction

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7179617B2 (en) * 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
EP1824988B1 (en) * 2004-11-12 2017-04-19 Bayer HealthCare LLC Site-directed modification of fviii
US7855279B2 (en) * 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US7846445B2 (en) * 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US7982010B2 (en) * 2006-03-31 2011-07-19 Baxter International Inc. Factor VIII polymer conjugates
EP2010222A1 (en) 2006-03-31 2009-01-07 Baxter International Inc. Pegylated factor viii
US7985839B2 (en) * 2006-03-31 2011-07-26 Baxter International Inc. Factor VIII polymer conjugates
WO2008074032A1 (en) 2006-12-15 2008-06-19 Baxter International Inc. Factor viia- (poly) sialic acid conjugate having prolonged in vivo half-life
WO2008154639A2 (en) 2007-06-12 2008-12-18 Neose Technologies, Inc. Improved process for the production of nucleotide sugars
US20100268055A1 (en) * 2007-07-19 2010-10-21 Arizona Board of Regents, a body corporate acting for and on behalf of Arizona State University Self-Anchoring MEMS Intrafascicular Neural Electrode
BRPI0815416A2 (en) * 2007-08-15 2014-10-21 Amunix Inc COMPOSITIONS AND METHODS FOR MODIFYING PROPERTIES OF BIOLOGICALLY ACTIVE POLYPEPTIDES
AU2014268202B2 (en) * 2008-08-01 2017-08-03 Takeda Pharmaceutical Company Limited Factor VIII Polymer Conjugates
US8680050B2 (en) * 2009-02-03 2014-03-25 Amunix Operating Inc. Growth hormone polypeptides fused to extended recombinant polypeptides and methods of making and using same
EP3178835B1 (en) 2009-02-03 2019-04-10 Amunix Pharmaceuticals, Inc. Extended recombinant polypeptides and compositions comprising same
US8703717B2 (en) * 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
TW201042257A (en) * 2009-05-26 2010-12-01 Baxter Int Detection of antibody that binds to water soluble polymer-modified polypeptides
JP5839597B2 (en) 2009-06-08 2016-01-06 アムニクス オペレーティング インコーポレイテッド Glucose-regulating polypeptide and methods for making and using the same
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US8642737B2 (en) 2010-07-26 2014-02-04 Baxter International Inc. Nucleophilic catalysts for oxime linkage
US8809501B2 (en) 2009-07-27 2014-08-19 Baxter International Inc. Nucleophilic catalysts for oxime linkage
DK2459224T3 (en) * 2009-07-27 2016-07-25 Baxalta GmbH Blodstørkningsproteinkonjugater
EP2470670A4 (en) 2009-08-24 2013-07-10 Amunix Operating Inc Coagulation factor vii compositions and methods of making and using same
BR112012013502A2 (en) * 2009-12-06 2017-01-10 Biogen Idec Hemophilia Inc "chimeric and hybrid factor viii-fc polypeptides, and methods of using them".
WO2011123830A2 (en) 2010-04-02 2011-10-06 Amunix Operating Inc. Alpha 1-antitrypsin compositions and methods of making and using same
GB201007357D0 (en) * 2010-04-30 2010-06-16 Leverton Licence Holdings Ltd Conjugated factor VIII
CA2822591C (en) 2010-12-22 2020-12-29 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
EP2714093A1 (en) * 2011-05-27 2014-04-09 Baxter International Inc. Therapeutic proteins with increased half-life and methods of preparing same
HUE046848T2 (en) 2012-02-15 2020-03-30 Bioverativ Therapeutics Inc Factor viii compositions and methods of making and using same
EP2822577B1 (en) 2012-02-15 2019-02-06 Bioverativ Therapeutics Inc. Recombinant factor viii proteins
CA3179537A1 (en) 2012-02-27 2013-09-06 Amunix Pharmaceuticals, Inc. Xten conjugate compositions and methods of making same
JP6533932B2 (en) * 2012-12-28 2019-06-26 アボット カーディオバスキュラー システムズ インコーポレイテッド Therapeutic composition comprising an antibody
AU2014237111B2 (en) * 2013-03-15 2018-06-21 Bayer Healthcare Llc Recombinant factor VIII formulations
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
JP6552286B2 (en) * 2015-06-05 2019-07-31 公立大学法人奈良県立医科大学 Method and reagent kit for obtaining information on complexing ability of coagulation factor VIII
JP6909203B2 (en) 2015-08-03 2021-07-28 バイオベラティブ セラピューティクス インコーポレイテッド Factor IX fusion proteins and their production and usage
CN108135968A (en) 2015-08-28 2018-06-08 阿穆尼克斯运营公司 Chimeric polyeptides assembly and its preparation and application
TW201936634A (en) * 2017-09-29 2019-09-16 日商中外製藥股份有限公司 Multispecific antigen-binding molecule having blood coagulation factor viii (FVIII) cofactor function-substituting activity, and pharmaceutical formulation containing said molecule as active ingredient

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4970300A (en) * 1985-02-01 1990-11-13 New York University Modified factor VIII
WO1991009122A1 (en) * 1989-12-15 1991-06-27 Kabivitrum Ab A recombinant human factor viii derivative
WO1994015625A1 (en) * 1993-01-15 1994-07-21 Enzon, Inc. Factor viii - polymeric conjugates
US6037452A (en) * 1992-04-10 2000-03-14 Alpha Therapeutic Corporation Poly(alkylene oxide)-Factor VIII or Factor IX conjugate
WO2000048635A1 (en) * 1999-02-22 2000-08-24 Baxter International Inc. Novel albumin-free factor viii formulations
EP1260582A1 (en) * 1995-09-29 2002-11-27 Biovitrum Ab Conjugates of factor IX and a biocompatible polymer
WO2004075923A2 (en) * 2003-02-26 2004-09-10 Nektar Therapeutics Al, Corporation Polymer-factor viii moiety conjugates
US20050106658A1 (en) * 2001-10-10 2005-05-19 Defrees Shawn Remodeling and glycoconjugation of peptides

Family Cites Families (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4356170A (en) * 1981-05-27 1982-10-26 Canadian Patents & Development Ltd. Immunogenic polysaccharide-protein conjugates
US4757006A (en) * 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
US4965199A (en) * 1984-04-20 1990-10-23 Genentech, Inc. Preparation of functional human factor VIII in mammalian cells using methotrexate based selection
JPS62502589A (en) 1985-04-11 1987-10-08 ザ・チヤイルドレンズ・メデイカル・センタ−・コ−ポレ−シヨン Von Willebrand factor
US5198349A (en) * 1986-01-03 1993-03-30 Genetics Institute, Inc. Method for producing factor VIII:C and analogs
US5250421A (en) * 1986-01-03 1993-10-05 Genetics Institute, Inc. Method for producing factor VIII:C-type proteins
CA1339946C (en) * 1987-03-31 1998-07-07 Michael J. Griffith Ultrapurification process for polypeptides
JPH0387173A (en) 1987-09-10 1991-04-11 Teijin Ltd Preparation of human active natural type factor viii c and transformant using the same
US5605884A (en) * 1987-10-29 1997-02-25 Rhone-Poulenc Rorer Pharmaceuticals Inc. Factor VIII formulations in high ionic strength media
US4877608A (en) * 1987-11-09 1989-10-31 Rorer Pharmaceutical Corporation Pharmaceutical plasma protein formulations in low ionic strength media
US5153265A (en) * 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US5122614A (en) * 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
DE4001451A1 (en) * 1990-01-19 1991-08-01 Octapharma Ag STABLE INJECTABLE SOLUTIONS OF FACTOR VIII AND FACTOR IX
SE466754B (en) * 1990-09-13 1992-03-30 Berol Nobel Ab COVALENT BINDING POLYMERS TO HYDROPHILIC SURFACES
CA2073511A1 (en) * 1990-11-14 1992-05-29 Matthew R. Callstrom Conjugates of poly(vinylsaccharide) with proteins for the stabilization of proteins
WO1992016555A1 (en) 1991-03-18 1992-10-01 Enzon, Inc. Hydrazine containing conjugates of polypeptides and glycopolypeptides with polymers
US5595732A (en) * 1991-03-25 1997-01-21 Hoffmann-La Roche Inc. Polyethylene-protein conjugates
DE4111393A1 (en) * 1991-04-09 1992-10-15 Behringwerke Ag STABILIZED FACTOR VIII PREPARATIONS
US5846951A (en) 1991-06-06 1998-12-08 The School Of Pharmacy, University Of London Pharmaceutical compositions
WO1993000357A1 (en) 1991-06-28 1993-01-07 Rhone-Poulenc Rorer International (Holdings) Inc. Therapeutic polypeptides based on von willebrand factor
AU5098193A (en) 1992-09-01 1994-03-29 Berlex Laboratories, Inc. Glycolation of glycosylated macromolecules
HU220194B (en) * 1992-10-02 2001-11-28 Genetics Institute Inc. Composition comprising coagulation factor viii formulation and process for its preparation
NZ250375A (en) 1992-12-09 1995-07-26 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives
US5298643A (en) * 1992-12-22 1994-03-29 Enzon, Inc. Aryl imidate activated polyalkylene oxides
FR2700268B1 (en) 1993-01-13 1995-03-31 Lvmh Rech Cosmetic or pharmaceutical composition, especially dermatological, containing an extract of Vismia.
US5621039A (en) 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
US5919455A (en) 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
DE4435485C1 (en) 1994-10-04 1996-03-21 Immuno Ag Process for obtaining high-purity von Willebrand factor
AU6255096A (en) 1995-06-07 1996-12-30 Mount Sinai School Of Medicine Of The City University Of New York, The Pegylated modified proteins
US6127153A (en) 1995-06-07 2000-10-03 Neose Technologies, Inc. Method of transferring at least two saccharide units with a polyglycosyltransferase, a polyglycosyltransferase and gene encoding a polyglycosyltransferase
US6005077A (en) 1995-11-10 1999-12-21 Immuno Aktiengesellschaft Use of von willebrand factor and pharmaceutical formulation
US6562781B1 (en) 1995-11-30 2003-05-13 Hamilton Civic Hospitals Research Development Inc. Glycosaminoglycan-antithrombin III/heparin cofactor II conjugates
US6214966B1 (en) 1996-09-26 2001-04-10 Shearwater Corporation Soluble, degradable poly(ethylene glycol) derivatives for controllable release of bound molecules into solution
AT405740B (en) 1996-12-13 1999-11-25 Immuno Ag FROM WILLEBRAND FACTOR DERIVATIVE AND A METHOD FOR ISOLATING PROTEINS
AT405403B (en) 1997-02-27 1999-08-25 Immuno Ag CLEANING OF WILLEBRAND FACTOR BY CATION EXCHANGER CHROMATOGRAPHY
CA2288994C (en) 1997-04-30 2011-07-05 Enzon, Inc. Polyalkylene oxide-modified single chain polypeptides
US6183738B1 (en) 1997-05-12 2001-02-06 Phoenix Pharamacologics, Inc. Modified arginine deiminase
AT405485B (en) 1997-05-28 1999-08-25 Immuno Ag A PHARMACEUTICAL PREPARATION CONTAINING THE VWF PROPEPTIDE
DK0985697T3 (en) 1998-03-24 2006-05-15 Nof Corp Oxirane derivatives and process for their preparation
EP2260872B1 (en) 1998-10-16 2017-08-30 Biogen MA Inc. Polymer conjugates of interferon beta-1A and uses thereof
DE19852729A1 (en) 1998-11-16 2000-05-18 Werner Reutter Recombinant glycoproteins, processes for their preparation, medicaments containing them and their use
AT407750B (en) 1999-02-19 2001-05-25 Immuno Ag METHOD FOR PRODUCING A VWF PREPARATION
WO2001005434A2 (en) 1999-07-20 2001-01-25 Amgen Inc. Hyaluronic acid-protein conjugates
US6531122B1 (en) 1999-08-27 2003-03-11 Maxygen Aps Interferon-β variants and conjugates
YU32402A (en) 1999-11-12 2005-03-15 Maxygen Holdings Ltd. Interferon gamma conjugates
SE515295C2 (en) 1999-11-23 2001-07-09 Medicarb Ab Process for the preparation of conjugates of a biologically active polysaccharide and a solid substrate, and such conjugates
JP2003521930A (en) 2000-02-11 2003-07-22 マキシゲン・エイピーエス Factor VII or Factor VIIa-like molecule
US6586398B1 (en) 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US7118737B2 (en) 2000-09-08 2006-10-10 Amylin Pharmaceuticals, Inc. Polymer-modified synthetic proteins
WO2002032957A1 (en) * 2000-10-16 2002-04-25 Chugai Seiyaku Kabushiki Kaisha Peg-modified erythropoietin
AUPR610501A0 (en) 2001-07-04 2001-07-26 Smart Drug Systems Inc Treatment of parasitic disease
US6913915B2 (en) 2001-08-02 2005-07-05 Phoenix Pharmacologics, Inc. PEG-modified uricase
US7265084B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
EP1446438A2 (en) * 2001-11-07 2004-08-18 Nektar Therapeutics Al, Corporation Branched polymers and their conjugates
US20050176108A1 (en) 2003-03-13 2005-08-11 Young-Min Kim Physiologically active polypeptide conjugate having prolonged in vivo half-life
CA2843439A1 (en) 2003-04-08 2004-10-21 Yeda Research And Development Co. Ltd Reversible pegylated drugs
WO2005032581A2 (en) 2003-10-07 2005-04-14 Novo Nordisk Health Care Ag Hybrid molecules having factor vii/viia activity
CA2549011A1 (en) 2003-12-10 2005-06-30 Nektar Therapeutics Al, Corporation Compositions comprising two different populations of polymer-active agent conjugates
KR20070092754A (en) 2004-12-27 2007-09-13 백스터 인터내셔널 인코포레이티드 Polymer-von willebrand factor-conjugates
US7645860B2 (en) 2006-03-31 2010-01-12 Baxter Healthcare S.A. Factor VIII polymer conjugates
EP2010222A1 (en) 2006-03-31 2009-01-07 Baxter International Inc. Pegylated factor viii
JP5570809B2 (en) 2006-09-01 2014-08-13 ノボ ノルディスク ヘルス ケア アーゲー Modified protein
WO2008074032A1 (en) 2006-12-15 2008-06-19 Baxter International Inc. Factor viia- (poly) sialic acid conjugate having prolonged in vivo half-life

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4970300A (en) * 1985-02-01 1990-11-13 New York University Modified factor VIII
WO1991009122A1 (en) * 1989-12-15 1991-06-27 Kabivitrum Ab A recombinant human factor viii derivative
US6037452A (en) * 1992-04-10 2000-03-14 Alpha Therapeutic Corporation Poly(alkylene oxide)-Factor VIII or Factor IX conjugate
WO1994015625A1 (en) * 1993-01-15 1994-07-21 Enzon, Inc. Factor viii - polymeric conjugates
EP1260582A1 (en) * 1995-09-29 2002-11-27 Biovitrum Ab Conjugates of factor IX and a biocompatible polymer
WO2000048635A1 (en) * 1999-02-22 2000-08-24 Baxter International Inc. Novel albumin-free factor viii formulations
US20050106658A1 (en) * 2001-10-10 2005-05-19 Defrees Shawn Remodeling and glycoconjugation of peptides
WO2004075923A2 (en) * 2003-02-26 2004-09-10 Nektar Therapeutics Al, Corporation Polymer-factor viii moiety conjugates

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Baxter Announces Collaborations to Develop Longer Acting Forms of Blood Clotting Factors", BAXTER NEWS, 29 September 2005 (2005-09-29), XP002446808, Retrieved from the Internet <URL:http://www.baxter.com/about_baxter/news_room/news_releases/2005/09-29-05-collaboration.html> *
SAENKO E L ET AL: "Strategies towards a longer acting factor VIII", HAEMOPHILIA, BLACKWELL SCIENCE, OXFORD, GB, vol. 12, no. SUPPL 3, 2006, pages 42 - 51, XP002434557, ISSN: 1351-8216 *
SAKURAGAWA N ET AL: "STUDIES ON THE STABILITY OF FACTOR VIII MODIFIED BY POLYETHYLENE GLYCOL", ACTA MEDICA BIOLOGICA, NIIGATA DAIGAKU IGAKUBU, NIIGATA,, JP, vol. 36, no. 1, 1988, pages 1 - 5, XP009011547, ISSN: 0567-7734 *
See also references of EP2010222A1 *
Severs Joanne; Samuels Nicole; Teare John; Tollner Amy; Regan Lisa "Characterization of PEGylated factor VIII molecules." Symposium of the International Society of Molecular Evolution; Guananacaste, COSTA RICA; January 08 -12, 2001, Abstract #4024. (Blood, Nov 2006 108(11-2), 84B, ISSN 0006-4971) *

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11020458B2 (en) 2006-03-31 2021-06-01 Takeda Pharmaceutical Company Limited Factor VIII polymer conjugates
WO2008116913A2 (en) * 2007-03-28 2008-10-02 Novo Nordisk A/S Peptide compounds with transient biodegradable pegylation
WO2008116913A3 (en) * 2007-03-28 2009-01-15 Novo Nordisk As Peptide compounds with transient biodegradable pegylation
US8173597B2 (en) 2007-11-09 2012-05-08 Baxter International Inc. Modified recombinant factor VIII and von Willebrand factor and methods of use
JP2011503101A (en) * 2007-11-09 2011-01-27 バクスター・インターナショナル・インコーポレイテッド Modified recombinant factor VIII and von Willebrand factor and methods of use thereof
US9547016B2 (en) 2007-12-27 2017-01-17 Baxalta Incorporated Method and compositions for specifically detecting physiologically acceptable polymer molecules
EP2416157A2 (en) 2007-12-27 2012-02-08 Baxter International Inc. Method and compositions for specifically detecting physiologically acceptable polymer molecules
US8557534B2 (en) 2007-12-27 2013-10-15 Baxter International Inc. Method and compositions for specifically detecting physiologically acceptable polymer molecules
WO2009108806A1 (en) * 2008-02-27 2009-09-03 Novo Nordisk A/S Conjugated factor viii molecules
US8536126B2 (en) 2008-02-27 2013-09-17 Novo Nordisk A/S Conjugated factor VIII molecules
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
EP2626080A3 (en) * 2008-02-27 2014-03-05 Novo Nordisk A/S Conjugated factor VIII molecules
EP2626079A3 (en) * 2008-02-27 2014-03-05 Novo Nordisk A/S Conjungated factor VIII molecules
CN101965200B (en) * 2008-02-27 2013-06-19 诺沃-诺迪斯克有限公司 Conjugated factor VIII molecules
JP2011519833A (en) * 2008-04-24 2011-07-14 セルティック ファーマ ピーイージー リミテッド Factor IX conjugates with extended half-life
US9290561B2 (en) 2008-06-24 2016-03-22 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
EP2865760A1 (en) 2008-06-24 2015-04-29 CSL Behring GmbH Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
JP2013500238A (en) * 2008-08-01 2013-01-07 バクスター・インターナショナル・インコーポレイテッド Factor FVIII polymer conjugate
JP2012505901A (en) * 2008-10-15 2012-03-08 バクスター・インターナショナル・インコーポレイテッド PEGylation of recombinant blood clotting factors in the presence of conjugated antibodies
EP3437661A1 (en) 2008-10-17 2019-02-06 Baxalta GmbH Modified blood factors comprising a low degree of water soluble polymer
EP3620177A1 (en) 2008-10-17 2020-03-11 Baxalta GmbH Modified blood factors comprising a low degree of water soluble polymer
EP2387413A4 (en) * 2009-01-19 2015-12-23 Bayer Healthcare Llc Protein conjugate having an endopeptidase-cleavable bioprotective moiety
CN102333788A (en) * 2009-02-19 2012-01-25 诺沃—诺迪斯克有限公司 Modification of factor viii
WO2010102886A1 (en) * 2009-02-19 2010-09-16 Novo Nordisk A/S Modification of factor viii
JP2013500375A (en) * 2009-07-27 2013-01-07 リポクセン テクノロジーズ リミテッド Glycopolysial oxidation of non-blood clotting proteins
JP2018024882A (en) * 2009-07-27 2018-02-15 リポクセン テクノロジーズ リミテッド Glycopolysialylation of non-blood coagulation protein
JP2016113626A (en) * 2009-07-27 2016-06-23 リポクセン テクノロジーズ リミテッド Glycopolysialylation of non-blood coagulation protein
US9018166B2 (en) 2010-02-16 2015-04-28 Novo Nordisk A/S Conjugated FVIII variants
US9309507B2 (en) 2010-04-30 2016-04-12 Polytherics Limited Conjugated blood coagulation factor VIIa
US20130183280A1 (en) * 2010-07-15 2013-07-18 Novo Nordisk A/S Stabilized factor viii variants
WO2012079979A1 (en) * 2010-12-16 2012-06-21 Novo Nordisk A/S Aqueous factor viii solution
US9486505B2 (en) 2011-09-23 2016-11-08 Novo Nordisk A/S Glucagon analogues
US9486506B2 (en) 2011-09-23 2016-11-08 Novo Nordisk A/S Glucagon analogues
US9458223B2 (en) 2012-02-15 2016-10-04 Csl Behring Gmbh Von willebrand factor variants having improved factor VIII binding affinity
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
GB2516388A (en) * 2012-04-16 2015-01-21 Cantab Biopharmaceuticals Patents Ltd Optimised subcutaneous therapeutic agents
EA033469B1 (en) * 2012-04-16 2019-10-31 Cantab Biopharmaceuticals Patents Ltd Subcutaneous administration of blood factor conjugates with polyethylene glycol
US11351112B2 (en) 2012-04-16 2022-06-07 Cantab Biopharmaceuticals Patents Limited Optimised subcutaneous therapeutic agents
WO2013156488A3 (en) * 2012-04-16 2014-01-16 Leverton Licence Holdings Limited Optimised subcutaneous therapeutic agents
US11078518B2 (en) 2012-09-28 2021-08-03 Chugai Seiyaku Kabushiki Kaisha Method for evaluating blood coagulation reaction
US9474790B2 (en) 2013-04-18 2016-10-25 Novo Nordisk A/S Stable, protracted GLP-1/glucagon receptor co-agonists for medical use
US9751927B2 (en) 2013-04-18 2017-09-05 Novo Nordisk A/S Stable, protracted GLP-1/glucagon receptor co-agonists for medical use
US9878017B2 (en) 2013-04-22 2018-01-30 Csl Ltd. Covalent complex of von Willebrand Factor and factor VIII, compositions, and uses relating thereto
WO2014173873A1 (en) 2013-04-22 2014-10-30 Csl Behring Gmbh Complex
US10570184B2 (en) 2014-06-04 2020-02-25 Novo Nordisk A/S GLP-1/glucagon receptor co-agonists for medical use
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US10253088B2 (en) 2014-07-02 2019-04-09 CSL Behring Lengnau AG Modified von Willebrand Factor
US10238718B2 (en) 2014-08-04 2019-03-26 Csl Limited Factor VIII formulation
US11155601B2 (en) 2015-03-06 2021-10-26 CSL Behring Lengnau AG Modified von Willebrand factor having improved half-life
WO2016142288A1 (en) 2015-03-06 2016-09-15 Csl Behring Recombinant Facility Ag Modified von willebrand factor having improved half-life
US10688157B2 (en) 2015-05-22 2020-06-23 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for treating hemophilia
US10905747B2 (en) 2015-05-22 2021-02-02 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
US11564976B2 (en) 2015-05-22 2023-01-31 CSL Behring Lengnau AG Methods for preparing modified von Willebrand Factor
US10772936B2 (en) 2015-05-22 2020-09-15 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
EP4089109A2 (en) 2015-05-22 2022-11-16 CSL Behring Lengnau AG Methods for preparing modified von willebrand factor
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
JP2015155469A (en) * 2015-06-01 2015-08-27 バクスター・インターナショナル・インコーポレイテッドBaxter International Incorp0Rated Fviii-polymer conjugates
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US10806774B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated truncated von Willebrand Factor
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
US11814421B2 (en) 2016-11-11 2023-11-14 CSL Behring Lengnau AG Truncated von Willebrand Factor polypeptides for treating hemophilia
US11890327B2 (en) 2016-11-11 2024-02-06 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
JP2018115170A (en) * 2018-03-02 2018-07-26 バクスアルタ ゲーエムベーハー Factor fviii-polymer conjugates
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii

Also Published As

Publication number Publication date
EP2010222A1 (en) 2009-01-07
AU2007245190B2 (en) 2011-07-21
MX2008012600A (en) 2008-12-12
AU2007245190A1 (en) 2007-11-08
US7683158B2 (en) 2010-03-23
BRPI0708832A2 (en) 2011-06-14
US20070244301A1 (en) 2007-10-18
KR20080108147A (en) 2008-12-11
US8053561B2 (en) 2011-11-08
CA2647314A1 (en) 2007-11-08
US20100168391A1 (en) 2010-07-01
NZ572050A (en) 2011-09-30
JP2009532351A (en) 2009-09-10
NO20084608L (en) 2008-12-30

Similar Documents

Publication Publication Date Title
AU2007245190B2 (en) Pegylated factor VIII
AU2009276625B2 (en) Factor VIII polymer conjugates
US7985839B2 (en) Factor VIII polymer conjugates
US7982010B2 (en) Factor VIII polymer conjugates
AU2019222949B2 (en) Factor VIII polymer conjugates
JP2015155469A (en) Fviii-polymer conjugates
JP2018115170A (en) Factor fviii-polymer conjugates

Legal Events

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

Ref document number: 07754130

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2007754130

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2647314

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2009502944

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/012600

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 200780012460.4

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2007245190

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 572050

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1020087026600

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2007245190

Country of ref document: AU

Date of ref document: 20070329

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: PI0708832

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080930