WO2013009627A2 - Factor viii chimeric and hybrid polypeptides, and methods of use thereof - Google Patents

Factor viii chimeric and hybrid polypeptides, and methods of use thereof Download PDF

Info

Publication number
WO2013009627A2
WO2013009627A2 PCT/US2012/045784 US2012045784W WO2013009627A2 WO 2013009627 A2 WO2013009627 A2 WO 2013009627A2 US 2012045784 W US2012045784 W US 2012045784W WO 2013009627 A2 WO2013009627 A2 WO 2013009627A2
Authority
WO
WIPO (PCT)
Prior art keywords
factor viii
subject
chimeric polypeptide
seq
polypeptide
Prior art date
Application number
PCT/US2012/045784
Other languages
French (fr)
Other versions
WO2013009627A3 (en
Inventor
Jennifer A. Dumont
Susan Low
Alan J. Bitonti
Glenn Pierce
Alvin Luk
Haiyan Jiang
Byron Mckinney
Matt Ottmer
Jurg Sommer
Karen Nugent
Lian Li
Robert Peters
Original Assignee
Biogen Idec Hemophilia Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to PL18211156T priority Critical patent/PL3513804T3/en
Priority to KR1020187003966A priority patent/KR102110736B1/en
Priority to JP2014519074A priority patent/JP2014522838A/en
Application filed by Biogen Idec Hemophilia Inc. filed Critical Biogen Idec Hemophilia Inc.
Priority to PL12811808T priority patent/PL2729161T3/en
Priority to CN201280043194.2A priority patent/CN103796670A/en
Priority to SI201231567T priority patent/SI2729161T1/en
Priority to NZ619438A priority patent/NZ619438B2/en
Priority to AU2012282875A priority patent/AU2012282875B2/en
Priority to RS20190355A priority patent/RS58578B1/en
Priority to KR1020147003130A priority patent/KR101829603B1/en
Priority to US14/131,600 priority patent/US10010622B2/en
Priority to BR112014000466A priority patent/BR112014000466A2/en
Priority to DK12811808.0T priority patent/DK2729161T3/en
Priority to EP18211156.7A priority patent/EP3513804B1/en
Priority to CA2841066A priority patent/CA2841066C/en
Priority to EA201490039A priority patent/EA029045B1/en
Priority to EP12811808.0A priority patent/EP2729161B1/en
Priority to ES12811808T priority patent/ES2722209T3/en
Priority to MYPI2014000038A priority patent/MY180714A/en
Priority to MX2014000202A priority patent/MX350581B/en
Priority to EP22163636.8A priority patent/EP4169525A1/en
Priority to LTEP12811808.0T priority patent/LT2729161T/en
Publication of WO2013009627A2 publication Critical patent/WO2013009627A2/en
Publication of WO2013009627A3 publication Critical patent/WO2013009627A3/en
Priority to IL230333A priority patent/IL230333B/en
Priority to AU2016204986A priority patent/AU2016204986B2/en
Priority to AU2018202936A priority patent/AU2018202936B2/en
Priority to US15/991,629 priority patent/US10881742B2/en
Priority to HRP20190535TT priority patent/HRP20190535T1/en
Priority to CY20191100325T priority patent/CY1122900T1/en
Priority to IL271870A priority patent/IL271870B/en
Priority to US17/112,280 priority patent/US20210220476A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • A61K38/37Factors VIII
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody 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
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • 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
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • 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
    • C07K14/755Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/86Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving blood coagulating time or factors, or their receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the present invention relates generally to the field of therapeutics for hemostatic disorders.
  • Hemophilia A is an X-linked bleeding disorder caused by mutations and/or deletions in the factor VIII (FVIII) gene resulting in a deficiency of FVIII activity (Peyvandi, F. et al. Haemophilia 72:82-89 (2006).
  • the disease is characterized by spontaneous hemorrhage and excessive bleeding after trauma. Over time, the repeated bleeding into muscles and joints, which often begins in early childhood, results in hemophilic arthropathy and irreversible joint damage. This damage is progressive and can lead to severely limited mobility of joints, muscle atrophy and chronic pain (Rodriguez-Merchan, E.C., Semin. Thromb. Hemost. 29:87-96 (2003), which is herein incorporated by reference in its entirety).
  • the A2 domain is necessary for the procoagulant activity of the factor VIII molecule.
  • porcine factor VIII has six-fold greater procoagulant activity than human factor VIII (Lollar, P., and E. T. Parker, J. Biol. Chem. 266: 12481-12486 (1991)), and that the difference in coagulant activity between human and porcine factor VIII appears to be based on a difference in amino acid sequence between one or more residues in the human and porcine A2 domains (Lollar, P., et al., J. Biol. Chem. 267:23652-23657 (1992)), incorporated herein by reference in its entirety.
  • Treatment of hemophilia A is by replacement therapy targeting restoration of FVIII activity to 1 to 5 % of normal levels to prevent spontaneous bleeding (Mannucci, P.M., et al, N. Engl. J. Med. 344:1773-1779 (2001), which is herein incorporated by reference in its entirety).
  • FVIII products available to treat bleeding episodes on- demand or to prevent bleeding episodes from occurring by treating prophylactically. Based on the short half-life of these products, however, e.g., 8-12 hours, treatment regimens require the administration of frequent intravenous injections. Such frequent administration is painful and inconvenient.
  • the present invention provides methods of administering Factor VIII; methods of administering chimeric polypeptides comprising Factor VIII and hybrids of such chimeric polypeptides; chimeric polypeptides comprising Factor VIII and hybrids of such chimeric polypeptides; polynucleotides encoding such chimeric and hybrid polypeptides; cells comprising such polynucleotides; and methods of producing such chimeric and hybrid polypeptides using such cells.
  • the present invention provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, at a dosing interval at least about one and one -half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • a chimeric Factor VIII polypeptide e.g., a chimeric Factor VIII-Fc polypeptide
  • the dosing interval may be at least about one and one -half to six times longer, one and one -half to five times longer, one and one-half to four times longer, one and one-half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., the Fc portion.
  • the dosing interval may be at least about one and one -half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one-half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., the Fc portion.
  • the dosing interval may be about every five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
  • the dosing interval may be at least about one and one -half to 5, one and one-half, 2, 3, 4, or 5 days or longer.
  • the present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, to obtain an area under the plasma concentration versus time curve (AUC) at least about one and one-quarter times greater than the AUC obtained by an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • AUC area under the plasma concentration versus time curve
  • the present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a polypeptide comprising a Factor VIII and an Fc at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
  • the methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
  • On-demand treatment includes treatment for a bleeding episode, hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis (head trauma), gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath.
  • the subject may be in need of surgical prophylaxis, peri-operative management, or treatment for surgery.
  • Such surgeries include, e.g., minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
  • the dosing interval of said chimeric polypeptide is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
  • the therapeutic doses that may be used in the methods of the invention are about 10 to about 100 IU/kg, more specifically, about 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 IU/kg, and more specifically, about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
  • the therapeutic doses that may be used in the methods of the invention are about 10 to about 150 IU/kg, more specifically, about 100-110, 110-120, 120-130, 130-140, 140-150 IU/kg, and more specifically, about 110, 115, 120, 125, 130, 135, 140, 145, or 150 IU/kg.
  • the subject in the methods of the invention is a human subject.
  • the determination of dosing interval and AUC may be carried out in a single subject or in a population of subjects.
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) is a human Factor VIII.
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may have a full or partial deletion of the B domain.
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90% or
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90%> or
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
  • the Fc portion (or Fc portion of a chimeric polypeptide) may be at least 90%> or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
  • the Fc portion (or Fc portion of a chimeric polypeptide) may be identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
  • the chimeric polypeptide may comprise a sequence at least 90% or 95% identical to the
  • the chimeric polypeptide may comprise a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
  • the chimeric polypeptide may be in the form of a hybrid comprising a second polypeptide in association with said chimeric polypeptide, wherein said second polypeptide comprises or consists essentially of an Fc.
  • the second polypeptide may comprise or consist essentially of a sequence at least 90% or
  • the second polypeptide may comprise or consist essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
  • the chimeric polypeptide or hybrid may be administered as part of a pharmaceutical composition comprising at least one excipient.
  • the invention also provides the above-described chimeric and hybrid polypeptides themselves, polynucleotides encoding them, a cultured human embryonic cells comprising the polynucleotides, and methods of producing such chimeric and hybrid polypeptides, and the polypeptides produced by such methods.
  • the present invention also provide a chimeric polypeptide that has Factor VIII activity comprising a Factor VIII portion and a second portion, wherein the Factor VIII portion is processed Factor VIII comprising two chains, a first chain comprising a heavy chain and a second chain comprising a light chain, wherein said first chain and said second chain are associated by a metal bond.
  • Factor VIII activity comprising a Factor VIII portion and a second portion
  • the Factor VIII portion is processed Factor VIII comprising two chains, a first chain comprising a heavy chain and a second chain comprising a light chain, wherein said first chain and said second chain are associated by a metal bond.
  • Factor VIII portion is processed Factor VIII comprising two chains, a first chain comprising a heavy chain and a second chain comprising a light chain, wherein said first chain and said second chain are associated by a metal bond.
  • the present invention includes a chimeric polypeptide that has Factor VIII activity, wherein the Factor VIII portion is single chain Factor VIII.
  • the single chain Factor VIII can contain an intact intracellular processing site. In one embodiment, at least about 1%), about 5%, about 10%, about 15%, about 20%, or about 25% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII. In another embodiment, at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 99%) of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII. In another aspect, the single chain FVIII does not contain an intracellular processing site.
  • the SCFVIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
  • the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648.
  • the substitution or mutation is an amino acid other than arginine, e.g., alanine.
  • the chimeric polypeptide comprising single chain Factor VIII has
  • Factor VIII activity at a level comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions, when the Factor VIII activity is measured in vitro by a chromogenic assay.
  • the chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity in vivo comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
  • the chimeric polypeptide comprising single chain Factor VIII has a Factor Xa generation rate comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
  • single chain Factor VIII in the chimeric polypeptide is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
  • the single chain Factor VIII in the chimeric polypeptide has a Factor IXa interaction rate comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
  • the single chain Factor VIII in the chimeric polypeptide binds to von Willebrand Factor at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and the processed Factor VIII.
  • the present invention further includes a composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%>, about 50%>, about 60%>, about 70%, about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%) of said polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion, wherein said single chain Factor VIII is at least 90%> or 95%> identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
  • the second portion can be an Fc.
  • the polypeptide is in the form of a hybrid comprising a second polypeptide, wherein said second polypeptide consists essentially of an Fc.
  • the polypeptide has a half-life at least one and one -half to six times longer, one and one -half to five times longer, one and one-half to four times longer, one and one -half to three times longer, or one and one-half to two times longer to a polypeptide consisting of said Factor VIII.
  • a method of treating a bleeding condition comprising administering a therapeutically effective amount of the composition.
  • the treatment can be prophylactic treatment or on-demand treatment or perioperative.
  • the bleeding coagulation disorder can be hemophilia.
  • the subject that is treated is a pediatric subject.
  • the present invention is also directed to a method of preventing, decreasing, or treating a bleeding episode in a subject comprising administering to the subject an effective amount of a long-acting Factor VIII (FVIII) protein, wherein the subject expresses a high level of von Willebrand Factor (VWF) in plasma.
  • VWF von Willebrand Factor
  • the subject has been identified as expressing a high level of VWF in plasma.
  • the present invention is also directed to a method of preventing, decreasing, or treating a bleeding episode in a subject comprising: (a) identifying a subject having high levels of VWF by measuring the level of VWF in the plasma of said subject, wherein a VWF level of at least about 100 IU/dL identifies the subject as having a high level of VWF; and (b) administering to the subject an effective amount of a long-acting FVIII protein.
  • the subject is a human. In another embodiment, the subject is a pediatric subject. In another embodiment, the subject has hemophilia A.
  • the high level of VWF is at least about 100 IU/dL. In another embodiment, the high level of VWF is between about 100 IU/dL and about 200 IU/dL. In another embodiment, the high level of VWF is about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
  • the subject has the blood serotype A, B, or AB.
  • the long-acting FVIII protein has a half-life in said subject of between about 20 and about 40 hours. In another embodiment, the long-acting FVIII protein has a half-life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours. In another embodiment, the long-acting FVIII protein has a half-life of between about 20 and 27 hours.
  • the long-acting FVIII protein has a half- life that is at least about 1.2 times greater than the half- life of said said long- acting FVIII protein when administered to an individual having average levels of VWF. In another embodiment, the long-acting FVIII protein has a half-life that is at least about about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold times greater than the half-life of said said long-acting FVIII protein when administered to an individual having average levels of VWF.
  • the effective amount of long-acting FVIII protein that is administered is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg.
  • the effective amount is at least about 65IU/kg to at least about 90IU/kg.
  • the effective amount is 80 IU/kg.
  • the long-acting FVIII protein is administered every 72 hours or longer. In another embodiment, the long-acting FVIII protein is administered about once a week or longer. In another embodiment, the long-acting FVIII protein is administered about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month. [0040] In one embodiment, the long-acting FVIII is administered at a dosage of 80 IU/kg once every 72 hours. In a further embodiment the long-acting FVIII is administered at a dosage of 80 IU/kg once every 72 hours to a pediatric subject.
  • administration of the long-acting FVIII protein resolves greater than
  • the administration prevents a bleeding episode in the subject.
  • the bleeding episode is spontaneous.
  • the administration resolves greater than 80-100%), greater than 80-90%), greater than 85-90%, greater than 90-100%, greater than 90-95%, or greater than 95-100% of bleeding episodes.
  • the administration maintains homeostatis in the population of the subjects in need of a surgery.
  • the long-acting FVIII protein is administered prior to, during, or after the surgery.
  • the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intraabdominal surgery, intrathoracic surgery, or joint replacement surgery.
  • the surgery is an emergency surgery.
  • the long-acting FVIII protein has a half-life longer than a polypeptide consisting of FVIII.
  • the long-acting FVIII protein is pegylated, hesylated, or polysialylated.
  • the long-acting FVIII protein is a chimeric protein comprising a
  • the second portion is an Fc region, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof.
  • the second portion is fused to the amino-terminus or the carboxy-terminus of the FVIII portion.
  • the second portion is inserted between two amino acids in the FVIII portion.
  • the chimeric protein is a FVIIIFc monomer dimer hybrid.
  • the FVIII portion is a single chain. In another embodiment, the FVIII portion comprises a heavy chain and a light chain. In another embodiment, the FVIII portion comprises full-length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain. In another embodiment, the FVIII portion comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In another embodiment, the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6.
  • the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
  • the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
  • the long-acting FVIII polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
  • the invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising measuring the half-life of FVIII-Fc in said subject, wherein a half-life that is at least about 1.2 times greater than the half- life of FVIII-Fc in a normal subject indicates the subject is a candidate for long interval dosing, and administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days.
  • the invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days to a subject, wherein the half-life of FVIII-Fc in said subject is at least about 1.2 times greater than the half- life of FVIII-Fc when administered to a subject having average levels of VWF.
  • the plasma half-life of FVIII-Fc in said subject is at least about 1.3
  • the FVIII-Fc plasma half-life is between 20-40 hours.
  • the long-acting FVIII protein has a half- life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours.
  • the long-acting FVIII protein has a half- life of between about 20 and 27 hours.
  • the invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising measuring the half- life of a short-acting FVIII administered to said subject, wherein a half-life that is at least about 1.2 times greater than the half-life of said short-acting FVIII in a subject having average VWF levels indicates that the subject is a candidate for long interval dosing, and administering a long-acting FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days.
  • the half-life of the short-acting FVIII in said subject is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold greater than the half-life of a short-acting FVIII when administered to a subject having average levels of VWF.
  • the subject is a human. In another embodiment, the subject is a pediatric subject. In another embodiment, the subject has hemophilia A. In another embodiment, the subject has the blood serotype A, B, or AB. [0050] In one embodiment, the long-acting FVIII-Fc is administered in an effective amount that is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg. In another embodiment, the effective amount is at least about 65IU/kg to at least about 90IU/kg.
  • the effective amount of the FVIII-Fc protein is administered about once every week, about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
  • the administration resolves greater than 5-20%, greater than 5-15%, greater than 5-10%, greater than 10-20%), or greater than 10-15%) of bleeding episodes.
  • the trough level of plasma Factor VIILC in the subjects is maintained above 1-3 or 3-5 IU/dl.
  • the administration prevents a bleeding episode in the subject.
  • the bleeding episode is spontaneous.
  • the administration resolves greater than 80-100%, greater than 80-90%, greater than 85-90%, greater than 90-100%, greater than 90-95%), or greater than 95-100%) of bleeding episodes.
  • the administration maintains homeostatis in the population of the subjects in need of a surgery.
  • the FVIII-Fc protein is administered prior to, during, or after the surgery.
  • the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
  • the surgery is an emergency surgery.
  • the FVIII-Fc protein has a half-life longer than a polypeptide consisting of FVIII.
  • the FVIII-Fc protein is pegylated, hesylated, or polysialylated.
  • the FVIII-Fc protein is a FVIIIFc monomer dimer hybrid.
  • the FVIII portion is a single chain.
  • the FVIII portion comprises a heavy chain and a light chain.
  • the FVIII portion comprises full- length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain.
  • the FVIII portion comprises an amino acid sequence at least 80%>, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In one embodiment, the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In one embodiment, the second portion of the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6. In one embodiment, the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
  • the FVIII-Fc polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
  • the invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing with a long-acting FVIII polypeptide, comprising measuring the expression levels of plasma VWF, wherein an VWF expression level of at least 100 IU/dL indicates that the subject is a candidate for long interval dosing using a long- acting FVIII polypeptide.
  • the VWF expression level is at least about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
  • the invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half- life of FVIII-Fc in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of FVIII-Fc when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing.
  • the half-life of FVIII-Fc is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2,
  • the invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half-life of short-acting FVIII in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of short-acting FVIII when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing.
  • the half-life is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3,
  • FIG. 1 Schematic Representation of rFVIIIFc monomer.
  • FIGS. 2A-E A-B. Non-reducing and reducing SDS-PAGE analysis of rFVIIIFc
  • C rFVIIIFc structure analyzed by LC/UV and LC/MS.
  • D Total Ion Current (TIC) chromatogram (LC/MS map) of rFVIIIFc after thrombin cleavage. Major digestion products are indicated.
  • E Deconvoluted Mass Spectrum of the A2 domain of rFVIIIFc and rBDDFVIII. Major products and their cognate masses are indicated, corresponding to thrombin- cleaved A2 domain (S373 to R740) and two truncated products, S373 to Y729 and S373 to E720.
  • FIGS. 3A-C Biochemical characterization of rFVIII-Fc: A. Activation of Factor X as a function of phospholipid vesicle concentration; B. Activation of Factor X as a function of FX concentration. C. Activation of Factor X as a function of Factor IXa concentration.
  • FIG. Activation of Factor X following cleavage by activated Protein C.
  • FIGS. 5A-D Observed group mean FVIII activity ( ⁇ SE) versus time profiles, sorted by dose level, grouped by compound (one stage assay, 25 IU/kg (A) or 65 IU/kg (B); and chromogenic assay, 25 IU/kg (C) or 65 IU/kg (D)) versus time.
  • FIGS. 6A-B Observed group mean FVIII activity ( ⁇ SE) versus time profiles, grouped by dose level and compound (one stage assay (A) or chromogenic assay (B)) versus time.
  • A Single chain rFVIILFc doses are shown as squares, and processed rFVIILFc doses are shown as circles.
  • B Percent survival following tail vein transection of 4.6 ⁇ g/kg, 1.38 ⁇ g/kg, and 0.46 ⁇ g/kg of rFVIIIFc or SC rFVIIIFc.
  • C Percent of non-bleeders following tail vein transection of 4.6 ⁇ g/kg (black circle or inverted triangle), 1.38 ⁇ g/kg (triangle or diamond), and 0.46 ⁇ g/kg (square and gray circle) of rFVIIIFc or SC rFVIIIFc, respectively.
  • FIG. 8 Study Design. Fig. 8 depicts the study design of the phase l/2a study, which was a dose-escalation, sequential design to evaluate the safety and PK of rFVIIIFc compared with AD V ATE ® after a single intravenous dose of either 25 IU/kg (low dose Cohort A) or 65 IU/kg (high dose Cohort B).
  • FIG. 9 Correlation of rFVIII Activity by One-Stage (aPTT) and Chromogenic Assays.
  • FIGS. 10(A)-(B). Group Mean Plasma FVIII Activity Pharmacokinetic Profiles for Low-
  • FIGS. 12(A)-(B). Ex Vivo Whole Blood ROTEM ® Results for Individual Subjects After
  • Activity comparison in thrombin generation assay TGA.
  • SC rFVIIIFc showed a reduced endogenous thrombin potential (ETP), and
  • ETP endogenous thrombin potential
  • B a reduced peak thrombin compared to rFVIIIFc.
  • FIGS. 14(A)-(C) In vitro ROTEM data.
  • ROTEM NATEM
  • results Mean ⁇ SD
  • CFT clot formation time
  • FIGS. 15(A)-(C). Ex vivo ROTEM data.
  • ROTEM NATEM
  • results Mean ⁇ SD
  • mice following a single intravenous administration of 50 IU/kg of XYNTHA, ADVATE, or rFVIIIFc at 5 min, 24, 48, 72, and 96 hours after dosing.
  • A Average clot time (CT)
  • B Clot formation time (CFT)
  • C alpha angle.
  • FIGS. 16(A)-(E) Real-time evaluation of the interaction of rFVIIIFc and single chain
  • the x-axis shows time in seconds and the y-axis shows response in response units (RU).
  • Individual lines indicate response at different ⁇ -thrombin concentrations. The uppermost line is the response at 0 U/mL a- thrombin, and each subsequent line runs in order for ⁇ -thrombin concentrations of 0.005, 0.01 , 0.02, 0.04, 0.08, 0.16, 0.31 , 0.63, 1.3, 2.5, 5.0, 10, and 20 U/mL.
  • D Double reference subtracted sensograms of thrombin mediated release phase for rFVIIIFc, SC rFVIIIFc, and rBDD FVIII at 25° C (top) and 37° C (bottom).
  • the x-axis shows time in seconds and the
  • the x-axis shows time in seconds and the y-axis shows response in response units (RU).
  • Individual lines indicate response at different ⁇ -thrombin concentrations. The uppermost line is the response at 20 U/mL a-thrombin, and each subsequent line runs in order for a-thrombin concentrations of 10, 5, 2.5, 1.3, 0.63, 0.31, 0.16, 0.08, 0.04, 0.02, 0.01, and 0.005 U/mL. (E).
  • EC 50 is half maximal effective concentration.
  • the x-axis is ⁇ -thrombin concentration in U/mL and the y-axis is maximum release rate in RU/second.
  • the present invention provides a method of treating Hemophilia A with Factor VIII
  • the present invention also provides improved Factor VIII chimeric polypeptides and methods of production.
  • Treatment of hemophilia A is by replacement therapy targeting restoration of FVIII activity to 1 to 5 % of normal levels to prevent spontaneous bleeding (Mannucci, P.M. et al, N. Engl. J. Med. 344: 177 -9 (2001), herein incorporated by reference in its entirety).
  • FVIII products available to treat bleeding episodes on-demand or to prevent bleeding episodes from occurring by treating prophylactically. Based on the short half-life of these products (8-12 hr) (White G.C., et al, Thromb. Haemost.
  • treatment regimens require frequent intravenous administration, commonly two to three times weekly for prophylaxis and one to three times daily for on-demand treatment (Manco-Johnson, M.J., et al, N. Engl. J. Med. 357:535-544 (2007)), each of which is incorporated herein by reference in its entirety. Such frequent administration is painful and inconvenient.
  • the present invention provides a method of administering Factor VIII to a human subject in need thereof (e.g., human patient), comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, or a hybrid of such a polypeptide at a dosing interval at least about one and one-half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • the present invention is also directed to a method of increasing dosing interval of Factor VIII administration in a human subject in need thereof comprising administering the chimeric Factor VIII polypeptide.
  • the dosing interval may be at least about one and one -half to six times longer, one and one -half to five times longer, one and one -half to four times longer, one and one -half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • the dosing interval may be at least about one and one -half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one -half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • the dosing interval may be about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
  • the dosing interval may be at least about one and one-half to 5, one and one -half, 2, 3, 4, or 5 days or longer.
  • the present invention also provides a method of administering Factor VIII to a human subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, or a hybrid of such a polypeptide to obtain an area under the plasma concentration versus time curve (AUC) at least about one and one-quarter times greater than the AUC obtained by an equivalent dose of said Factor VIII without non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
  • the present invention thus includes a method of increasing or extending AUC of Factor VIII activity in a human patient in need thereof comprising administering the chimeric Factor VIII polypeptide.
  • the present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a polypeptide comprising a Factor VIII and an Fc or a hybrid of such a polypeptide at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
  • the methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
  • administering means to give a pharmaceutically acceptable Factor VIII polypeptide of the invention to a subject via a pharmaceutically acceptable route.
  • Routes of administration can be intravenous, e.g., intravenous injection and intravenous infusion. Additional routes of administration include, e.g., subcutaneous, intramuscular, oral, nasal, and pulmonary administration.
  • Chimeric polypeptides and hybrid proteins may be administered as part of a pharmaceutical composition comprising at least one excipient.
  • AUC rea under the plasma concentration versus time curve
  • AUC is the same as the term of art in pharmacology, and is based upon the rate and extent of absorption of Factor VIII following administration. AUC is determined over a specified time period, such as 12, 18, 24, 36, 48, or 72 hours, or for infinity using extrapolation based on the slope of the curve. Unless otherwise specified herein, AUC is determined for infinity. The determination of AUC may be carried out in a single subject, or in a population of subjects for which the average is calculated.
  • a "B domain" of Factor VIII is the same as the B domain known in the art that is defined by internal amino acid sequence identity and sites of proteolytic cleavage by thrombin, e.g., residues Ser741-Argl648 of full length human factor VIII.
  • the other human factor VIII domains are defined by the following amino acid residues: Al, residues Alal-Arg372; A2, residues Ser373-Arg740; A3, residues Serl690-Ile2032; CI, residues Arg2033-Asn2172; C2, residues Ser2173-Tyr2332.
  • the A3-C1-C2 sequence includes residues Serl690-Tyr2332.
  • the remaining sequence, residues Glul649-Argl689, is usually referred to as the factor VIII light chain activation peptide.
  • the locations of the boundaries for all of the domains, including the B domains, for porcine, mouse and canine factor VIII are also known in the art.
  • the B domain of Factor VIII is deleted ("B domain deleted factor VIII" or "BDD FVIII”).
  • BDD FVIII is REFACTO ® (recombinant BDD FVIII), which has the same sequence as the Factor VIII portion of the sequence in Table 2A(i) (amino acids 1 to 1457 or 20 to 1457 of SEQ ID NO:2).
  • the B domain deleted Factor VIII contains an intact intracellular processing site, which corresponds to Arginine at residue 754 of B domain deleted Factor VIII, which corresponds to Arginine residue 773 of SEQ ID NO: 2, or residue 1648 of full- length Factor VIII, which corresponds to Arginine residue 1667 of SEQ ID NO: 6.
  • sequence residue numbers used herein without referring to any SEQ ID Numbers correspond to the Factor VIII sequence without the signal peptide sequence (19 amino acids) unless otherwise indicated.
  • S743/Q1638 of full-length Factor VIII corresponds to S762/Q1657 of SEQ ID NO: 6 due to the 19 amino acid signal peptide sequence.
  • the B domain deleted FVIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
  • the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648.
  • the substitution or mutation is an amino acid other than arginine, e.g., alanine.
  • a "B domain deleted factor VIII" may have the full or partial deletions disclosed in U.S.
  • a B domain deleted factor VIII sequence of the present invention comprises any one of the deletions disclosed at col. 4, line 4 to col. 5, line 28 and examples 1-5 of U.S. Patent No. 6,316,226 (also in US 6,346,513).
  • a B domain deleted factor VIII of the present invention has a deletion disclosed at col.
  • a B domain deleted factor VIII has a deletion described in col. 1, lines 25 to col. 2, line 40 of US Patent No. 5,972,885; col. 6, lines 1-22 and example 1 of U.S. Patent no. 6,048,720; col. 2, lines 17-46 of U.S. Patent No. 5,543,502; col. 4, line 22 to col. 5, line 36 of U.S. Patent no. 5,171,844; col. 2, lines 55-68, figure 2, and example 1 of U.S. Patent No. 5,112,950; col. 2, line 2 to col.
  • a B domain deleted factor VIII has a deletion of most of the B domain, but still contains amino- terminal sequences of the B domain that are essential for in vivo proteolytic processing of the primary translation product into two polypeptide chain (i.e., intracellular processing site), as disclosed in WO 91/09122, which is incorporated herein by reference in its entirety.
  • a B domain deleted factor VIII is constructed with a deletion of amino acids 747- 1638, i.e., virtually a complete deletion of the B domain. Hoeben R.C., et al. J. Biol. Chem. 265 (13): 7318-7323 (1990), incorporated herein by reference in its entirety.
  • a B domain deleted factor VIII may also contain a deletion of amino acids 771-1666 or amino acids 868-1562 of factor VIII.
  • Additional B domain deletions that are part of the invention include, e.g.,: deletion of amino acids 982 through 1562 or 760 through 1639 (Toole et al., Proc. Natl. Acad. Sci. U.S.A. 53:5939-5942 (1986)), 797 through 1562 (Eaton et al, Biochemistry 25:8343-8347 (1986)), 741 through 1646 (Kaufman (PCT published application No.
  • the B domain deleted Factor VIII portion in the chimeric polypeptide is processed into two chains connected (or associated) by a metal bond, the first chain comprising a heavy chain (Al -A2 -partial B) and a second chain comprising a light chain (A3-C1 -C2) .
  • the B domain deleted Factor VIII portion is a single chain Factor VIII.
  • the single chain Factor VIII can comprise an intracellular processing site, which corresponds to Arginine at residue 754 of B domain deleted Factor VIII (residue 773 of SEQ ID NO: 2) or at residue 1648 of full-length Factor VIII (residue 1657 of SEQ ID NO: 6).
  • the metal bond between the heavy chain and the light chain can be any metal known in the art.
  • the metals useful for the invention can be a divalent metal ion.
  • the metals that can be used to associate the heavy chain and light chain include, but not limited to, Ca 2+ , Mn , or Cu . Fatouros et ah, Intern. J. Pharm. 155(1): 121-131 (1997); Wakabayashi et ah, JBC. 279(13): 12677-12684 (2004).
  • Chimeric polypeptide means a polypeptide that includes within it at least two polypeptides (or subsequences or peptides) from different sources.
  • Chimeric polypeptides may include, e.g., two, three, four, five, six, seven, or more polypeptides from different sources, such as different genes, different cDNAs, or different animal or other species.
  • Chimeric polypeptides may include, e.g., one or more linkers joining the different subsequences.
  • the subsequences may be joined directly or they may be joined indirectly, via linkers, or both, within a single chimeric polypeptide.
  • Chimeric polypeptides may include, e.g., additional peptides such as signal sequences and sequences such as 6His and FLAG that aid in protein purification or detection.
  • chimeric polypeptides may have amino acid or peptide additions to the N- and/or C-termini.
  • the chimeric polypeptide comprises a Factor VIII portion and a non-Factor VIII portion.
  • Exemplary non-Factor VIII portions include, e.g., Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of human chorionic gonadotropin (hCG) with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, and albumin-binding small molecules.
  • Exemplary chimeric polypeptides of the invention include, e.g., chimeric Factor VIII-Fc polypeptides, chimeric Factor VIII-XTEN polypeptides, chimeric Factor Vlll-albumin polypeptides, chimeric Factor VIII-PAS polypeptides, chimeric Factor Vlll-transferrin polypeptides, chimeric Factor VIII-CTP polypeptides, chimeric Factor VIII-PEG polypeptides, chimeric Factor VIII-HES polypeptides, chimeric Facotr Vlll-albumbin binding polypeptide polypeptides, and chimeric Factor VIII.- albumin-binding small molecule polypeptides.
  • Exemplary chimeric Factor VIII-Fc polypeptides include, e.g., SEQ ID NO:2 or 6 (Table
  • the chimeric polypeptide may comprise a sequence at least 90% or 95% identical to the
  • the Factor VIII activity can be measured by activated Partial Thromboplastin Time (aPPT) assay, chromogenic assay, or other known methods.
  • the chimeric polypeptide may comprise a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
  • exemplary chimeric polypeptides include Factor VIII fused to one or more XTEN polypeptides. Schellenburger et al, Nat. Biotech. 27: 1186-90 (2009), which is incorporated herein by reference in its entirety.
  • the XTEN polypeptide can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII.
  • a protease site may be included between the XTEN portion and the Factor VIII portion to allow such processing.
  • XTEN polypeptides include, e.g., those disclosed in WO 2009/023270, WO 2010/091122, WO 2007/103515, US 2010/0189682, and US 2009/0092582, each of which is incorporated herein by reference in its entirety.
  • exemplary chimeric polypeptides also include Factor VIII fused to one or more albumin polypeptides, albumin binding polypeptides, or albumin-binding small molecules.
  • the albumin is human albumin.
  • the albumin or albumin binding protein can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII.
  • albumin e.g., fragments thereof, that may be used in the present invention are known, e.g., U.S. Patent No. 7,592,010; U.S. Patent No. 6,686,179; and Schulte, Thrombosis Res. 124 Suppl. 2:S6-S8 (2009), each of which is incorporated herein by reference in its entirety.
  • the albumin binding polypeptides can compromise, without limitation, bacterial albumin- binding domains, albumin-binding peptides, or albumin-binding antibody fragments that can bind to albumin.
  • Domain 3 from streptococcal protein G, as disclosed by Kraulis et al, FEBS Lett. 378: 190-194 (1996) and Linhult et al, Protein Sci. 11 :206-213 (2002) is an example of a bacterial albumin-binding domain.
  • albumin-binding peptides include a series of peptides having the core sequence DICLPRWGCLW (SEQ ID NO: 7). See, e,g., Dennis et al., J. Biol. Chem.
  • albumin-binding antibody fragments are disclosed in Muller and Kontermann, Curr. Opin. Mol. Ther. 9:319-326 (2007); Rooverset et al, Cancer Immunol. Immunother. 56:303-317 (2007), and Holt et al., Prot. Eng. Design Sci., 21 :283-288 (2008), which are incorporated herein by reference in their entireties..
  • a recombinant FVIII polypeptide of the invention comprises at least one attachment site for a non-polypeptide small molecule, variant, or derivative that can bind to albumin thereof.
  • albumin binding moieties is 2-(3-maleimidopropanamido)- 6-(4-(4-iodophenyl)butanamido)hexanoate ("Albu" tag) as disclosed by Trusselet et al, Bioconjugate Chem. 20:2286-2292 (2009).
  • exemplary chimeric polypeptides also include Factor VIII fused to at least one ⁇ subunit of the C-terminal peptide (CTP) of human chorionic gonadotropin or fragment, variant, or derivative thereof.
  • CTP C-terminal peptide
  • the CTP can be fused to Factor VIII either the N- terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII.
  • One or more CTP peptides fused to or inserted into a recombinant protein is known to increase the in vivo half-life of that protein. See, e.g., U.S. Patent No. 5,712,122, incorporated by reference herein in its entirety.
  • Exemplary CTP peptides include
  • SSSSKAPPPSLPSPSRLPGPSDTPILPQ (SEQ ID NO: 9). See, e.g., U.S. Patent Application Publication No. US 2009/0087411 Al, incorporated by reference.
  • exemplary chimeric polypeptides also include Factor VIII fused to at least one PAS sequence or fragment, variant, or derivative thereof.
  • the PAS sequence can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII.
  • a PAS peptide or PAS sequence as used herein, means an amino acid sequence comprising mainly alanine and serine residues or comprising mainly alanine, serine, and proline residues, the amino acid sequence forming random coil conformation under physiological conditions.
  • the PAS sequence is a building block, an amino acid polymer, or a sequence cassette comprising, consisting essentially of, or consisting of alanine, serine, and proline which can be used as a part of the heterologous moiety in the chimeric protein.
  • An amino acid polymer also can form random coil conformation when residues other than alanine, serine, and proline are added as a minor constituent in the PAS sequence.
  • amino acids other than alanine, serine, and proline can be added in the PAS sequence to a certain degree, e.g., up to about 12%, i.e., about 12 of 100 amino acids of the PAS sequence, up to about 10%, up to about 9%>, up to about 8%>, about 6%, about 5%, about 4%>, about 3%), i.e. about 2%>, or about 1%>, of the amino acids.
  • the amino acids different from alanine, serine and proline cab be selected from the group consisting of Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, He, Leu, Lys, Met, Phe, Thr, Trp, Tyr, and Val.
  • a PAS peptide forms a random coil conformation and thereby can mediate an increased in vivo and/or in vitro stability to a recombinant protein of the invention, and has procoagulant activity.
  • Non-limiting examples of the PAS peptides include ASPAAPAPASPAAPAPSAPA
  • exemplary chimeric polypeptides also include Factor VIII fused to at least one transferrin peptide or fragment, variant, or derivative thereof.
  • At least one transferrin peptide can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII.
  • Any transferrin can be fused to or inserted into a recombinant FVIII protein of the invention.
  • wild-type human Tf (Tf) is a 679 amino acid protein, of approximately 75 KDa (not accounting for glycosylation), with two main domains, N (about 330 amino acids) and C (about 340 amino acids), which appear to originate from a gene duplication.
  • Transferrin transports iron through transferrin receptor (Tf ) -mediated endocytosis.
  • Tf After the iron is released into an endosomal compartment and Tf-TfR complex is recycled to cell surface, the Tf is released back extracellular space for next cycle of iron transporting.
  • Tf possesses a long half-life that is in excess of 14-17 days (Li et al, Trends Pharmacol. Sci. 23:206- 209 (2002)).Transferrin fusion proteins have been studied for half-life extension, targeted deliver for cancer therapies, oral delivery and sustained activation of proinsulin (Brandsma et al, Biotechnol. Adv., 29: 230-238 (2011); Bai et al, Proc. Natl. Acad. Sci. USA 102:7292-7296 (2005); Kim et al., J. Pharmacol. Exp. Ther., 334:682-692 (2010); Wang et al., J. Controlled Release 155:386-392 (2011)).
  • exemplary chimeric polypeptides also include Factor VIII fused to at least one polyethylene glycol (PEG) moieties.
  • PEG polyethylene glycol
  • PEGylated FVIII can refer to a conjugate formed between FVIII and at least one polyethylene glycol (PEG) molecule.
  • PEG is commercially available in a large variety of molecular weights and average molecular weight ranges. Typical examples of PEG average molecular weight ranges include, but are not limited to, about 200, about 300, about 400, about 600, about 1000, about 1300-1600, about 1450, about 2000, about 3000, about 3000-3750, about 3350, about 3000-7000, about 3500-4500, about 5000-7000, about 7000-9000, about 8000, about 10000, about 8500-11500, about 16000-24000, about 35000, about 40000, about 60000, and about 80000 daltons. These average molecular weights are provided merely as examples and are not meant to be limiting in any way.
  • a recombinant FVIII protein of the invention can be PEGylated to include mono- or poly-
  • PEGylation can be carried out by any of the PEGylation reactions known in the art.
  • Methods for preparing a PEGylated protein product will generally include (i) reacting a polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the peptide of the invention becomes attached to one or more PEG groups; and (ii) obtaining the reaction product(s).
  • polyethylene glycol such as a reactive ester or aldehyde derivative of PEG
  • the optimal reaction conditions for the reactions will be determined case by case based on known parameters and the desired result.
  • FVIII variants can contain cysteine substitutions in one or more insertion sites in FVIII, and the cysteines can be further conjugated to PEG polymer. See Mei et al, Blood 116:270-279 (2010) and U.S. Patent No. 7,632,921, which are incorporated herein by reference in their entireties.
  • exemplary chimeric polypeptides also include Factor VIII fused to at least one hydroxyethyl starch (HES) polymer.
  • HES is a derivative of naturally occurring amylopectin and is degraded by alpha-amylase in the body. HES exhibits advantageous biological properties and is used as a blood volume replacement agent and in hemodilution therapy in the clinics. See, e.g., Sommermeyer et al., Whypharmazie 8:271-278 (1987); and Weidler et al., Arzneim.-Forschung/Drug Res. 41 : 494-498 (1991).
  • HES is mainly characterized by the molecular weight distribution and the degree of substitution.
  • HES has a mean molecular weight (weight mean) of from 1 to 300 kD, from 2 to 200kD, from 3 to 100 kD, or from 4 to 70kD.
  • Hydroxyethyl starch can further exhibit a molar degree of substitution of from 0.1 to 3, from 0.1 to 2,from 0.1 to 0.9, or from 0.1 to 0.8, and a ratio between C2:C6 substitution in the range of from 2 to 20 with respect to the hydroxyethyl groups.
  • HES with a mean molecular weight of about 130 kD is Voluven ® from Fresenius.
  • Voluven ® is an artificial colloid, employed, e.g. , for volume replacement used in the therapeutic indication for therapy and prophylaxis of hypovolaemia.
  • HES attachment methods available to those skilled in the art, e.g., the same PEG attachment methods described above.
  • a chimeric polypeptide comprising a Factor VIII portion has an increased half-life (tl/2) over a polypeptide consisting of the same Factor VIII portion without the non Factor VIII portion.
  • a chimeric Factor VIII polypeptide with an increased tl/2 may be referred to herein as a long-acting Factor VIII.
  • Long-acting chimeric Factor VIII polypeptides include, e.g., Factor VIII fused to Fc (including, e.g., chimeric Factor VIII polypeptides in the form of a hybrid such as a FVIIIFc monomer dimer hybrid; see Example 1 , Fig. 1 , and Table 2A; and US Patent Nos. 7,404,956 and 7,348,004), Factor VIII fused to XTEN, and Factor VIII fused to albumin.
  • Culture means to incubate cells under in vitro conditions that allow for cell growth or division or to maintain cells in a living state.
  • Cultured cells means cells that are propagated in vitro.
  • Factor VIII means functional factor VIII polypeptide in its normal role in coagulation, unless otherwise specified.
  • Factor VIII includes variant polypeptides that are functional.
  • Factor VIII proteins can be the human, porcine, canine, and murine factor VIII proteins.
  • the full length polypeptide and polynucleotide sequences are known, as are many functional fragments, mutants and modified versions. Examples of human factor VIII sequences are shown as subsequences in SEQ ID NOs:2 or 6 (Table 2).
  • Factor VIII polypeptides include, e.g., full-length factor VIII, full- length factor VIII minus Met at the N-terminus, mature factor VIII (minus the signal sequence), mature factor VIII with an additional Met at the N-terminus, and/or factor VIII with a full or partial deletion of the B domain.
  • Factor VIII variants include B domain deletions, whether partial or full deletions.
  • Pat. No. 4,965,199 discloses a recombinant DNA method for producing factor VIII in mammalian host cells and purification of human factor VIII.
  • Human factor VIII expression in CHO (Chinese hamster ovary) cells and BHKC (baby hamster kidney cells) has been reported.
  • Human factor VIII has been modified to delete part or all of the B domain (U.S. Pat. Nos. 4,994,371 and 4,868,112, each of which is incorporated herein by reference in its entirety), and replacement of the human factor VIII B domain with the human factor V B domain has been performed (U.S. Pat. No. 5,004,803, incorporated herein by reference in its entirety).
  • the cDNA sequence encoding human factor VIII and predicted amino acid sequence are shown in SEQ ID NOs: l and 2, respectively, of US Application Publ. No. 2005/0100990, incorporated herein by reference in its entirety.
  • a number of functional factor VIII molecules, including B-domain deletions, are disclosed in the following patents US 6,316,226 and US 6,346,513, both assigned to Baxter; US 7,041,635 assigned to In2Gen; US 5,789,203, US 6,060,447, US 5,595,886, and US 6,228,620 assigned to Chiron; US 5,972,885 and US 6,048,720 assigned to Biovitrum, US 5,543,502 and US 5,610,278 assigned to Novo Nordisk; US 5,171,844 assigned to Immuno Ag; US 5,112,950 assigned to Transgene S.A.; US 4,868,112 assigned to Genetics Institute, each of which is incorporated herein by reference in its entirety.
  • porcine factor VIII sequence is published, (Toole, J. J., et al, Proc. Natl. Acad. Sci.
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90% or
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2; and amino acids 20 to 2351 of SEQ ID NO:6).
  • the Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90%> or
  • Exavalent dose means the same dose of Factor VIII activity as expressed in International Units, which is independent of molecular weight of the polypeptide in question.
  • IU of factor VIII activity corresponds approximately to the quantity of factor VIII in one milliliter of normal human plasma.
  • assays are available for measuring Factor VIII activity, including the European Pharmacopoeia chromogenic substrate assay and a one stage clotting assay.
  • Fc neonatal Fc receptor (FcRn) binding partners, unless otherwise specified.
  • An FcRn binding partner is any molecule that can be specifically bound by the FcRn receptor with consequent active transport by the FcRn receptor of the FcRn binding partner.
  • Fc includes any variants of IgG Fc that are functional.
  • the region of the Fc portion of IgG that binds to the FcRn receptor has been described based on X-ray crystallography (Burmeister et al. , Nature 372:379 (1994), incorporated herein by reference in its entirety).
  • the major contact area of the Fc with the FcRn is near the junction of the CH2 and CH3 domains.
  • Fc-FcRn contacts are all within a single Ig heavy chain.
  • the FcRn binding partners include, e.g., whole IgG, the Fc fragment of IgG, and other fragments of IgG that include the complete binding region of FcRn.
  • the major contact sites include amino acid residues 248, 250-257, 272, 285, 288, 290-291, 308-311, and 314 of the CH2 domain and amino acid residues 385-387, 428, and 433-436 of the CH3 domain.
  • References made to amino acid numbering of immunoglobulins or immunoglobulin fragments, or regions, are all based on Kabat et al. 1991, Sequences of Proteins of Immunological Interest, U. S.
  • FcRn receptor has been isolated from several mammalian species including humans. The sequences of the human FcRn, rat FcRn, and mouse FcRn are known (Story et al., J. Exp. Med. 180: 2377 (1994), incorporated herein by reference in its entirety.)
  • An Fc may comprise the CH2 and CH3 domains of an immunoglobulin with or without the hinge region of the immunoglobulin. Exemplary Fc variants are provided in WO 2004/101740 and WO 2006/074199, incorporated herein by reference in its entirety.
  • Fc (or Fc portion of a chimeric polypeptide) may contain one or more mutations, and combinations of mutations.
  • Fc (or Fc portion of a chimeric polypeptide) may contain mutations conferring increased half-life such as M252Y, S254T, T256E, and combinations thereof, as disclosed in Oganesyan et al., Mol. Immunol. 46: 1750 (2009), which is incorporated herein by reference in its entirety; H433K, N434F, and combinations thereof, as disclosed in Vaccaro et al, Nat. Biotechnol.
  • Fc (or Fc portion of a chimeric polypeptide) may also include, e.g., the following mutations:
  • the Fc region of IgG can be modified according to well recognized procedures such as site directed mutagenesis and the like to yield modified IgG or Fc fragments or portions thereof that will be bound by FcRn.
  • Such modifications include, e.g., modifications remote from the FcRn contact sites as well as modifications within the contact sites that preserve or even enhance binding to the FcRn.
  • modifications include, e.g., modifications remote from the FcRn contact sites as well as modifications within the contact sites that preserve or even enhance binding to the FcRn.
  • Fcyl single amino acid residues in human IgGl Fc
  • alanine may be substituted for the wildtype amino acids at the positions specified above. Mutations may be introduced singly into Fc giving rise to more than one hundred FcRn binding partners distinct from native Fc. Additionally, combinations of two, three, or more of these individual mutations may be introduced together, giving rise to hundreds more FcRn binding partners. Certain of these mutations may confer new functionality upon the FcRn binding partner. For example, one embodiment incorporates N297A, removing a highly conserved N-glycosylation site.
  • the effect of this mutation is to reduce immunogenicity, thereby enhancing circulating half-life of the FcRn binding partner, and to render the FcRn binding partner incapable of binding to FcyRI, FcyRIIA, FcyRIIB, and FcyRIIIA, without compromising affinity for FcRn (Routledge et al. 1995, Transplantation 60:847, which is incorporated herein by reference in its entirety; Friend et al. 1999, Transplantation 68: 1632, which is incorporated herein by reference in its entirety; Shields et al. 1995, J. Biol. Chem. 276:6591 , which is incorporated herein by reference in its entirety).
  • At least three human Fc gamma receptors appear to recognize a binding site on IgG within the lower hinge region, generally amino acids 234-237. Therefore, another example of new functionality and potential decreased immunogenicity may arise from mutations of this region, as for example by replacing amino acids 233-236 of human IgGl "ELLG” to the corresponding sequence from IgG2 "PVA” (with one amino acid deletion). It has been shown that FcyRI, FcyRII, and FcyRIII which mediate various effector functions will not bind to IgGl when such mutations have been introduced (Ward and Ghetie, Therapeutic Immunology 2:11 (1995), which is incorporated herein by reference in its entirety; and Armour et al., Eur. J.
  • affinity for FcRn may be increased beyond that of wild type in some instances. This increased affinity may reflect an increased "on” rate, a decreased “off rate or both an increased “on” rate and a decreased "off rate. Mutations believed to impart an increased affinity for FcRn include, e.g., T256A, T307A, E380A, and N434A (Shields et al. , J. Biol. Chem. 276:6591 (2001), which is incorporated herein by reference in its entirety).
  • the Fc (or Fc portion of a chimeric polypeptide) may be at least 90% or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
  • the Fc (or Fc portion of a chimeric polypeptide) may be identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 and amino acids 2352 to 2578 of SEQ ID NO:6).
  • Hybrid polypeptides and proteins means a combination of a chimeric polypeptide with a second polypeptide.
  • the chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via protein-protein interactions, such as charge-charge or hydrophobic interactions.
  • the chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via disulfide or other covalent bond(s).
  • Hybrids are described in WO 2004/101740 and WO 2006/074199, each of which is incorporated herein by reference in its entirety. See also U.S. Patent Nos. 7,404,956 and 7,348,004, each of which is incorporated herein by reference in its entirety.
  • the second polypeptide may be a second copy of the same chimeric polypeptide or it may be a non-identical chimeric polypeptide. See, e.g., Figure 1, Example 1, and Table 2.
  • the second polypeptide is a polypeptide comprising an Fc.
  • the chimeric polypeptide is a chimeric Factor VIII-Fc polypeptide and the second polypeptide consists essentially of Fc, e.g., the hybrid polypeptide of Example 1, which is a rFVIIIFc recombinant fusion protein consisting of a single molecule of recombinant B- domain deleted human FVIII (BDD-rFVIII) fused to the dimeric Fc domain of the human IgGl, with no intervening linker sequence.
  • This hybrid polypeptide is referred to herein as FVIIIFc monomelic Fc fusion protein, FVIIIFc monomer hybrid, monomeric FVIIIIFc hybrid, and FVIIIFc monomer-dimer. See Example 1, Fig. 1, and Table 2A. The Examples provide preclinical and clinical data for this hybrid polypeptide.
  • the second polypeptide in a hybrid may comprise or consist essentially of a sequence at least 90%) or 95%> identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95%> identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO-:4).
  • the second polypeptide may comprise or consist essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
  • FIG. 1 is a schematic showing the structure of a B domain deleted factor VIII-Fc chimeric polypeptide, and its association with a second polypeptide that is an Fc polypeptide.
  • the coding sequence of human recombinant B-domain deleted FVIII was obtained by reverse transcription-polymerase chain reaction (RT-PCR) from human liver poly A RNA (Clontech) using FVIII-specific primers.
  • the FVIII sequence includes the native signal sequence for FVIII.
  • the B-domain deletion was from serine 743 (S743; 2287 bp) to glutamine 1638 (Q1638; 4969 bp) for a total deletion of 2682 bp.
  • the coding sequence for human recombinant Fc was obtained by RT-PCR from a human leukocyte cDNA library (Clontech) using Fc specific primers. Primers were designed such that the B-domain deleted FVIII sequence was fused directly to the N-terminus of the Fc sequence with no intervening linker.
  • the FVIIIFc DNA sequence was cloned into the mammalian dual expression vector pBUDCE4.1 (Invitrogen) under control of the CMV promoter.
  • a second identical Fc sequence including the mouse Igk signal sequence was obtained by RT-PCR and cloned downstream of the second promoter, EFla, in the expression vector pBUDCE4.1.
  • the rFVIIIFc expression vector was transfected into human embryonic kidney 293 cells
  • the conditioned medium contained Fc and monomelic rFVIIIFc. It is possible that the size of dimeric rFVIIIFc was too great and prevented efficient secretion from the cell. This result was beneficial since it rendered the purification of the monomer less complicated than if all three proteins had been present.
  • the material used in these studies had a specific activity of approximately 9000 IU/mg.
  • the polypeptides of the invention are administered to a patient who expresses high level of von Willebrand factor (VWF).
  • VWF von Willebrand factor
  • Subject or “patient” as used herein means a human individual.
  • a subject can be a patient who is currently suffering from a bleeding disorder or is expected to be in need of such a treatment.
  • Subject can include an adult or a pediatric subject.
  • the pediatric subject can be a pediatric patient under the age of 12.
  • the term “pediatrics” as used herein is the branch of medicine that deals with the care of infants and children and the treatment of their diseases.
  • the subject is a pediatric patient who has a diagnosis of severe hemophilia A.
  • pediatric subjects are treated with a long-acting Factor VIII polypeptide of the invention.
  • VWF is a plasma protein having a multimer structure in which the molecular weight of the various forms varies between approximately 230 kDa for each monomer subunit and up to more than 20 million Da in the multimer forms of greater molecular weight, thus forming the largest known soluble protein. Its plasma concentration is approximately around 5-10 ⁇ g/ml (Siedlecki et ah, Blood, vol 88: 2939-2950 (1996)) and the plasma form of smaller size is that corresponding to the dimer, with an approximate size of 500 kDa.
  • VWF has an essential role to play in primary haemostasis, being responsible for the adhesion of platelets to damaged vascular surfaces and therefore formation of the platelet plug on which the mechanisms for formation of the fibrin coagulate develop. It is suggested that the higher molecular weight multimers support platelet adhesion mechanisms to the sub-endothelium with greater efficiency and the clinical efficacy of VWF concentrates has been related to the concentration of these multimers of higher molecular weight (Metzner et al, Haemophilia 4:25- 32 (1998).
  • the average range of VWF in plasma is between about 50 IU/dL and about 200 IU/dL. In one embodiment, the average level of VWF in plasma is about 50 IU/dL. In another embodiment, a VWF level in plasma of at least about 100 IU/dL is considered a high VWF level. In another embodiment, a high level of VWF in plasma is between about 100 IU/dL and about 200 IU/dL.
  • a high level of VWF in plasma is at least about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
  • subjects expressing at least about 100 IU/dL of plasma are expressing at least about 100 IU/dL of plasma.
  • VWF are administered a long-acting FVIII polypeptide of the invention at a long interval dosing regimen.
  • the long-acting FVIII polypeptide is administered at a dosing interval of at least about 3 days.
  • the long-acting FVIII polypeptide is administered at a dosing interval of at least about once every week, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
  • the subjects were previously identified as having high levels of
  • subjects having a blood serotype other than O ⁇ i.e., A, B, or AB) require less frequent dosing of long-acting FVIII because the long-acting FVIII has a longer half- life in these subjects. In these subjects, the increased half-life is due to their elevated VWF levels.
  • pharmacokinetic data defined as the study of the time course of drug absorption, distribution, metabolism, and excretion, can be used as an identifier of subjects eligible for longer or shorter dosing intervals using a long-acting FVIII polypeptide of the invention. Cinical pharmacokinetics is the application of pharmacokinetic principles to the safe and effective therapeutic management of drugs in an individual patient. The primary goals of clinical pharmacokinetics include enhancing efficacy and decreasing toxicity of a patient's drug therapy. The development of strong correlations between drug concentrations and their pharmacologic responses has enabled clinicians to apply pharmacokinetic principles to actual patient situations.
  • the half-life of a FVIII-Fc polypeptide of the invention is used to identify patients who express high levels of VWF.
  • the range of half-life of FVIII-Fc is between about 10 and about 40 hours, depending at least in part on the levels of VWF also present. On average however, the half- life of FVIII-Fc is about 18 hours.
  • FVIII-Fc exhibits an increased half-life of at least about 1.2-fold in patients having high levels of VWF compared to the half-life of FVIII-Fc when administered to individuals having average levels of VWF.
  • FVIII-Fc exhibits an increased half-life of at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold compared to the half-life of FVIII-Fc when administered to individuals having average levels of VWF.
  • the half-life of FVIII-Fc in subjects expressing high levels of VWF, is between at least about 20 hours and about 40 hours.
  • the half-life of FVIII-Fc is at least about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours. In one embodiment the half- life of FVIII-Fc is between about 20 and about 27 hours in subjects having high levels of VWF. Thus, in one embodiment, an increased half-life of FVIII-Fc compared to average values is indicative of a subject that is eligible for a longer dosing interval with a long-acting FVIII polypeptide of the invention.
  • the half-life of a short-acting FVIII polypeptide is used to identify patients who express high levels of VWF.
  • short-acting FVIII refers to a FVIII polypeptide in which no extenders of half-life have been added.
  • short-acting FVIII polypeptides consist of full-length or B domain-deleted FVIII. Examples of short-acting FVIII polypeptides are Advate ® and ReFacto ® .
  • short-acting FVIII polypeptides can also be used to identify patients that are eligible for a longer dosing interval of a long-acting FVIII polypeptide of the invention.
  • the short-acting FVIII exhibits an increased half-life of at least about 1.2-fold in individuals expressing high levels of VWF compared to the half-life of the short-acting FVIII when administered to individuals having average levels of VWF.
  • the short- acting FVIII exhibits an increased half-life of at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold in individuals expressing high levels of VWF compared to the half- life of the short-acting FVIII when administered to individuals having average levels of VWF.
  • individuals that demonstrate an increased half- life of at least about 1.2-fold when they are administered a short-acting FVIII are eligible for a longer dosing interval with a long-acting FVIII polypeptide of the invention.
  • Dosing interval means the dose of time that elapses between multiple doses being administered to a subject.
  • the comparison of dosing interval may be carried out in a single subject or in a population of subjects and then the average obtained in the population may be calculated.
  • the dosing interval when administering a chimeric Factor VIII polypeptide may be at least about one and one-half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII).
  • the dosing interval may be at least about one and one -half to six times longer, one and one-half to five times longer, one and one-half to four times longer, one and one -half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII).
  • the dosing interval may be at least about one and one-half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one-half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII).
  • the dosing interval may be about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
  • the dosing interval may be at least about one and one-half to 5, one and one-half, 2, 3, 4, or 5 days or longer.
  • the dosing interval of said chimeric polypeptide or hybrid is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
  • the effective dose is 25-80 IU/kg (25, 26, 27, 28, 29, 30, 31, 32, 33,
  • the dosing interval is once every 3-5, 3-6, 3-7, 3, 4, 5, 6, 7, or 8 or more days, or three times per week, or no more than three times per week.
  • the effective dose is 80 IU/kg and the dosing interval is once every 3 days.
  • the effective dose of 80 IU/kg given at a dosing interval of every 3 days is administered to a pediatric subject.
  • the effective dose is 65 IU/kg and the dosing interval is once weekly, or once every 6-7 days.
  • the doses can be administered repeatedly as long as they are necessary (e.g., at least 10, 20, 28, 30, 40, 50, 52, or 57 weeks, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 years).
  • the effective dose for on-demand treatment is 20-50IU/Kg (20,
  • the on-demand treatment can be one time dosing or repeated dosing.
  • the dosing interval can be every 12-24 hours, every 24-36 hours, every 24- 48 hours, every 36-48 hours, or every 48-72 hours.
  • Long-acting Factor VIII is a Factor VIII having an increased half-life (also referred to herein as tl/2, tl/2 beta, elimination half-life and HL) over a reference Factor VIII.
  • the increased half-life of a long-acting Factor VIII may be due to fusion to one or more non-Factor VIII polypeptides such as, e.g., Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof.
  • non-Factor VIII polypeptides such as, e.g., Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of h
  • the increased half-life may be due to one or more modification, such as, e.g., pegylation.
  • exemplary long-acting Factor VIII polypeptides include, e.g., chimeric Factor VIII polypeptides comprising Fc, chimeric Factor VIII polypeptides comprising XTEN and chimeric Factor VIII polypeptides comprising albumin. Additional exemplary long-acting Factor VIII polypeptides include, e.g., pegylated Factor VIII.
  • the "reference" polypeptide in the case of a long-acting chimeric Factor VIII polypeptide, is a polypeptide consisting essentially of the Factor VIII portion of the chimeric polypeptide, e.g., the same Factor VIII portion without the Fc portion, without the XTEN portion, or without the albumin portion.
  • the reference polypeptide in the case of a modified Factor VIII is the same Factor VIII without the modification, e.g., a Factor VIII without the pegylation.
  • the long-acting Factor VIII has one or more of the following properties when administered to a subject:
  • MRT mean residence time
  • a clearance (CL) (activity) in said subject of about 1.22-5.19 mL/hour/kg or less;
  • a tl/2beta (activity) in said subject of about 11-26.4 hours;
  • K value activity; observed
  • Vss activity in said subject of about 37.7-79.4 mL/kg
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • K- Value a mean incremental recovery (activity; observed) greater that 1.38 IU/dL per IU/kg; a mean incremental recovery (K- Value) (activity; observed) of at least about 1.5, at least about 1.85, or at least about 2.46 IU/dL per IU/kg.
  • a mean clearance (CL) (activity) in said patient population of about 2.33 ⁇ 1.08 mL/hour/kg or less;
  • a mean clearance (CL) (activity) in said patient population of about 1.8-2.69 mL/hour/kg;
  • a mean clearance (CL) (activity) in said patient population that is about 65% of the clearance of a polypeptide comprising said Factor VIII without modification;
  • MRT mean mean residence time
  • a mean MRT (activity) in said patient population of about 25.9 - 26.5 hours;
  • a mean MRT (activity) in said patent population that is about 1.5 fold longer than the mean MRT of a polypeptide comprising said Factor VIII without modification;
  • a mean tl/2beta (activity) in said patient population of about 18.3 ⁇ 5.79 hours;
  • a mean tl/2beta (activity)in said patient population that is about 18 - 18.4 hours;
  • a mean tl/2beta (activity) in said patient population that is about 1.5 fold longer than the mean tl/2beta of a polypeptide comprising said Factor VIII without modification;
  • K value a mean incremental recovery (activity; observed) in said patient population of about 2.01 ⁇ 0.44 IU/dL per IU/kg;
  • K value a mean incremental recovery (activity; observed) in said patient population of about 1.85 - 2.46 IU/dL per IU/kg;
  • K value activity; observed
  • a mean AUC/dose (activity) in said patient population of about 49.9 ⁇ 18.2 IU*h/dL per IU/kg; a mean AUC/dose (activity) in said patient population of about 44.8 - 57.6 IU*h/dL per IU/kg.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • a Cmax OBS in said subject administered with the chimeric polypeptide is comparable to the Cmax OBS in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • a C mx _OBS in said subject of about 60.5 IU/dL, about 60.5 ⁇ 1 IU/dL, about 60.5 ⁇ 2 IU/dL, about 60.5 ⁇ 3 IU/dL, about 60.5 ⁇ 4 IU/dL, about 60.5 ⁇ 5 IU/dL, about 60.5 ⁇ 6 IU/dL, about 60.5 ⁇ 7 IU/dL, about 60.5 ⁇ 8 IU/dL, about 60.5 ⁇ 9 IU/dL, or about 60.5 ⁇ 10 IU/dL as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide
  • a Cmax OBS in said subject of about 103 - 136 IU/dL as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • a C ⁇ OBS in said subject of about 76.5 IU/dL, about 76.5 ⁇ 1 IU/dL, about 76.5 ⁇ 2 IU/dL, about 76.5 ⁇ 3 IU/dL, about 76.5 ⁇ 4 IU/dL, about 76.5 ⁇ 5 IU/dL, about 76.5 ⁇ 6 IU/dL, about 76.5 ⁇ 7 IU/dL, about 76.5 ⁇ 8 IU/dL, about 76.5 ⁇ 9 IU/dL, about 76.5 ⁇ 10 IU/dL, about 76.5 ⁇ 11 IU/dL, about 76.5 ⁇ 12 IU/dL, about 76.5 ⁇ 13 IU/dL, about 76.5 ⁇ 14 IU/dL, or about 76.5 ⁇ 15 IU/dL, as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • a Cmax OBS in said subject of about 64.9 - 90.1 IU/dL as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • a C max _OBS in said subject of about 146 - 227 IU/dL, about 146 ⁇ 5 IU/dL, about 146 ⁇ 10 IU/dL, about 227 ⁇ 5 IU/dL, or about 146 ⁇ 10 IU/dL as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • a tl/2beta (activity) in said subject that is at least 1.48, 1.49, 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, or 1.90 times higher than the tl/2beta (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • a tl/2beta (activity) in said subject of about 16.7 hours, 16.7 ⁇ 1 hours, 16.7 ⁇ 2 hours, 16.7 ⁇ 3 hours, 16.7 ⁇ 4 hours, 16.7 ⁇ 5 hours, 16.7 ⁇ 6 hours, 16.7 ⁇ 7 hours, 16.7 ⁇ 8 hours, 16.7 ⁇ 9 hours, 16.7 ⁇ 10 hours, or 16.7 ⁇ 11 hours as measured by a two stage (chromogenic) assay;
  • a tl/2beta (activity) in said subject of about 13.8 - 20.1 hours as measured by a two stage (chromogenic) assay;
  • a tl/2beta (activity) in said subject of about 19.8 hours, 19.8 ⁇ 1 hours, 19.8 ⁇ 2 hours, 19.8 ⁇ 3 hours, 19.8 ⁇ 4 hours, 19.8 ⁇ 5 hours, 19.8 ⁇ 6 hours, 19.8 ⁇ 7 hours, 19.8 ⁇ 8 hours, 19.8 ⁇ 9 hours, 19.8 ⁇ 10 hours, or 19.8 ⁇ 11 hours as measured by a two stage (chromogenic) assay; or
  • a tl/2beta (activity) in said subject of about 14.3 - 27.5 hours as measured by a two stage (chromogenic) assay.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • a clearance (CL) (activity) in said subject is 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, or 0.70 times lower than the clearance in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • a clearance (CL) (activity) in said subject of about 1.68 mL/hour/kg, 1.68 ⁇ 0.1 mL/hour/kg, 1.68 ⁇ 0.2 mL/hour/kg, 1.68 ⁇ 0.3 mL/hour/kg, 1.68 ⁇ 0.4 mL/hour/kg, 1.68 ⁇ 0.5 mL/hour/kg, 1.68 ⁇ 0.6 mL/hour/kg, or 1.68 ⁇ 0.7 mL/hour/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • aPTT one stage
  • a clearance (CL) activity
  • aPTT one stage assay when about 25 IU/kg of the chimeric polypeptide is administered
  • a clearance (CL) (activity) in said subject of about 2.32 mL/hour/kg, 2.32 ⁇ 0.1 mL/hour/kg, 2.32 ⁇ 0.2 mL/hour/kg, 2.32 ⁇ 0.3 mL/hour/kg, 2.32 ⁇ 0.4 mL/hour/kg, 2.32 ⁇ 0.5 mL/hour/kg, 2.32 ⁇ 0.6 mL/hour/kg, or 2.32 ⁇ 0.7 mL/hour/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • aPTT one stage
  • a clearance (CL) activity
  • aPTT oa ne stage
  • a clearance (CL) (activity) in said subject of about 1.49 mL/hour/kg, 1.49 ⁇ 0.1 mL/hour/kg, 1.49 ⁇ 0.2 mL/hour/kg, 1.49 ⁇ 0.3 mL/hour/kg, 1.49 ⁇ 0.4 mL/hour/kg, 1.49 ⁇ 0.5 mL/hour/kg, 1.49 ⁇ 0.6 mL/hour/kg, or 1.49 ⁇ 0.7 mL/hour/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • a clearance (CL) (activity) in said subject of about 1.05-2.20 mL/hour/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • a MRT in said subject is at least 1.46, 1.47, 1.48, 1.49, 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.90, 1.91, 1.92, or 1.93 times higher than the MRT in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • a MRT (activity) in said subject of about 27 hours, 27 ⁇ 1 hours, 27 ⁇ 2 hours, 27 ⁇ 3 hours, 27 ⁇ 4 hours, 27 ⁇ 5 hours, 27 ⁇ 6 hours, 27 ⁇ 7 hours, 27 ⁇ 8 hours, 27 ⁇ 9 hours, or 27 ⁇ 10 hours as meansured by a one stage (aPTT) assay;
  • a MRT activity in said subject of about 20.6 - 35.3 hours as meansured by a one stage (aPTT) assay;
  • a MRT (activity) in said subject of about 23.9 - 28.5 hours as measured by a two stage (chromogenic) assay;
  • a MRT (activity) in said subject of about 19.8 - 28.9 hours as measured by a two stage (chromogenic) assay;
  • a MRT (activity) in said subject of about 20.5 - 39.6 hours as measured by a two stage (chromogenic) assay.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • an incremental recovery in said subject that is comparable to the Incremental Recovery in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • an incremental recovery in said subject of about 1.59 - 2.10 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • an incremental recovery in said subject of about 2.61-3.66 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; or
  • an incremental recovery in said subject of about 2.24-3.50 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • Vss activity in said subject that is comparable to the Vss (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • a Vss activity in said subject of about 45.5 mL/kg, 45.5 ⁇ 1 mL/kg, 45.5 ⁇ 2 mL/kg, 45.5 ⁇ 3 mL/kg, 45.5 ⁇ 4 mL/kg, 45.5 ⁇ 5 mL/kg, 45.5 ⁇ 6 mL/kg, 45.5 ⁇ 7 mL/kg, 45.5 ⁇ 8 mL/kg, 45.5 ⁇ 9 mL/kg, 45.5 ⁇ 10 mL/kg, or 45.5 ⁇ 11 mL/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • aPTT one stage
  • Vss activity in said subject of about 39.3 - 52.5 mL/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered; a Vss (activity) in said subject of about 62.8 mL/kg, 62.8 ⁇ 1 mL/kg, 62.8 ⁇ 2 mL/kg, 62.8 ⁇ 3 mL/kg, 62.8 ⁇ 4 mL/kg, 62.8 ⁇ 5 mL/kg, 62.8 ⁇ 6 mL/kg, 62.8 ⁇ 7 mL/kg, 62.8 ⁇ 8 mL/kg, 62.8 ⁇ 9 mL/kg, 62.8 ⁇ 10 mL/kg, 62.8 ⁇ 1 1 mL/kg, 62.8 ⁇ 12 mL/kg, 62.8 ⁇ 13 mL/kg, 62.8 ⁇ 14 mL/kg, 62.8 ⁇ 15 mL/kg, or 62.8 ⁇ 16 mL/kg
  • Vss activity in said subject of about 55.2 - 71.5 mL/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • a Vss activity in said subject of about 35.9 mL/kg, 35.9 ⁇ 1 mL/kg, 35.9 ⁇ 2 mL/kg, 35.9 ⁇ 3 mL/kg, 35.9 ⁇ 4 mL/kg, 35.9 ⁇ 5 mL/kg, 35.9 ⁇ 6 mL/kg, 35.9 ⁇ 7 mL/kg, 35.9 ⁇ 8 mL/kg, 35.9 ⁇ 9 mL/kg, 35.9 ⁇ 10 mL/kg, 35.9 ⁇ 1 1 mL/kg, 35.9 ⁇ 12 mL/kg, or 35.9 ⁇ 13 mL/kg, as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • Vss activity in said subject of about 30.4-42.3 mL/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • Vss activity in said subject of about 43.4 mL/kg, 43.4 ⁇ 1 mL/kg, 43.4 ⁇ 2 mL/kg, 43.4 ⁇ 3 mL/kg, 43.4 ⁇ 4 mL/kg, 43.4 ⁇ 5 mL/kg, 43.4 ⁇ 6 mL/kg, 43.4 ⁇ 7 mL/kg, 43.4 ⁇ 8 mL/kg, 43.4 ⁇ 9 mL/kg, 43.4 ⁇ 10 mL/kg, 43.4 ⁇ 1 1 mL/kg, 43.4 ⁇ 12 mL/kg, 43.4 ⁇ 13 mL/kg, 43.4 ⁇ 14 mL/kg, 43.4 ⁇ 15 mL/kg, or 43.4 ⁇ 16 mL/kg, as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; or
  • Vss activity in said subject of about 38.2-49.2 mL/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
  • the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
  • an AUCi NF in said subject that is at least 1.45 1.46, 1.47, 1.48, 1.49, 1.50, 1.51 , 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61 , 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71 , 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81 , 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.90 times higher than the AUC in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
  • an AUCi NF in said subject of about 1440 ⁇ 316 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • an AUCi NF in said subject of about 1 160 - 1880 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • an AUC in said subject of about 1480 hr*IU/dL per IU/kg, 1480 ⁇ 100 hr*IU/dL per IU/kg, 1480 ⁇ 200 hr*IU/dL per IU/kg, 1480 ⁇ 300 hr*IU/dL per IU/kg, 1480 ⁇ 400 hr*IU/dL per IU/kg, 1480 ⁇ 500 hr*IU/dL per IU/kg, 1480 ⁇ 600 hr*IU/dL per IU/kg, 1480 ⁇ 700 hr*IU/dL per IU/kg, 1480 ⁇ 800 hr*IU/dL per IU/kg, 1480 ⁇ 900 hr*IU/dL per IU/kg, or 1480 ⁇ 1000 hr*IU/dL per IU/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
  • aPTT
  • an AUC INP in said subject of about 2910 ⁇ 1320 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • an AUCi NF in said subject of about 1980 - 3970 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • an AUC in said subject of about 2800 hr*IU/dL per IU/kg, 2800 ⁇ 100 hr*IU/dL per IU/kg, 2800 ⁇ 200 hr*IU/dL per IU/kg, 2800 ⁇ 300 hr*IU/dL per IU/kg, 2800 ⁇ 400 hr*IU/dL per IU/kg, 2800 ⁇ 500 hr*IU/dL per IU/kg, 2800 ⁇ 600 hr*IU/dL per IU/kg, 2800 ⁇ 700 hr*IU/dL per IU/kg, 2800 ⁇ 800 hr*IU/dL per IU/kg, 2800 ⁇ 900 hr*IU/dL per IU/kg, or 2800 ⁇ 1000 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
  • aPTT one
  • On-demand treatment means treatment that is intended to take place over a short course of time and is in response to an existing condition, such as a bleeding episode, or a perceived need such as planned surgery.
  • Conditions that may require on-demand treatment include, e.g., a bleeding episode, hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath.
  • the subject may be in need of surgical prophylaxis, peri-operative management, or treatment for surgery.
  • Such surgeries include, e.g., minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
  • on-demand treatment resolves greater than 80% (greater than 80%>, greater than 81%>, greater than 82%, greater than 83%, greater than 84%, greater than 85%, greater than 86%, greater than 87%, greater than 88%, greater than 89%, greater than 90%, greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, greater than 99%, or 100%) or 80-100%, 80-90%, 85-90%, 90-100%, 90-95%, or 95-100% of bleeds (e.g., spontaneous bleeds) in a single dose.
  • bleeds e.g., spontaneous bleeds
  • greater than 80% greater than 81%, greater than 82%, greater than 83%o, greater than 84%, greater than 85%, greater than 86%, greater than 87%, greater than 88%, greater than 89%, greater than 90%, greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or 100%
  • 80-100%, 80-90%, 85-90%, 90-100%, 90-95%, or 95-100% of bleeding episodes are rated excellent or good by physicians after on-demand treatment.
  • greater than 5%o (greater than 6%, greater than 7%, greater than 8%, greater than 9%, greater than 10%, greater than 11%, greater than 12%, greater than 13%, greater than 14%, greater than 15%, greater than 16%, greater than 17%, greater than 18%, greater than 19%, greater than 20%), or 5- 20%o, 5-15%), 5-10%), 10-20%), or 10-15% of bleeding episodes are rated as fair by physicians after on-demand treatment.
  • Polypeptide “peptide” and “protein” are used interchangeably and refer to a polymeric compound comprised of covalently linked amino acid residues.
  • Polynucleotide and “nucleic acid” are used interchangeably and refer to a polymeric compound comprised of covalently linked nucleotide residues.
  • Polynucleotides may be DNA, cDNA, RNA, single stranded, or double stranded, vectors, plasmids, phage, or viruses.
  • Polynucleotides include, e.g., those in Table 1, which encode the polypeptides of Table 2 (see Table 1).
  • Polynucleotides also include, e.g., fragments of the polynucleotides of Table 1, e.g., those that encode fragments of the polypeptides of Table 2, such as the Factor VIII, Fc, signal sequence, 6His and other fragments of the polypeptides of Table 2.
  • Prophylactic treatment means administering a Factor VIII polypeptide in multiple doses to a subject over a course of time to increase the level of Factor VIII activity in a subject's plasma.
  • the increased level can be sufficient to decrease the incidence of spontaneous bleeding or to prevent bleeding, e.g., in the event of an unforeseen injury.
  • the plasma protein level in the subject may not fall below the baseline level for that subject, or below the level of Factor VIII that characterizes severe hemophilia ( ⁇ 1 IU/dl [1%]).
  • the prophylaxis regimen is "tailored" to the individual patient, for example, by determining PK data for each patient and administering Factor VIII of the invention at a dosing interval that maintains a trough level of 1-3% FVIII activity. Adjustments may be made when a subject experiences unacceptable bleeding episodes defined as >2 spontaneous bleeding episodes over a rolling two-month period. In this case, adjustment will target trough levels of 3-5%.
  • prophylactic treatment results in prevention and control of bleeding, sustained control of bleeding, sustained protection from bleeding, and/or sustained benefit.
  • Prophylaxis e.g., sustained protection can be demonstrated by an increased AUC to last measured time point (AUC -LAST) and reduced clearance, resulting in increased terminal tl/2 compared to short acting FVIII.
  • Prophylaxis can be demonstrated by better Cmax, better Tmax, and/or greater mean residence time versus short-acting FVIII.
  • prophylaxis results in no spontaneous bleeding episodes within about 24, 36, 48, 72, or 96 hours (e.g., 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 96, 87, 88, 89, 90, 91, 92, 93, 94, 95, or 96 hours), after injection (e.g., the last injection).
  • injection e.g., the last injection
  • prophylaxis results in greater than 30%> (e.g., greater than 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 96, 87, 88, 89, or 90%, for example, greater than 50%), mean reduction in annualized bleeding episodes with once weekly dosing (e.g., at 65 IU/kg).
  • mean reduction in annualized bleeding episodes with once weekly dosing e.g., at 65 IU/kg.
  • Therapeutic dose means a dose that achieves a therapeutic goal, as described herein.
  • the calculation of the required dosage of factor VIII is based upon the empirical finding that, on average, 1 IU of factor VIII per kg body weight raises the plasma factor VIII activity by approximately 2 IU/dL.
  • the required dosage is determined using the following formula:
  • the therapeutic doses that may be used in the methods of the invention are about 10-100
  • IU/kg more specifically, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 IU/kg, and more specifically, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
  • 10 to about 150 IU/kg more specifically, about 100-110, 110-120, 120-130, 130-140, 140-150 IU/kg, and more specifically, about 110, 115, 120, 125, 130, 135, 140, 145, or 150 IU/kg.
  • Variant refers to a polynucleotide or polypeptide differing from the original polynucleotide or polypeptide, but retaining essential properties thereof, e.g., factor VIII coagulant activity or Fc (FcRn binding) activity. Generally, variants are overall closely similar, and, in many regions, identical to the original polynucleotide or polypeptide. Variants include, e.g., polypeptide and polynucleotide fragments, deletions, insertions, and modified versions of original polypeptides.
  • Variant polynucleotides may comprise, or alternatively consist of, a nucleotide sequence which is at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for example, the nucleotide coding sequence in SEQ ID NO: l, 3, or 5 (the factor VIII portion, the Fc portion, individually or together) or the complementary strand thereto, the nucleotide coding sequence of known mutant and recombinant factor VIII or Fc such as those disclosed in the publications and patents cited herein or the complementary strand thereto, a nucleotide sequence encoding the polypeptide of SEQ ID NO:2, 4, or 6 (the factor VIII portion, the Fc portion, individually or together), and/or polynucleotide fragments of any of these nucleic acid molecules (e.g., those fragments described herein). Polynucleotides which hybridize to these nucleic acid molecules under stringent hybridization conditions or lower stringency conditions are also included as variant
  • Variant polypeptides may comprise, or alternatively consist of, an amino acid sequence which is at least 85%, 90%, 95%, 96%, 97%, 98%, 99% identical to, for example, the polypeptide sequence shown in SEQ ID NOS:2, 4, or 6 (the factor VIII portion, the Fc portion, individually or together), and/or polypeptide fragments of any of these polypeptides (e.g., those fragments described herein).
  • nucleic acid having a nucleotide sequence at least, for example, 95% "identical" to a reference nucleotide sequence it is intended that the nucleotide sequence of the nucleic acid is identical to the reference sequence except that the nucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence.
  • nucleic acid having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • the query sequence may be, for example, the entire sequence shown in SEQ ID NO: l or 3, the ORF (open reading frame), or any fragment specified as described herein.
  • nucleic acid molecule or polypeptide is at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence or polypeptide of the present invention can be determined conventionally using known computer programs.
  • a method for determining the best overall match between a query sequence (reference or original sequence) and a subject sequence also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et ah, Comp. App. Biosci. 6:237-245 (1990), which is herein incorporated by reference in its entirety
  • sequence alignment the query and subject sequences are both DNA sequences.
  • RNA sequence can be compared by converting U's to T's.
  • the result of said global sequence alignment is in percent identity.
  • the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
  • a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%.
  • a 90 base subject sequence is compared with a 100 base query sequence.
  • deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a query amino acid sequence of the present invention it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • the amino acid sequence of the subject polypeptide may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, (indels) or substituted with another amino acid.
  • These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • a method for determining the best overall match between a query sequence (reference or original sequence) and a subject sequence can be determined using the FASTDB computer program based on the algorithm of Brutlag et al., Comp. App. Biosci. 6:237-245(1990), incorporated herein by reference in its entirety.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C- terminal residues of the subject sequence.
  • a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence.
  • deletions are internal deletions so there are no residues at the N- or C- termini of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • the polynucleotide variants may contain alterations in the coding regions, non-coding regions, or both.
  • the polynucleotide variants contain alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide.
  • nucleotide variants are produced by silent substitutions due to the degeneracy of the genetic code.
  • variants in which 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination.
  • Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to others, e.g., a bacterial host such as E. coli).
  • Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
  • variants may be generated to improve or alter the characteristics of the polypeptides. For instance, one or more amino acids can be deleted from the N-terminus or C-terminus of the secreted protein without substantial loss of biological function.
  • Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al, J. Biotechnology 7:199-216 (1988), incorporated herein by reference in its entirety.)
  • polypeptide variants include, e.g., modified polypeptides.
  • Modifications include, e.g., acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation (Mei et al., Blood 776:270-79 (2010), which is incorporated herein by reference in its entirety), prote
  • Factor VIII is modified, e.g., pegylated, at any convenient location.
  • Factor VIII is pegylated at a surface exposed amino acid of Factor VIII, e.g., a surface exposed cysteine, which may be an engineered cysteine. Id..
  • modified Factor VIII e.g., pegylated Factor VIII, is a long-acting Factor VIII.
  • Vss Volume of distribution at steady state
  • Process Factor VIII means Factor VIII that has been cleaved at Arginine 1648 (for full-length Factor VIII) or Arginine 754 (for B- domain deleted Factor VIII), i.e., intracellular processing site. Due to the cleavage at the intracellular processing site, processed Factor VIII comprises two polypeptide chains, the first chain being a heavy chain and the second chain being a light chain.
  • the processed Factor VIII-Fc fusion protein (i.e., Heavy chain and Light chain fused to Fc) run at approximately 90 kDa and 130 kDa on a non-reducing SDS-PAGE, respectively, and 90 kDa and 105 kDa on a reducing SDS-PAGE, respectively. Therefore, in one embodiment, at least about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII.
  • about 50%>, about 60%>, about 70%>, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%> of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII.
  • the chimeric polypeptide comprising processed Factor VIII is purified (or isolated) from the chimeric polypeptide comprising single chain Factor VIII, and at least about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%) of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII.
  • Single chain Factor VIII means Factor IGF
  • single chain Factor VIII in the chimeric polypeptide used herein comprises a single chain.
  • the single chain Factor VIII contains an intact intracellular processing site.
  • the single chain Factor VIII of the invention comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
  • the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648.
  • the substitution or mutation is an amino acid other than arginine, e.g., isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, histidine, ornithine, pyrrolysine, or taurine.
  • arginine e.g., isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, histidine, ornithine, pyrroly
  • the single chain Factor VIII-Fc fusion protein can run at approximately 220 kDa on a non reducing SDS-PAGE and at approximately 195 kDa on a reducing SDS-PAGE.
  • the chimeric polypeptide comprising single chain Factor VIII is purified (or isolated) from the chimeric polypeptide comprising processed Factor VIII, and at least about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99%, or about 100% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII.
  • At least about 1%>, about 5%), about 10%>, about 15%>, about 20%>, about 25%>, about 30%>, or about 35%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
  • about 1%-about 10%, about 5%-about 15%, about 10%-about 20%, about 15%-about 25%, about 20%- about 30%, about 25%-about 35%, about 30%-about 40% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII.
  • about 1%, about 5%, about 10%, about 15%, about 20% about 25%, about 30%, about 35% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII.
  • the ratio of the single chain Factor VIII to the processed Factor VIII of the chimeric polypeptide is (a) about 25%> of single chain Factor VIII and about 75%> of processed Factor VIII; (b) about 20%) of single chain Factor VIII and about 80%> of processed Factor VIII; (c) about 15%> of single chain Factor VIII and about 85%> of processed Factor VIII; (d) about 10%> of single chain Factor VIII and about 90%> of processed Factor VIII; (e) about 5%> of single chain Factor VIII and about 95%) of processed Factor VIII; (f) about 1%> of single chain Factor VIII and about 99%> of processed Factor VIII; (g) about 100% of processed Factor VIII, (h) about 30%> of single chain Factor VIII and about 70%> of processed Factor VIII, (i) about 35%> of single chain Factor VIII and about 65%> of processed Factor VIII, or (j) about 40%> of single chain Factor VIII and about 60%) of processed Factor VIII.
  • the ratio of the single chain Factor VIII to the processed Factor VIII of the chimeric polypeptide is (a) about 30%> of single chain Factor VIII and about 70%> of processed Factor VIII; (b) about 40%> of single chain Factor VIII and about 60%) of processed Factor VIII; (c) about 50%> of single chain Factor VIII and about 50%> of processed Factor VIII; (d) about 60%> of single chain Factor VIII and about 40%> of processed Factor VIII; (e) about 70%> of single chain Factor VIII and about 30%> of processed Factor VIII; (f) about 80%) of single chain Factor VIII and about 20%> of processed Factor VIII; (g) about 90%> of single chain Factor VIII and about 10%> of processed Factor VIII; (h) about 95%> of single chain Factor VIII and about 5%> of processed Factor VIII; (i) about 99%> of single chain Factor VIII and about 1%) of processed Factor VIII; or (j) about 100% of single chain Factor VIII.
  • the Factor VIII portion in the chimeric polypeptide used herein has Factor VIII activity.
  • Factor VIII activity can be measured by any known methods in the art.
  • one of those methods can be a chromogenic assay.
  • the chromogenic assay mechanism is based on the principles of the blood coagulation cascade, where activated Factor VIII accelerates the conversion of Factor X into Factor Xa in the presence of activated Factor IX, phospholipids and calcium ions.
  • the Factor Xa activity is assessed by hydrolysis of a p-nitroanilide (pNA) substrate specific to Factor Xa.
  • pNA p-nitroanilide
  • the initial rate of release of p-nitroaniline measured at 405 nM is directly proportional to the Factor Xa activity and thus to the Factor VIII activity in the sample.
  • the chromogenic assay is recommended by the Factor VIII and Factor IX Subcommittee of the Scientific and Standardization Committee (SSC) of the International Society on Thrombosis and Hemostatsis (ISTH). Since 1994, the chromogenic assay has also been the reference method of the European Pharmacopoeia for the assignment of FVIII concentrate potency.
  • the chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions), when the Factor VIII activity is measured in vitro by a chromogenic assay.
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions
  • the chimeric polypeptide comprising single chain Factor VIII of this invention has a Factor Xa generation rate comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • activated Factor IX hydrolyses one arginine-isoleucine bond in Factor X to form Factor Xa in the presence of Ca2+, membrane phospholipids, and a Factor VIII cofactor. Therefore, the interaction of Factor VIII with Factor IX is critical in coagulation pathway.
  • the chimeric polypeptide comprising single chain factor VIII can interact with Factor IXa at a rate comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • Factor VIII is bound to von Willebrand Factor while inactive in circulation.
  • Factor VIII degrades rapidly when not bound to vWF and is released from vWF by the action of thrombin.
  • the chimeric polypeptide comprising single chain Factor VIII binds to von Willebrand Factor at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • Factor VIII can be inactivated by activated protein C in the presence of calcium and phospholipids.
  • Activated protein C cleaves Factor VIII heavy chain after Arginine 336 in the Al domain, which disrupts a Factor X substrate interaction site, and cleaves after Arginine 562 in the A2 domain, which enhances the dissociation of the A2 domain as well as disrupts an interaction site with the Factor IXa. This cleavage also bisects the A2 domain (43 kDa) and generates A2-N (18 kDa) and A2-C (25 kDa) domains. Thus, activated protein C can catalyze multiple cleavage sites in the heavy chain.
  • the chimeric polypeptide comprising single chain Factor VIII is inactivated by activated Protein C at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • the chimeric polypeptide comprising single chain Factor VIII has
  • Factor VIII activity in vivo comparable to a chimeric polypeptide comprising processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • the chimeric polypeptide comprising single chain Factor VIII is capable of protecting a HemA mouse at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions) in a HemA mouse tail vein transection model.
  • processed Factor VIII e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions
  • the term "comparable” as used herein means a compared rate or level resulted from using the chimeric polypeptide is equal to, substantially equal to, or similar to the reference rate or level.
  • the term “similar” as used herein means a compared rate or level has a difference of no more than 10% or no more than 15%> from the reference rate or level (e.g., FXa generation rate by a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions).
  • the term “substantially equal” means a compared rate or level has a difference of no more than 0.01%, 0.5%) or 1%) from the reference rate or level.
  • the present invention further includes a composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%>, about 40%>, about 50%>, about 60%>, about 70%, about 80%, about 85%, about 90%, about 95%, or about 99% of the chimeric polypeptide comprises a Factor VIII portion, which is single chain Factor VIII and a second portion.
  • about 30%>, about 40%>, about 50%>, about 60%>, about 70%>, about 80%>, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% of the chimeric polypeptide in the composition is single chain Factor VIII.
  • the second portion is an Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C- terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof.
  • CTP 28 amino acid C- terminal peptide (CTP) of hCG with its 4 O-glycans
  • PEG polyethylene glycol
  • HES hydroxyethyl starch
  • albumin binding polypeptide albumin-binding small molecules, or two or more combinations thereof.
  • the composition of the present invention comprises a combination of a chimeric polypeptide comprising processed Factor VIII and a chimeric polypeptide comprising single chain Factor VIII, (a) wherein about 30% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 70%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (b) wherein about 40%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 60%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (c) wherein about 50%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 50%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (d) wherein about 60%) of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 40%o of the Factor VIII portion of the chimeric polypeptide being processed Factor VIII;
  • the composition of the present invention has Factor VIII activity comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions), when the Factor VIII activity is measured in vitro by a chromogenic assay.
  • processed Factor VIII e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions
  • the composition of the invention has a Factor Xa generation rate comparable to a composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • the composition comprising single chain factor VIII can interact with Factor IXa at a rate comparable to a composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc).
  • the single chain Factor VIII in the chimeric polypeptide of the present composition is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide of a composition (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • the composition comprising single chain Factor VIII has Factor VIII activity in vivo comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
  • processed Factor VIII e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions.
  • the composition comprising single chain Factor VIII of the invention is capable of protecting HemA mouse at a level comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions) in HemA mouse tail vein transection model.
  • processed Factor VIII e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions
  • the present invention further provides a method for treating a bleeding condition in a human subject using the composition of the invention.
  • An exemplary method comprises administering to the subject in need thereof a therapeutically effective amount of a pharmaceutical composition/formulation comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 85%, about 90%), about 95%), or about 99%> of the chimeric polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion.
  • the bleeding condition can be caused by a blood coagulation disorder.
  • a blood coagulation disorder can also be referred to as a coagulopathy.
  • the blood coagulation disorder, which can be treated with a pharmaceutical composition of the current disclosure is hemophilia or von Willebrand disease (vWD).
  • vWD von Willebrand disease
  • the blood coagulation disorder, which can be treated with a pharmaceutical composition of the present disclosure is hemophilia A.
  • the type of bleeding associated with the bleeding condition is selected from hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intraabdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, and bleeding in the illiopsoas sheath.
  • the subject suffering from bleeding condition is in need of treatment for surgery, including, e.g., surgical prophylaxis or peri-operative management.
  • the surgery is selected from minor surgery and major surgery.
  • exemplary surgical procedures include tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, joint replacement surgery (e.g., total knee replacement, hip replacement, and the like), heart surgery, and caesarean section.
  • the subject is concomitantly treated with FIX.
  • the compounds of the invention are capable of activating FIXa, they could be used to pre-activate the FIXa polypeptide before administration of the FIXa to the subject.
  • the methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
  • compositions comprising at least 30% of single chain Factor VIII may be formulated for any appropriate manner of administration, including, for example, topical (e.g., transdermal or ocular), oral, buccal, nasal, vaginal, rectal or parenteral administration.
  • topical e.g., transdermal or ocular
  • oral e.g., buccal
  • nasal e.g., vaginal
  • rectal e.g., parenteral administration.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intracranial, intrathecal, intraocular, periocular, intraorbital, intrasynovial and intraperitoneal injection, as well as any similar injection or infusion technique
  • the composition can be also for example a suspension, emulsion, sustained release formulation, cream, gel or powder.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • the pharmaceutical formulation is a liquid formulation, e.g., a buffered, isotonic, aqueous solution.
  • the pharmaceutical composition has a pH that is physiologic, or close to physiologic.
  • the aqueous formulation has a physiologic or close to physiologic osmolarity and salinity. It can contain sodium chloride and/or sodium acetate.
  • the composition of the present invention is lyophilized.
  • the coding sequence of human FVIII (Genbank accession number NM 000132), including its native signal sequence, was obtained by reverse transcription-polymerase chain reactions (RT-PCR) from human liver polyA RNA. Due to the large size of FVIII, the coding sequence was obtained in several sections from separate RT-PCR reactions, and assembled through a series of PCR reactions, restriction digests and ligations into an intermediate cloning vector containing a B domain deleted (BDD) FVIII coding region with a fusion of serine 743 (S743) to glutamine 1638 (Q1638), eliminating 2682 bp from the B domain of full length FVIII.
  • BDD B domain deleted
  • S743 serine 743
  • Q1638 glutamine 1638
  • the human IgGl Fc sequence (e.g., GenBank accession number Y14735) was obtained by PCR from a leukocyte cDNA library, and the final expression cassette was made in such a way that the BDD FVIII sequence was fused directly to the N-terminus of the Fc sequence (hinge, CH2 and CH3 domains, beginning at D221 of the IgGl sequence, EU numbering) with no intervening linker.
  • the mouse IgK (kappa) light chain signal sequence was created with synthetic oligonucleotides and added to the Fc coding sequence using PCR to enable secretion of this protein product.
  • the FVIIIFc and Fc chain coding sequences were cloned into a dual expression vector, pBudCE4.1 (Invitrogen, Carlsbad, CA).
  • HEK 293H cells (Invitrogen, Carlsbad, CA) were transfected with the pSYN-FVIII-013 plasmid using Lipofectamine transfection reagent (Invitrogen, Carlsbad, CA)), and a stable cell line was selected with zeocin.
  • Cells were grown in serum free suspension culture, and rFVIIIFc protein purified from clarified harvest media using a four column purification process, including a FVIII-specific affinity purification step (McCue J. et ah, J. Chromatogr. A., 1216(45): 7824-30 (2009)), followed by a combination of anion exchange columns and a hydrophobic interaction column.
  • rFVIIIFc Processed recombinant FVIII-Fc (rFVIIIFc) is synthesized as two polypeptide chains, one chain consisting of BDD-FVIII (S743-Q1638 fusion, 1438 amino acids) fused to the Fc domain (hinge, CH2 and CH3 domains) of IgGl (226 amino acids, extending from D221 to G456, EU numbering), for a total chain length of 1664 amino acids, the other chain consisting of the same Fc region alone (226 amino acids).
  • a third band was also detected at approximately 220 kDa, consistent with the predicted molecular weight for single chain FVIIIFc (SC FVIIIFc; HC+LCFc2), in which the arginine residue at position 754 (1648 with respect to the full length sequence) is not cleaved during secretion.
  • SC FVIIIFc single chain FVIIIFc
  • major bands were seen migrating at approximately 25 kDa, 90 kDa, 105 kDa, and 195 kDa, consistent with the predicted molecular weights for the single chain Fc, HC, LCFc, and SC FVIIIFc ( Figure 2B, lane 3).
  • Cotransfection with human PC5 a member of the proprotein convertase subtlisin/kexin (PCSK) type proteases, resulted in full processing of the rFVIIIFc product ( Figure 2A, B, lane 2).
  • rFVIIIFc structure was further analyzed by thrombin cleavage, reduction, and analysis by
  • the four Factor VIII fragments generated by thrombin can be detected by UV absorbance (Figure 2C), corresponding to the following segments of the protein: Fc (peak 1), light-chain-Fc (peak 2); the Al domain from the heavy chain (peak 3) and the A2 domain from the heavy chain (peak 4).
  • the 14 amino acid B domain linker and ⁇ 6 kDa a3 -related peptides are not detected by UV absorbance due to their small size.
  • FVIII samples were passed through Detergent-OUTDTG-100X columns (GBioscience, Maryland Heights, MO) for the removal of Tween, fully digested with thrombin, reduced and analyzed either by RP-HPLC-UV (POROS Rl/10, Applied Biosystems) or RP- HPLC-MS (Agilent 1200 coupled to an Agilent 6210TOF mass spectrometer) using gradients of acetonitrile in water + 0.1 % formic acid . Peptide sequence was also confirmed with LysC peptide mapping, analyzed by RP-HPLC/MS (Thermo Finnigan LTQ-XL-ETD).
  • TIC total ion current
  • Figure 2D the total ion current (TIC) chromatogram of rFVIIIFc
  • Figure 2C the UV chromatogram
  • Five of the expected six products can be detected by LC/MS, including two forms of the a3 acidic region generated from the processed and single chain isoforms, as well as the thrombin used for the digestion.
  • An additional truncated form of the a3 acidic region was also observed, and is described more fully below.
  • rBDD FVIII yielded a similar TIC chromatogram, but without the free Fc chain and having a different mass for the LC compared to the rFVIIIFc LC-Fc, consistent with the lack of an Fc region (data not shown).
  • the 6 kDa N-terminal peptide released from the LC after cleavage of R1689 is predicted to comprise the a3 acidic region (amino acids El 649 to R1689) if derived from the processed isoform, and the 14 amino acid truncated B domain fused to the a3 acidic region if derived from the single chain isoform.
  • Both rFVIIIFc and rBDD FVIII were found to contain both forms of the a3 region, proportional to the expected levels based on SDS-PAGE analysis.
  • both proteins contained a truncated form of the a3 region corresponding to amino acids D1658-R1689, as has been reported for other FVIII products, though this was found in greater abundance in rBDD FVIII than in rFVIIIFc.
  • the A2 domain contains three potential tyrosine sulfation sites, but no glycosylation sites that could result in complex heterogeneity, and therefore the exact masses of this region can be calculated.
  • rFVIIIFc the mass of the S373 to R740 sequence
  • two additional forms were identified corresponding to known truncations of the FVIII HC, correlating with an A2 domain truncated at E720 and Y729. These reported truncated forms were also observed directly in the deconvoluted spectrum of rBDD FVIII A2 ( Figure 2E).
  • Both rFVIIIFc and rBDD FVIII A2 contained similar relative amounts of the form truncated at Y729 while the rBDD FVIII A2 domain contained a notably greater level of the form truncated at E720 as compared to the same form in rFVIIIFc ( Figure 2E).
  • rFVIIIFc The primary sequence of rFVIIIFc was confirmed by peptide mapping with lysyl endopeptidase (Lys-C) digests followed by UV and mass spectrometric detection. Of the 99 theoretical peptides produced from rFVIIIFc, 81 were detected, corresponding to 98% of the total sequence. The posttranslational modifications of rFVIIIFc were also characterized by this method. FVIII contains 6 potential tyrosine sulfation sites, corresponding to positions 346, 718, 719, 723, 1664, and 1680.
  • BDD FVIII also contains 6 potential N- glycosylation sites, four of which have been reported to be glycosylated in recombinant FVIII products. Consistent with this, rFVIIIFc was found to have the same 4 sites glycosylated; N239 and N2118 were found to contain high mannose structures, while N41 and N1810 were found to contain more complex carbohydrates, similar to those found on rBDD FVIII.
  • Fc region N- linked glycosylation was found to match the GO, Gl , and G2 isoforms found with the thrombin map by LC/MS.
  • FVIII has been reported to have O-glycosylation sites at Ser 741 and 743 that are partially occupied, and this was found to be the case with rFVIIIFc as well.
  • SC FVIIIFc 15-25% single chain FVIIIFc (SC FVIIIFc), which differs from processed rFVIIIFc by a single peptide bond between R754 and E755 (R1648/E1649 with respect to the full length FVIII).
  • This isoform was purified and characterized in all of the biochemical assays described above, and found to be comparable to rFVIIIFc as shown below.
  • the activity of purified single chain FVIIIFc was found to be similar to rFVIIIFc in a chromogenic assay as well as by the various functional assays described below.
  • FVIII activity was measured by a FVIII chromogenic assay.
  • the average specific activity from four separate batches of rFVIIIFc was found to be 9762 ⁇ 449 IU/mg by the chromogenic assay, corresponding to 2148 ⁇ 99 IU/nmol.
  • the average specific activity from fourteen separate batches of rFVIIIFc was found to be 8460 ⁇ 699 IU/mg by the aPTT assay, and 9348 ⁇ 1353 IU/mg by the chromogenic assay, corresponding to 1861 ⁇ 154 and 2057 ⁇ 298 IU/nmol, respectively.
  • FVIII activity of single chain FVIILFc was also measured by the chromogenic assay and compared to the completely processed rFVllLFc or rFVllLFc DS (containing about 25% single chain rFVllLFc).
  • Table 3A shows, single chain rFVIIIFc showed no significant difference in FVIII activity compared to the Factor VIII activity of completely processed FVIIIFc or rFVIIIFc DS by the chromogenic assay, both in the presence and the absence of von Willebrand Factor (VWF).
  • Table 3B shows that full activity of SCrFVIIIFc, as measured by one- stage activated partial thromboplastin time (aPTT) assay, was observed in the absence of VWF. TABLE _3A_FVIII Activity by Chromogenic Assay
  • FVIII activity was also measured in the context of the Xase complex, by incubating activated FIX and thrombin-activated REFACTO® or rFVIIIFc protein on a phospholipid surface in the presence of calcium, and monitoring the conversion of FX to FXa as measured by cleavage of a chromogenic or fluorogenic substrate, from which FXa generation rates were determined. This assay was then modified by varying one component of the assay while keeping the others constant in order to examine the interactions with each individual component.
  • the FXa generation rate was determined as a function of varying phospholipid concentrations for rFVIIIFc DS, rBDD FVIII, and single chain rFVIIIFc ( Figure 3A), using synthetic phospholipid vesicles (25% phosphotadyl serine/75%) phosphotadyl choline). Both proteins were found to have a similar activity profile, with peak activity at approximately 156 ⁇ phospholipids.
  • FVIII is inactivated by cleavage by activated protein C (APC), as well as by dissociation of the A2 domain.
  • APC activated protein C
  • rFVIIIFc and rBDD FVIII were both activated by thrombin, then incubated with APC for different times and activity determined in a FXa generation assay ( Figure 4). In the absence of thrombin activation, little FXa generation was detected, and this was increased significantly with thrombin digestion. Treatment with APC for 90 min led to a significant decrease in FXa generation rates, similar to non-activated samples, and these results were similar for rFVIIIFc DS, rBDD FVIII, and single chain rFVIIIFc.
  • rFVIIIFc Recombinant B-domain deleted Factor VIIIFc
  • HC FVIII heavy chain
  • LC FVIII light chain
  • SC rFVIIIFc single chain isoform of rFVIIIFc
  • vWF Willebrand factor
  • Interactions with vWF were measured by real-time biomolecular interaction analysis (BIAcore), based on surface Plasmon resonance (SPR) technology, to determine the kinetics of binding of rFVIIIFc and SC rFVIIIFc towards vWF (Table 11 and Figure 16A).
  • BiAcore real-time biomolecular interaction analysis
  • SPR surface Plasmon resonance
  • FVIII-free human plasma derived vWF was immobilized by amine coupling on the surface of a biosensor at levels low enough to prevent mass transport limitation.
  • rFVIIIFc and SC rFVIIIFc were sequentially injected in single-cycle kinetics mode at concentrations ranging from 0.13 to 5.0 nM. Sensorgram data was fit to a 1 : 1 interaction model.
  • thrombin-mediated release of rFVIIIFc, SC rFVIIIFc, and B-domain deleted FVIII lacking Fc moieties was measured at both 25° C and 37° C.
  • Human vWF was immobilized by amine coupling at similar levels on three flow cells on the biosensor surface. The remaining flow cell served as a blank for reference purposes.
  • the rFVIIIFc and SC rFVIIIFc proteins were captured by vWF and allowed to slowly disassociate, and the concentrations of rFVIIIFc and SC rFVIIIFc were adjusted to obtain equimolar capture levels by the end of the dissociation phase.
  • Human a-thrombin solutions were prepared by 2-fold serial dilution and applied at concentrations that ranged from 0.005 to 20 U/mL, resulting in proteolytic release of FVIIIa species from vWF.
  • Thrombin mediated release of activated rFVIIIFc, SC rFVIIIFc, and rBDD FVIII from vWF was monitored in real-time using an SPR-based optical biosensor (Figure 16B). Following blank reference subtraction (Figure 16C), the release rate as a function of a- thrombin concentration was determined ( Figure 16D).
  • the thrombin half maximal effective concentration (EC 50 ) for SC rFVIIIFc was 12 ⁇ 1 U/mL compared to 3.9 ⁇ 0.3 U/mL for rFVIIIFc at 25° C and was 15 ⁇ 1 U/mL for SC rFVIIIFc compared to 4.8 ⁇ 0.2 U/mL for rFVIIIFc at 37° C ( Figure 16E).
  • rFVIIIFc had a similar thrombin EC 50 value compared to rBDD FVIII, having values of 3.9 ⁇ 0.3 U/mL and 3.3 ⁇ 0.3 U/mL, respectively at 25° C and values of 4.8 ⁇ 0.2 U/mL and 4.0 ⁇ 0.2 U/mL, respectively at 37° C.
  • SC rFVIIIFc had a thrombin EC 50 value that was approximately 3-fold higher than rFVIIIFc. This impairment of thrombin mediated release from vWF may underlie the specific reduction in specific activity for SC rFVIIIFc observed in the aPTT assay in which vWF was present (Table 3B).
  • each subject For the first three (3) subjects dosed with 25 IU/kg of rFVIIIFc, each subject underwent an inhibitor assessment at 14- days (336 hours) post-injection of rFVIIIFc. Dosing of the next subject (for the first three subjects only) occurred once the inhibitor testing is completed. After the 3rd subject completed the 14 day inhibitor assessment, the remaining three subjects at 25 IU/kg and the six subjects at 65 IU/kg began enrollment sequentially at least 1 day apart within each dose group.
  • each subject in the 65 IU/kg cohort received a single 65 IU/kg dose of ADVATE® followed by a 4-day (96 hours) PK profiling then crossover and receive a 65 IU/kg single, open-label dose of rFVIIIFc for a 10-day (240 hours) profiling. If a bleeding episode occurred before the first injection of rFVIIIFc in any cohort, subject's pre-study FVIII product was used for treatment and an interval of at least 4 days had to pass before receiving the first injection of rFVIIIFc for the PK profile.
  • FVIIIFc had about a 50% increase in systemic exposure (AUC INF ), about 50% reduction in clearance (CI), and about 50-70%) increase in elimination half-life and MRT compared to ADVATE ® (full length rFVIII).
  • AUC INF systemic exposure
  • CI reduction in clearance
  • FVIIIFc showed increased CI 68, TBLP1, TBLP3, and TBLP5 values compared to ADVATE ® .
  • Beta HL Elimination phase half-life also referred to as ti ⁇ p
  • a recombinant B-domain-deleted factor VIII-Fc (rFVIIIFc) fusion protein has been created as an approach to extend the half-life of FVIII.
  • the pharmacokinetics (PK) of rFVIIIFc were compared to rFVIII in hemophilia A mice. We found that the terminal half-life was twice as long for rFVIIIFc compared to rFVIII.
  • the PK were evaluated in FcRn knockout and human FcRn transgenic mice.
  • ROTEM ® rotation thromboelastometry
  • rFVIIIFc showed significantly improved CT at 72 and 96 hr post dose, and CFT and a-angle were also improved at 96 hrs compared to both XYNTHA® (FVIII) and ADVATE® (FVIII), consistent with prolonged PK of rFVIIIFc. Therefore construction of an Fc fusion of FVIII produces a molecule with a defined mechanism of action that has an increased half-life and the potential to provide prolonged protection from bleeding.
  • This Example presents final analysis results for FVIII activity from 16 patients treated with 25 and 65 IU/kg FVIII products. See Example 3.
  • rFVIIIFc is a recombinant fusion protein comprised of a single molecule of recombinant B-domain deleted human FVIII (BDD-rFVIII) fused to the dimeric Fc domain of the human IgGl, with no intervening linker sequence.
  • This protein construct is also referred to herein as rFVIIIFc heterodimeric hybrid protein, FVIIIFc monomeric Fc fusion protein, FVIIIFc monomer hybrid, monomeric FVIIIIFc hybrid, and FVIIIFc monomer-dimer. See Example 1, Fig. 1 , and Table 2A.
  • the primary objective of this study was to assess the safety and tolerability of single administration of two doses of rFVIIIFc (25 and 65 IU/kg) in previously treated patients (PTPs) aged 12 and above with severe hemophilia A.
  • the secondary objectives were to determine the pharmacokinetics (PK) parameters determined by pharmacodynamic (PD) activity of FVIII over time after a single administration of 25 or 65 IU/kg of rFVIIIFc compared to ADVATE ® in one-stage clotting and chromogenic assays.
  • PK pharmacokinetics
  • PD pharmacodynamic
  • TBLP1 Model-predicted time after dose when FVIII activity has declined to approximately 1 IU/dL above baseline.
  • TBLP3 Model-predicted time after dose when FVIII activity has declined to approximately 3 IU/dL above baseline
  • KV_M Cmax_M/Actual Dose (IU/kg)
  • KV OB Cmax OB/Actual Dose (IU/kg)
  • Total systemic FVIII exposure (assessed by AUC ) was ⁇ 48% and 61% greater following rFVIIIFc administration than ADVATE ® at 25 and 65 IU/kg dose levels, respectively.
  • Mean ( ⁇ SD) model-predicted AUC values were 974 ⁇ 259 and 1810 ⁇ 606 hr*IU/dL for ADVATE ® and 1440 ⁇ 316 and 2910 ⁇ 1320 hr*IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • ADVATE ® Mean ( ⁇ SD) model-predicted MRT values were 17.1 ⁇ 4.29 and 14.9 ⁇ 4.38 hr for ADVATE ® and 25.9 ⁇ 5.60 and 26.5 ⁇ 10.1 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. MRT values appeared to be dose-independent over the dose range evaluated for both FVIII products.
  • V values were comparable between ADVATE ® and rFVIIIFc with mean ( ⁇ SD) model-predicted V values of 43.9 ⁇ 4.27 and 56.1 ⁇ 13.4 mL/kg for ADVATE ® and 45.3 ⁇ 7.23 and 61.6 ⁇ 10.6 mL/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • Slight increases in mean CL and V values were noted with increasing dose of ADVATE ® and rFVIIIFc; however, the increase in standard deviations at the 65 IU/kg dose coupled with limited dose levels confounded an assessment of the dose-dependency of these parameters.
  • the CV% geometric mean CL value for the rFVIIIFc treatment group increased from 23.0% (25 IU/kg) to 48.6% (65 IU/kg).
  • secondary PK parameters e.g. K-values, IVR, etc.
  • K-values e.g. K-values, IVR, etc.
  • IVR and K-values for ADVATE ® and rFVIIIFc appeared to be comparable.
  • a slight increase in TBLP1 and TBLP3 values were observed with increasing dose of ADVATE ® and rFVIIIFc.
  • slight decreases in mean IVR and K-values were noted with increasing dose of ADVATE ® and rFVIIIFc.
  • an assessment of the dose dependency of these parameters is confounded by limited dose levels.
  • Mean ( ⁇ SD) observed TBLP3 were 2.06 ⁇ 0.527 and 2.26 ⁇ 0.666 IU/dL per IU/kg for ADVATE ® and 3.09 ⁇ 0.623 and 3.93 ⁇ 1.59 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • Mean IVR and K-values calculated using observed Cmax values were generally greater than values determined using model- predicted Cmax values; consistent with slight underestimation of the observed peak activity using the one-compartment model.
  • Mean ( ⁇ SD) observed K-values were 2.57 ⁇ 0.198 and 2.13 ⁇ 0.598 IU/dL per IU/kg for ADVATE ® and 2.46 ⁇ 0.330 and 1.85 ⁇ 0.332 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • Total systemic FVIII exposure (assessed by AUC ) was ⁇ 53% and 84% greater following rFVIIIFc administration than ADVATE ® at 25 and 65 IU/kg dose levels, respectively.
  • Mean ( ⁇ SD) model-predicted AUC values were 1080 ⁇ 236 and 2320 ⁇ 784 hr*IU/dL for ADVATE ® and 1650 ⁇ 408 and 4280 ⁇ 1860 hr*IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • ADVATE ® Mean ( ⁇ SD) model-predicted MRT values were 15.3 ⁇ 2.86 and 14.8 ⁇ 4.72 hr for ADVATE ® and 23.4 ⁇ 4.22 and 27.3 ⁇ 11.4 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. MRT values appeared to be dose-independent over the dose range evaluated for both FVIII products.
  • V values were comparable between ADVATE ® and rFVIIIFc with mean ( ⁇ SD) model-predicted V values of 35.8 ⁇ 5.52 and 43.6 ⁇ 11.2 mL/kg for ADVATE ® and 35.9 ⁇ 6.65 and 42.7 ⁇ 8.91 mL/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • Increases in mean CL and V values were noted with increasing dose of ADVATE ® and rFVIIIFc; however, the increase in standard deviations at 65 IU/kg coupled with limited dose levels confounded an assessment of the dose-dependency of these parameters.
  • secondary PK parameters e.g. K-values, IVR, etc.
  • K-values e.g. K-values, IVR, etc.
  • IVR and K-values for ADVATE ® and rFVIIIFc appeared to be comparable.
  • Mean IVR and K- values calculated using observed Cmax values were generally greater than values determined using model- predicted Cmax values; consistent with slight underestimation of the observed peak activity using the one -compartment model.
  • Mean ( ⁇ SD) observed K-values were 3.08 ⁇ 0.429 and 2.85 ⁇ 0.721 IU/dL per IU/kg for ADVATE ® and 3.12 ⁇ 0.451 and 2.92 ⁇ 0.985 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
  • PK parameters obtained from the Chromogenic Assay results generally agreed with those from the One-Stage Assay, except that the Chomogenic Assay yielded a higher estimation of exposure parameters (e.g., Cmax, AUCINF, etc.).
  • exposure parameters e.g., Cmax, AUCINF, etc.
  • A-LONG is an open label, multi-center evaluation of the safety, pharmacokinetics, and efficacy of recombinant Factor VIII Fc fusion (FVIILFc) in the prevention and treatment of bleeding in previously treated subjects with severe hemophilia A (defined as ⁇ 1 IU/dL [ ⁇ 1%] endogenous FVIII).
  • FVIILFc Factor VIII Fc fusion
  • Approximately 106 subjects will be enrolled into one of three regimens: a tailored prophylaxis regimen (arm 1), a weekly dosing regimen (arm 2), and an on-demand regimen (arm 3).
  • arm 1 a tailored prophylaxis regimen
  • arm 2 a weekly dosing regimen
  • arm 3 an on-demand regimen
  • Arm 1 will include an overall group and a PK subgroup. Approximately 66 subjects will be enrolled. The initial regimen will be twice weekly at 25 IU/kg on the first day, followed by 50 IU/kg on the fourth day of the week. Subjects will administer rFVIIIFc on this weekly prophylaxis regimen until PK results for rFVIIIFc are available. Based on these results, a tailored prophylaxis regimen will be established for each individual, in which the dose and interval will be determined to maintain a trough level of 1-3% FVIII activity. Each subject will then administer his individually tailored prophylaxis regimen throughout the study.
  • rFVIIIFc PK profiling Approximately 20 subjects will be enrolled/randomized and undergo abbreviated rFVIIIFc PK profiling as follows: Washout of at least 96 hours; a single dose of rFVIIIFc 65 IU/kg; Abbreviated sampling beginning on rFVIIIFc Day 0, including pre-injection and 10 ( ⁇ 2) minutes, 3 hours ( ⁇ 15 minutes), 72 ( ⁇ 2) hours [Day 3], and 96 ( ⁇ 2) hours [Day 4] from the start of injection. Following the abbreviated PK profiling, subjects will then administer a fixed dose of 65 IU/kg rFVIIIFc every 7 days at least for 28 weeks and up to 52 weeks. Arm 3: On-demand Regimen
  • Major surgery is defined as any surgical procedure (elective or emergent) that involves general anesthesia and/or respiratory assistance in which a major body cavity is penetrated and exposed, or for which a substantial impairment of physical or physiological functions is produced (e.g., laparotomy, thoracotomy, craniotomy, joint replacement, and limb amputation).
  • rFVIIIFc For prophylaxis during surgery, subjects will be treated with 20 to 50 IU/kg rFVIIIFc every 12 to 24 hours. Prior to surgery, the physician will review the subject's rFVIIIFc PK profile and assess the dose regimen of Factor VIII replacement generally required for the type of planned surgery and the clinical status of the subject. Recommendation for the appropriate dosing of rFVIIIFc in the surgical treatment period, including any rehabilitation time, will take these factors into consideration.
  • the primary objectives of this study are (a) to evaluate the safety and tolerability of rFVIIIFc administered as prophylaxis, on-demand, and surgical treatment regimens; and (b) to evaluate the efficacy of rFVIIIFc administered as prophylaxis, on-demand, and surgical treatment regimens.
  • the secondary objectives of this study are (a) to characterize the PK profile of rFVIIIFc and compare the PK of FVIIIFc with the currently marketed product, AD V ATE ® ; (b) to evaluate individual responses with FVIIIFc; and (c) to evaluate FVIIIFc consumption.
  • rFVIIIFc and AD V ATE ® appear to be comparably active in clot formation when spiked into subjects' blood prior to rFVIIIFc treatment.
  • the clotting time (CT) was linear with respect to the dose of rFVIIIFc and AD V ATE ® in the range of approximately 1% of 100% of normal, and the dose response was comparable between rFVIIIFc and ADVATE ® in the same subject.
  • mice HemA mice were used for tail clip studies. The mice were first anesthetized and then injected with 4.6 ⁇ g/kg, 1.38 ⁇ g/kg, or 0.46 ⁇ g/kg of either processed rFVIIIFc (Drug Substance, which contain about 75%-85% processed rFVIIIFc) and purified single chain rFVIIIFc. After the injection, the tail was cut from the tip and immediately placed into a tube to collect blood. Percentage of protection on survival was measured for rFVIIIFc processed (drug substance) and single chain FVIIIFc as shown in Table 7 and Figure 7.
  • processed rFVIIIFc Drug Substance, which contain about 75%-85% processed rFVIIIFc
  • purified single chain rFVIIIFc purified single chain rFVIIIFc
  • ROTEM Thromboelastometry
  • the clot was initiated by the addition of CaCl 2 (NATEM) and clotting time (CT), clot formation time (CFT), alpha-angle and maximum clot firmness (MCF) were recorded on the ROTEM system (Pentapharm GmbH, Kunststoff, Germany).
  • the clotting time (CT), clot formation time (CFT), and alpha angle for the three proteins spiked in HemA mouse blood at escalating doses from 0.1 - 100 % of normal FVIII levels are shown in Figure 14. In the wide range of 0.1 to 100% of normal, the CT and CFT are comparable among rFVIIIFc, rBDD FVIII and rflFVIII.
  • the alpha angle is only significantly different (p ⁇ 0.05) between rFVIIIFc and rBDD FVIII at 10%.
  • Recombinant factor VIIIFc is comprised of a B domain deleted (BDD) rFVIII protein genetically fused to the Fc domain of human immunoglobulin Gl (IgGl).
  • BDD B domain deleted
  • IgGl human immunoglobulin Gl
  • HC FVIII heavy chain
  • LC+Fc light chain
  • rFVIIIFc is complexed with von Willebrand factor (VWF) and released upon activation in a manner that is indistinguishable from native FVIII.
  • VWF von Willebrand factor
  • Spontaneous dissociation of the HC and LC is thought to contribute to the loss of FVIII activity in plasma and during storage of FVIII drug products.
  • SC rFVIIIFc single chain non-processed isoform of rFVIIIFc
  • SC rFVIIIFc was purified from rFVIIIFc, which contains a fraction of the non-processed isoform. Compared to rFVIIIFc, SC rFVIIIFc showed equivalent chromogenic activity but approximately 60% reduced activity by the one stage (aPTT) assay, (Table 3A-B).
  • Thrombin generation assay (TGA) was performed using calibrated automated thrombogram (from Thrombinoscope®). In a thrombin generation assay (TGA), SC rFVIIIFc also showed a reduced thrombin potential ( Figure 13 A), and peak thrombin ( Figure 13B) compared to rFVIIIFc.
  • mice HemA male mice were treated with indicated doses of either rFVIIIFc drug product or SC rFVIIIFc 48 hours prior to TVT. Tail re -bleeding and survival were monitored hourly up to 12 hours post TVT with final observation performed at 24-hour post TVT. SC rFVIIIFc and the rFVIIIFc demonstrated equivalent in vivo efficacy in this model, with an ED50 of 1.17 ⁇ g/kg and 1.23 ⁇ g /kg respectively when TVT was performed at 48 hours post infusion (Figure 7(A)).
  • rFVIIIFc Current factor VIII (FVIII) products display a half- life (t 1/2 ) of approximately 8-12 hours, requiring frequent intravenous injections for prophylaxis and treatment of hemophilia A patients.
  • rFVIIIFc is a recombinant fusion protein composed of a single molecule of FVIII covalently linked to the Fc domain of human IgGi to extend circulating rFVIII half-life.
  • This first-in-human study in previously-treated male subjects with severe hemophilia A investigated safety and pharmacokinetics of rFVIIIFc. Sixteen subjects received a single dose of ADVATE ® at 25 or 65 IU/kg followed by an equal dose of rFVIIIFc.
  • rFVIIIFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia A.
  • Hemophilia A is an inherited bleeding disorder that results in frequent spontaneous and traumatic bleeding into the joints and soft tissues. Mannucci PM, Tuddenham EGD, N Engl J Med, 344:1773-1779 (2001). When inadequately treated, this bleeding leads to chronic arthropathy, disability, and increased risk of death. Soucie JM et al., Blood. 96(2):437-442 (2000).
  • Plasma-derived FVIII (pdFVIII) and recombinant human FVIII (rFVIII) products are utilized for treatment (on-demand therapy) and prevention (prophylaxis therapy) of bleeding episodes.
  • rFVIII was developed to reduce the risk of blood-borne pathogen transmission following the widespread contamination of plasma products with HIV and hepatitis viruses, and to secure an adequate supply of FVIII product.
  • hemostatic protection with current FVIII products is temporally limited due to a short half-life (t 1/2 ) of approximately 8-12 hours, requiring prophylactic injections three times per week or every other day for most patients in order to maintain FVIII levels above 1%, a level that has been established as protective against most spontaneous bleeding episodes.
  • t 1/2 short half-life
  • prophylactic therapy Compared to on-demand treatment, prophylactic therapy also decreases disability, hospitalization rate, and time lost from school or work; 6 ' 16 and improves quality of life for patients and their families. 17
  • prophylactic therapy often requires use of central venous access devices in children, and their attendant risks of infection, sepsis, and thrombosis.
  • acceptance of and compliance with prophylaxis decreases with age, in part because of inconvenience and invasiveness. '
  • a rFVIII product with a prolonged plasma ti 2 would potentially be of benefit. Lillicrap D., Current Opinion in Hematology 77:393-397 (2010).
  • rFVIIIFc is a recombinant fusion protein composed of a single molecule of B-domain deleted rFVIII covalently linked to the human IgGi Fc domain.
  • Potential advantages of Fc-fusion proteins include better tolerability and prolonged hemostatic protection, and the Fc domain represents a natural molecule with no known inherent toxicity.
  • 21 ' 22 Attachment to the IgGi Fc domain permits binding to the neonatal Fc receptor (FcRn), which is expressed in many cell types, including endothelial cells. FcRn expression remains stable throughout life and is responsible for protecting IgGi and Fc-fusion proteins from lysosomal degradation, thus prolonging the t 1/2 of the protein.
  • FcRn neonatal Fc receptor
  • Numerous proteins within the circulation are internalized into the cells lining the vasculature via nonspecific pinocytosis and are trafficked to endosomal and lysosomal degradation pathways.
  • Fc proteins interact with FcRn, resident within endosomes. Endosomes containing FcRn direct the Fc fusion proteins back to the plasma membrane, releasing them into circulation in a pH-dependent manner, 24 thereby avoiding lysosomal degradation.
  • This recycling approach has been used successfully to extend the t 1/2 of therapeutic biologies; a number of Fc fusion-based drugs have been approved for clinical use (eg etanercept, romiplostim) and others are in development. 25 ' 26
  • Plasma FVIII activity was measured in subjects before injection, 10 and 30 minutes, 1, 3, 6, 9, 24, 48, 72, 96, 120, and 168 hours (7 days) after rFVIIIFc injection, with additional samples at 192, 216, and 240 hours (10 days) for subjects dosed at 65 IU/kg of rFVIIIFc. Plasma FVIII activity was measured at the same time points after ADVATE ® treatment, through 72 hours for the 25 IU/kg group and 96 hours for the 65 IU/kg group.
  • Subjects Male subjects were at least 12 years of age with severe hemophilia A (defined as FVIII activity level ⁇ 1%) and had at least 100 documented prior exposure days to FVIII concentrates (pdFVIII or rFVIII). Subjects with known hypersensitivity to mouse or hamster protein, history of inhibitor or detectable inhibitor titer at screening, or who were taking any medications that could affect hemostasis or systemic immunosuppressive drugs, or who experienced an active bacterial or viral infection (other than hepatitis or HIV) within 30 days of screening were excluded. Subject's genotype was recorded at study entry, when known.
  • Treatment Product The human rFVIIIFc and Fc transgenes were stably transfected into
  • the purified drug product is composed of a monomeric B- domain-deleted FVIII covalently linked through its carboxy-terminus to the N-terminus of an Fc monomer, which forms a disulfide bond with a second Fc monomer during synthesis and secretion from the cells.
  • rFVIIIFc was purified by chromatography and nanofiltration, and was fully active in one-stage and chromogenic clotting assays relative to commercially available rFVIII preparations.
  • FVIII activity was measured by the one-stage clotting (aPTT) assay on a Siemens
  • FVIII inhibitors were measured by the - *Nijmegen-modified Bethesda assay and less than 0.6 BU/mL was considered negative.
  • Anti- rFVIIIFc antibodies were assessed using a specific bridging electrochemiluminescent immunoassay which uses biotin and sulfo-tagged rFVIIIFc. Assay sensitivity was determined to be 89 ng/mL using an anti-human FVIII monoclonal antibody as a surrogate control. Exploratory whole blood rotation thromboelastometry (ROTEM ® ) was performed in two subjects, one from each dose level, at various time points to assess the improvement in global hemostasis following injection with ADVATE ® and rFVIIIFc.
  • ROTEM ® Exploratory whole blood rotation thromboelastometry
  • Statistical Analyses Selected PK parameters for rFVIIIFc and ADVATE ® were compared using an analysis of variance model. PK parameters were log-transformed for these analyses and estimated means, mean differences, and confidence intervals on the log-scale were transformed to obtain estimates for geometric means, geometric mean ratios (GMR), and confidence intervals, respectively, on the original scale.
  • the GMR is the geometric mean of the intra-subject ratio of the rFVIIIFc PK parameter value to the ADVATE ® PK parameter value.
  • Subject Disposition- Nineteen subjects were enrolled in the study; 16 underwent PK evaluation for both ADVATE ® and rFVIIIFc. One subject self-administered his previous product prior to completing the wash-out period following the dose with ADVATE ® and was thus excluded from the PK analysis, but was included in the safety analysis. Three subjects were discontinued from the study before receiving either study drug: one voluntarily withdrew; a second was withdrawn by the Investigator for non-compliance; and one was withdrawn at the Sponsor's request due to completion of study enrollment. Of the subjects dosed, six subjects received 25 IU/kg and 10 subjects received 65 IU/kg of both ADVATE ® and rFVIIIFc.
  • Plasma - ADVATE ® and rFVIIIFc activities were determined in the same assays using commercially available reagents and calibration against normal human plasma standards. There was a strong correlation between the results obtained by the one-stage clotting assay and the chromogenic assay in samples that had an activity above the LLOQ. Correlation coefficients (Pearson R 2 ) of 0.94 and 0.95 were observed between the two assay results for 151 samples following ADVATE ® dosing and 185 samples following rFVIIIFc dosing, respectively.
  • CI Confidence Interval
  • Geom. Mean Geometric Mean
  • OBS observed.
  • Estimated means, 95% CI for means, and mean differences were transformed to obtain estimated geometric means, 95% CI for geometric means, and geometric mean ratios, respectively.
  • CI Confidence Interval
  • Geom. Mean Geometric Mean
  • OBS observed.
  • Estimated means, 95%> CI for means, and mean differences were transformed to obtain estimated geometric means, 95%> CI for geometric means, and geometric mean ratios, respectively.
  • rFVIIIFc was well tolerated by subjects at both doses. There were no clinically significant changes observed in hematology, blood chemistry, or urinalysis parameters. The majority of AEs were mild, unrelated to rFVIIIFc, and resolved without sequelae. No serious AEs or deaths occurred during the study, and no subjects at either dose developed neutralizing or binding antibodies to rFVIIIFc.
  • ADVATE ® with ⁇ ⁇ 2 and MRT across dose levels being 1.54 to 1.71-fold longer, as measured by the one-stage (aPTT) clotting assay and 1.59 to 1.84-fold longer by the two-stage chromogenic assay.
  • the prolonged activity of rFVIIIFc predicts possible prolonged efficacy, allowing for a less frequent dosing regimen in the prophylactic treatment of patients with Hemophilia A.
  • this Phase l/2a clinical trial is the first trial to demonstrate the safety and prolonged t 1/2 of rFVIIIFc in patients with severe hemophilia A.
  • a pivotal Phase 3 study is ongoing with rFVIIIFc to establish effective prophylaxis dosing regimens for individuals with hemophilia A.
  • SC-rFVIIIFc immunoglobulin Fc domain
  • SC-rFVIIIFc purified SC counterpart
  • ⁇ -thrombin EC 50 values for rFVIIIFc and rBDD-FVIII were comparable (3.7 ⁇ 0.2 U/mL and 3.2 ⁇ 0.3 U/mL, respectively), whereas the EC 50 value for SC-rFVIIIFc was greater than 3- fold higher (11.7 ⁇ 0.9 U/mL).
  • ATGTGCACTC AGGCCTGATT GGACCCCTTC TGGTCTGCCA CACTAACACA CTGAACCCTG 3661 CTCATGGGAG ACAAGTGACA GTACAGGAAT TTGCTCTGTT TTTCACCATC TTTGATGAGA
  • AAACCATCTC CAAAGCCAAA GGGCAGCCCC GAGAACCACA GGTGTACACC CTGCCCCCAT
  • B-Domain Deleted FVIII-Fc Monomer Hybrid BDD FVIIIFc monomer dimer: created by coexpressing BDD FVIIIFc and Fc chains.
  • Construct HC-LC-Fc fusion.
  • An Fc expression cassette is cotransfected with BDDFVIII-Fc to generate the BDD FVIIIFc monomer-.
  • the Fc sequence is shown in bold; HC sequence is shown in double underline; remaining B domain sequence is shown in italics. Signal peptides are underlined.
  • Construct HC-B-LC-Fc fusion.
  • An Fc expression cassette is cotransfected with full length FVIII-Fc to generate the full length FVIIIFc monomer.
  • the Fc sequence is shown in bold; HC sequence is shown in double underline; B domain sequence is shown in italics. Signal peptides are underlined.

Abstract

The present invention provides methods of administering Factor VIII (processed FVIII, single chain FVIII, or a combination thereof); methods of administering chimeric and hybrid polypeptides comprising Factor VIII; chimeric and hybrid polypeptides comprising Factor VIII; polynucleotides encoding such chimeric and hybrid polypeptides; cells comprising such polynucleotides; and methods of producing such chimeric and hybrid polypeptides using such cells

Description

FACTOR VIII CHIMERIC AND HYBRID POLYPEPTIDES, AND METHODS
OF USE THEREOF
REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY
[0001] The content of the electronically submitted sequence listing in ASCII text file
(Name: sequencelisting_ascii.txt; Size: bytes; and Date of Creation: ) filed with the application is incorporated herein by reference in its entirety.
Field of the Invention
[0002] The present invention relates generally to the field of therapeutics for hemostatic disorders.
Background Art
[0003] Hemophilia A is an X-linked bleeding disorder caused by mutations and/or deletions in the factor VIII (FVIII) gene resulting in a deficiency of FVIII activity (Peyvandi, F. et al. Haemophilia 72:82-89 (2006). The disease is characterized by spontaneous hemorrhage and excessive bleeding after trauma. Over time, the repeated bleeding into muscles and joints, which often begins in early childhood, results in hemophilic arthropathy and irreversible joint damage. This damage is progressive and can lead to severely limited mobility of joints, muscle atrophy and chronic pain (Rodriguez-Merchan, E.C., Semin. Thromb. Hemost. 29:87-96 (2003), which is herein incorporated by reference in its entirety).
[0004] The A2 domain is necessary for the procoagulant activity of the factor VIII molecule.
Studies show that porcine factor VIII has six-fold greater procoagulant activity than human factor VIII (Lollar, P., and E. T. Parker, J. Biol. Chem. 266: 12481-12486 (1991)), and that the difference in coagulant activity between human and porcine factor VIII appears to be based on a difference in amino acid sequence between one or more residues in the human and porcine A2 domains (Lollar, P., et al., J. Biol. Chem. 267:23652-23657 (1992)), incorporated herein by reference in its entirety.
[0005] Treatment of hemophilia A is by replacement therapy targeting restoration of FVIII activity to 1 to 5 % of normal levels to prevent spontaneous bleeding (Mannucci, P.M., et al, N. Engl. J. Med. 344:1773-1779 (2001), which is herein incorporated by reference in its entirety). There are plasma-derived and recombinant FVIII products available to treat bleeding episodes on- demand or to prevent bleeding episodes from occurring by treating prophylactically. Based on the short half-life of these products, however, e.g., 8-12 hours, treatment regimens require the administration of frequent intravenous injections. Such frequent administration is painful and inconvenient.
[0006] Reduced mortality, prevention of joint damage and improved quality of life have been important achievements due to the development of plasma-derived and recombinant FVIII. Prolonged protection from bleeding would represent another key advancement in the treatment of hemophilia A patients. However, to date, no products that allow for prolonged hemostatic protection have been developed. Therefore, there remains a need for improved methods of treating hemophilia due to factor VIII deficiency that are more tolerable, longer lasting, and more effective than current therapies.
BRIEF SUMMARY OF THE INVENTION
[0007] The present invention provides methods of administering Factor VIII; methods of administering chimeric polypeptides comprising Factor VIII and hybrids of such chimeric polypeptides; chimeric polypeptides comprising Factor VIII and hybrids of such chimeric polypeptides; polynucleotides encoding such chimeric and hybrid polypeptides; cells comprising such polynucleotides; and methods of producing such chimeric and hybrid polypeptides using such cells.
[0008] The present invention provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, at a dosing interval at least about one and one -half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
[0009] The dosing interval may be at least about one and one -half to six times longer, one and one -half to five times longer, one and one-half to four times longer, one and one-half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., the Fc portion. The dosing interval may be at least about one and one -half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one-half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., the Fc portion. The dosing interval may be about every five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
[0010] The dosing interval may be at least about one and one -half to 5, one and one-half, 2, 3, 4, or 5 days or longer. [0011] The present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, to obtain an area under the plasma concentration versus time curve (AUC) at least about one and one-quarter times greater than the AUC obtained by an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion.
[0012] The present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a polypeptide comprising a Factor VIII and an Fc at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
[0013] The methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
[0014] On-demand treatment includes treatment for a bleeding episode, hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis (head trauma), gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath. The subject may be in need of surgical prophylaxis, peri-operative management, or treatment for surgery. Such surgeries include, e.g., minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
[0015] For on-demand treatment, the dosing interval of said chimeric polypeptide is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
[0016] The therapeutic doses that may be used in the methods of the invention are about 10 to about 100 IU/kg, more specifically, about 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 IU/kg, and more specifically, about 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
[0017] The therapeutic doses that may be used in the methods of the invention are about 10 to about 150 IU/kg, more specifically, about 100-110, 110-120, 120-130, 130-140, 140-150 IU/kg, and more specifically, about 110, 115, 120, 125, 130, 135, 140, 145, or 150 IU/kg.
[0018] The subject in the methods of the invention is a human subject. The determination of dosing interval and AUC may be carried out in a single subject or in a population of subjects. [0019] The Factor VIII (or Factor VIII portion of a chimeric polypeptide) is a human Factor VIII.
The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may have a full or partial deletion of the B domain.
[0020] The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90% or
95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 4 to 2351 of SEQ ID NO: 6). The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
[0021] The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90%> or
95% identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6). The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
[0022] The Fc portion (or Fc portion of a chimeric polypeptide) may be at least 90%> or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6). The Fc portion (or Fc portion of a chimeric polypeptide) may be identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
[0023] The chimeric polypeptide may comprise a sequence at least 90% or 95% identical to the
Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2). The chimeric polypeptide may comprise a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
[0024] The chimeric polypeptide may be in the form of a hybrid comprising a second polypeptide in association with said chimeric polypeptide, wherein said second polypeptide comprises or consists essentially of an Fc.
[0025] The second polypeptide may comprise or consist essentially of a sequence at least 90% or
95%) identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4). The second polypeptide may comprise or consist essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
[0026] The chimeric polypeptide or hybrid may be administered as part of a pharmaceutical composition comprising at least one excipient.
[0027] The invention also provides the above-described chimeric and hybrid polypeptides themselves, polynucleotides encoding them, a cultured human embryonic cells comprising the polynucleotides, and methods of producing such chimeric and hybrid polypeptides, and the polypeptides produced by such methods.
[0028] The present invention also provide a chimeric polypeptide that has Factor VIII activity comprising a Factor VIII portion and a second portion, wherein the Factor VIII portion is processed Factor VIII comprising two chains, a first chain comprising a heavy chain and a second chain comprising a light chain, wherein said first chain and said second chain are associated by a metal bond. For example, at least about 50%, about 60%>, about 70%, about 75%, about 80%, about 85%), about 90%>, about 95%, or about 99% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
[0029] In addition, the present invention includes a chimeric polypeptide that has Factor VIII activity, wherein the Factor VIII portion is single chain Factor VIII. In one aspect, the single chain Factor VIII can contain an intact intracellular processing site. In one embodiment, at least about 1%), about 5%, about 10%, about 15%, about 20%, or about 25% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII. In another embodiment, at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 99%) of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII. In another aspect, the single chain FVIII does not contain an intracellular processing site. For example, the SCFVIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII. The amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648. In certain embodiments, the substitution or mutation is an amino acid other than arginine, e.g., alanine.
[0030] In some embodiments, the chimeric polypeptide comprising single chain Factor VIII has
Factor VIII activity at a level comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions, when the Factor VIII activity is measured in vitro by a chromogenic assay. In other embodiments, the chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity in vivo comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions. In still other embodiments, the chimeric polypeptide comprising single chain Factor VIII has a Factor Xa generation rate comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions. In certain embodiments, single chain Factor VIII in the chimeric polypeptide is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII. In yet other embodiments, the single chain Factor VIII in the chimeric polypeptide has a Factor IXa interaction rate comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII. In further embodiments, the single chain Factor VIII in the chimeric polypeptide binds to von Willebrand Factor at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and the processed Factor VIII.
[0031] The present invention further includes a composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%>, about 50%>, about 60%>, about 70%, about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%) of said polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion, wherein said single chain Factor VIII is at least 90%> or 95%> identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6). In one embodiment, the second portion can be an Fc. In another embodiment, the polypeptide is in the form of a hybrid comprising a second polypeptide, wherein said second polypeptide consists essentially of an Fc. In other embodiments, the polypeptide has a half-life at least one and one -half to six times longer, one and one -half to five times longer, one and one-half to four times longer, one and one -half to three times longer, or one and one-half to two times longer to a polypeptide consisting of said Factor VIII.
[0032] Also provided is a method of treating a bleeding condition comprising administering a therapeutically effective amount of the composition. The treatment can be prophylactic treatment or on-demand treatment or perioperative. The bleeding coagulation disorder can be hemophilia. In one embodiment, the subject that is treated is a pediatric subject.
[0033] The present invention is also directed to a method of preventing, decreasing, or treating a bleeding episode in a subject comprising administering to the subject an effective amount of a long-acting Factor VIII (FVIII) protein, wherein the subject expresses a high level of von Willebrand Factor (VWF) in plasma. In one embodiment, the subject has been identified as expressing a high level of VWF in plasma. The present invention is also directed to a method of preventing, decreasing, or treating a bleeding episode in a subject comprising: (a) identifying a subject having high levels of VWF by measuring the level of VWF in the plasma of said subject, wherein a VWF level of at least about 100 IU/dL identifies the subject as having a high level of VWF; and (b) administering to the subject an effective amount of a long-acting FVIII protein.
[0034] In one embodiment, the subject is a human. In another embodiment, the subject is a pediatric subject. In another embodiment, the subject has hemophilia A.
[0035] In one embodiment, the high level of VWF is at least about 100 IU/dL. In another embodiment, the high level of VWF is between about 100 IU/dL and about 200 IU/dL. In another embodiment, the high level of VWF is about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
[0036] In one embodiment the subject has the blood serotype A, B, or AB.
[0037] In one embodiment, the long-acting FVIII protein has a half-life in said subject of between about 20 and about 40 hours. In another embodiment, the long-acting FVIII protein has a half-life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours. In another embodiment, the long-acting FVIII protein has a half-life of between about 20 and 27 hours. In another embodiment, the long-acting FVIII protein has a half- life that is at least about 1.2 times greater than the half- life of said said long- acting FVIII protein when administered to an individual having average levels of VWF. In another embodiment, the long-acting FVIII protein has a half-life that is at least about about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold times greater than the half-life of said said long-acting FVIII protein when administered to an individual having average levels of VWF.
[0038] In one embodiment, the effective amount of long-acting FVIII protein that is administered is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg. In another embodiment, the effective amount is at least about 65IU/kg to at least about 90IU/kg. In another embodiment, the effective amount is 80 IU/kg.
[0039] In one embodiment,, the long-acting FVIII protein is administered every 72 hours or longer. In another embodiment, the long-acting FVIII protein is administered about once a week or longer. In another embodiment, the long-acting FVIII protein is administered about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month. [0040] In one embodiment, the long-acting FVIII is administered at a dosage of 80 IU/kg once every 72 hours. In a further embodiment the long-acting FVIII is administered at a dosage of 80 IU/kg once every 72 hours to a pediatric subject.
[0041] In one embodiment, administration of the long-acting FVIII protein resolves greater than
5-20%, greater than 5-15%, greater than 5-10%, greater than 10-20%), or greater than 10-15% of bleeding episodes. In one embodiment, the trough level of plasma Factor VIILC in the subjects is maintained above 1-3 or 3-5 IU/dl. In one embodiment, the administration prevents a bleeding episode in the subject. In another embodiment, the bleeding episode is spontaneous. In another embodiment, the administration resolves greater than 80-100%), greater than 80-90%), greater than 85-90%, greater than 90-100%, greater than 90-95%, or greater than 95-100% of bleeding episodes.
[0042] In one embodiment, the administration maintains homeostatis in the population of the subjects in need of a surgery. In another embodiment, the long-acting FVIII protein is administered prior to, during, or after the surgery. In another embodiment, the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intraabdominal surgery, intrathoracic surgery, or joint replacement surgery. In another embodiment, the surgery is an emergency surgery.
[0043] In one embodiment, the long-acting FVIII protein has a half-life longer than a polypeptide consisting of FVIII. In another embodiment, the long-acting FVIII protein is pegylated, hesylated, or polysialylated.
[0044] In one embodiment, the long-acting FVIII protein is a chimeric protein comprising a
FVIII portion and a second portion. In another embodiment, the second portion is an Fc region, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof. In another embodiment, the second portion is fused to the amino-terminus or the carboxy-terminus of the FVIII portion. In another embodiment, the second portion is inserted between two amino acids in the FVIII portion. In another embodiment, the chimeric protein is a FVIIIFc monomer dimer hybrid. In another embodiment, the FVIII portion is a single chain. In another embodiment, the FVIII portion comprises a heavy chain and a light chain. In another embodiment, the FVIII portion comprises full-length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain. In another embodiment, the FVIII portion comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In another embodiment, the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In another embodiment, the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6. In another embodiment, the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6. In another embodiment, the long-acting FVIII polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
[0045] The invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising measuring the half-life of FVIII-Fc in said subject, wherein a half-life that is at least about 1.2 times greater than the half- life of FVIII-Fc in a normal subject indicates the subject is a candidate for long interval dosing, and administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days.
[0046] The invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days to a subject, wherein the half-life of FVIII-Fc in said subject is at least about 1.2 times greater than the half- life of FVIII-Fc when administered to a subject having average levels of VWF.
[0047] In one embodiment, the plasma half-life of FVIII-Fc in said subject is at least about 1.3,
1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold times greater than the plasma half-life of FVIII-Fc when administered to a subject having average levels of VWF. In another embodiment, the FVIII-Fc plasma half-life is between 20-40 hours. In another embodiment, the long-acting FVIII protein has a half- life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours. In another embodiment, the long-acting FVIII protein has a half- life of between about 20 and 27 hours.
[0048] The invention also provides a method of treating a subject diagnosed with bleeding disorder, comprising measuring the half- life of a short-acting FVIII administered to said subject, wherein a half-life that is at least about 1.2 times greater than the half-life of said short-acting FVIII in a subject having average VWF levels indicates that the subject is a candidate for long interval dosing, and administering a long-acting FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days. In one embodiment, the half-life of the short-acting FVIII in said subject is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold greater than the half-life of a short-acting FVIII when administered to a subject having average levels of VWF.
[0049] In one embodiment, the subject is a human. In another embodiment, the subject is a pediatric subject. In another embodiment, the subject has hemophilia A. In another embodiment, the subject has the blood serotype A, B, or AB. [0050] In one embodiment, the long-acting FVIII-Fc is administered in an effective amount that is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg. In another embodiment, the effective amount is at least about 65IU/kg to at least about 90IU/kg.
[0051] In one embodiment, the effective amount of the FVIII-Fc protein is administered about once every week, about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
[0052] In one embodiment, the administration resolves greater than 5-20%, greater than 5-15%, greater than 5-10%, greater than 10-20%), or greater than 10-15%) of bleeding episodes. In one embodiment, the trough level of plasma Factor VIILC in the subjects is maintained above 1-3 or 3-5 IU/dl.
[0053] In one embodiment, the administration prevents a bleeding episode in the subject. In one embodiment, the bleeding episode is spontaneous. In one embodiment, the administration resolves greater than 80-100%, greater than 80-90%, greater than 85-90%, greater than 90-100%, greater than 90-95%), or greater than 95-100%) of bleeding episodes. In one embodiment, the administration maintains homeostatis in the population of the subjects in need of a surgery. In one embodiment, the FVIII-Fc protein is administered prior to, during, or after the surgery. In one embodiment, the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery. In one embodiment the surgery is an emergency surgery.
[0054] In one embodiment, the FVIII-Fc protein has a half-life longer than a polypeptide consisting of FVIII. In one embodiment, the FVIII-Fc protein is pegylated, hesylated, or polysialylated. In one embodiment, the FVIII-Fc protein is a FVIIIFc monomer dimer hybrid. In one embodiment, the FVIII portion is a single chain. In one embodiment, the FVIII portion comprises a heavy chain and a light chain. In one embodiment, the FVIII portion comprises full- length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain. In one embodiment, the FVIII portion comprises an amino acid sequence at least 80%>, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In one embodiment, the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6. In one embodiment, the second portion of the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6. In one embodiment, the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
[0055] In one embodiment, the FVIII-Fc polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
[0056] The invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing with a long-acting FVIII polypeptide, comprising measuring the expression levels of plasma VWF, wherein an VWF expression level of at least 100 IU/dL indicates that the subject is a candidate for long interval dosing using a long- acting FVIII polypeptide. In one embodiment, the VWF expression level is at least about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
[0057] The invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half- life of FVIII-Fc in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of FVIII-Fc when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing. In one embodiment, the half-life of FVIII-Fc is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2,
2.3, 2.4, or 2.5-fold greater than the half-life of FVIII-Fc when administered to a subject having average levels of VWF.
[0058] The invention also provides a method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half-life of short-acting FVIII in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of short-acting FVIII when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing. In one embodiment, the half-life is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3,
2.4, or 2.5-fold greater than the half-life of FVIII-Fc when administered to a subject having average levels of VWF.
BRIEF DESCRIPTION OF THE DRAWINGS/FIGURES [0059] FIG. 1. Schematic Representation of rFVIIIFc monomer.
[0060] FIGS. 2A-E. A-B. Non-reducing and reducing SDS-PAGE analysis of rFVIIIFc
(processed or single chain). C. rFVIIIFc structure analyzed by LC/UV and LC/MS. D. Total Ion Current (TIC) chromatogram (LC/MS map) of rFVIIIFc after thrombin cleavage. Major digestion products are indicated. E. Deconvoluted Mass Spectrum of the A2 domain of rFVIIIFc and rBDDFVIII. Major products and their cognate masses are indicated, corresponding to thrombin- cleaved A2 domain (S373 to R740) and two truncated products, S373 to Y729 and S373 to E720.
[0061] FIGS. 3A-C. Biochemical characterization of rFVIII-Fc: A. Activation of Factor X as a function of phospholipid vesicle concentration; B. Activation of Factor X as a function of FX concentration. C. Activation of Factor X as a function of Factor IXa concentration.
[0062] FIG 4. Activation of Factor X following cleavage by activated Protein C.
[0063] FIGS. 5A-D. Observed group mean FVIII activity (±SE) versus time profiles, sorted by dose level, grouped by compound (one stage assay, 25 IU/kg (A) or 65 IU/kg (B); and chromogenic assay, 25 IU/kg (C) or 65 IU/kg (D)) versus time.
[0064] FIGS. 6A-B. Observed group mean FVIII activity (±SE) versus time profiles, grouped by dose level and compound (one stage assay (A) or chromogenic assay (B)) versus time.
[0065] FIGS. 7(A)-(C). In Vivo Efficacy of Single Chain FVIILFc in HemA Mouse Tail Vein
Transection Model. (A) Single chain rFVIILFc doses are shown as squares, and processed rFVIILFc doses are shown as circles. (B) Percent survival following tail vein transection of 4.6 μg/kg, 1.38 μg/kg, and 0.46 μg/kg of rFVIIIFc or SC rFVIIIFc. (C) Percent of non-bleeders following tail vein transection of 4.6 μg/kg (black circle or inverted triangle), 1.38 μg/kg (triangle or diamond), and 0.46 μg/kg (square and gray circle) of rFVIIIFc or SC rFVIIIFc, respectively.
[0066] FIG. 8. Study Design. Fig. 8 depicts the study design of the phase l/2a study, which was a dose-escalation, sequential design to evaluate the safety and PK of rFVIIIFc compared with AD V ATE® after a single intravenous dose of either 25 IU/kg (low dose Cohort A) or 65 IU/kg (high dose Cohort B).
[0067] FIG. 9. Correlation of rFVIII Activity by One-Stage (aPTT) and Chromogenic Assays.
Correlation between one-stage clotting (aPTT) and chromogenic assay results measuring FVIII activity (IU/mL) following injection of ADVATE® (♦) and rFVIIIFc (□).
[0068] FIGS. 10(A)-(B). Group Mean Plasma FVIII Activity Pharmacokinetic Profiles for Low-
Dose and High-Dose Cohorts. The plasma FVIII activity (one stage aPTT assay) versus time curve after a single intravenous injection of rFVIIIFc or ADVATE® are shown for (A) 25 IU/kg (low-dose cohort, n=6); and (B) 65 IU/kg (high dose cohort, n=10 [ADVATE®]; n=9 [rFVIIIFc]). Results presented are group mean ± standard error of mean (SEM).
[0069] FIGS. 11(A)-(B). Effect of VWF Antigen Levels on CI and t1/2 of FVIII Activity after
Injection of ADVATE® or rFVIIIFc. Correlation between VWF antigen levels and (A) the weight-adjusted CI of ADVATE® (R2=0.5415 and p=0.0012) and rFVIIIFc (R2=0.5492 and p=0.0016); and (B) the t1/2 of ADVATE® (R2=0.7923 and p<0.0001) and rFVIIIFc (R2= 0.6403 and p=0.0003). Each dot represents an individual subject.
[0070] FIGS. 12(A)-(B). Ex Vivo Whole Blood ROTEM® Results for Individual Subjects After
Injection of ADVATE® or rFVIIIFc. Blood was sampled from subjects prior to and after treatment at doses of (A) 25 IU/kg ADVATE and rFVIIIFc; and (B) 65 IU/kg ADVATE and rFVIIIFc at specified time points. Clotting time was determined by NATEM initiated with Ca++ on a ROTEM® instrument. Results presented are mean ± standard error of mean (SEM) from triplicate channel readings for each individual sample.
[0071] FIGS. 13(A)-(B). Activity comparison in thrombin generation assay (TGA). (A) SC rFVIIIFc showed a reduced endogenous thrombin potential (ETP), and (B) a reduced peak thrombin compared to rFVIIIFc.
[0072] FIGS. 14(A)-(C): In vitro ROTEM data. ROTEM (NATEM) results (Mean ± SD) for varying concentratios of XYNTHA, ADVATE< and rFVIIIFc spked in pooled whole blood obtained from naive HemA mice. (A). Average clot time (CT) (Fig. 14A), (B). clot formation time (CFT) , and (C). alpha angle.
[0073] FIGS. 15(A)-(C). Ex vivo ROTEM data. ROTEM (NATEM) results (Mean ± SD) from
HemA mice following a single intravenous administration of 50 IU/kg of XYNTHA, ADVATE, or rFVIIIFc at 5 min, 24, 48, 72, and 96 hours after dosing. (A). Average clot time (CT), (B). Clot formation time (CFT), and (C). alpha angle.
[0074] FIGS. 16(A)-(E): Real-time evaluation of the interaction of rFVIIIFc and single chain
(SC) rFVIIIFc with vWF, and real-time evaluation of thrombin mediated release of rFVIIIFc and SC rFVIIIFc from vWF. (A). Surface plasmon resonance (SPR) analysis of rFVIIIFc and SC rFVIIIFc affinity for vWF. Depicted are the binding curve and the 1 : 1 fit interaction model. The x-axis shows time in seconds and the y-axis shows response in response units (RU). (B). Reference subtracted sensograms of thrombin-mediated release of activated rFVIIIFc, SC rFVIIIFc, and B-domain deleted rFVIII lacking Fc moieties (rBDD FVIII) at 25° C (top) and 37° C (bottom). The x-axis shows time in seconds and the y-axis shows response in response units (RU). Individual lines indicate the response at different a-thrombin concentrations. The uppermost line is the response at 0 U/mL a-thrombin, and each subsequent line runs in order for a-thrombin concentrations of 0.005, 0.01 , 0.02, 0.04, 0.08, 0.16, 0.31 , 0.63, 1.3, 2.5, 5, 10, and 20 U/mL. (C). Double reference subtracted sensograms of thrombin mediated release phase for rFVIIIFc, SC rFVIIIFc, and rBDD FVIII at 25° C (top) and 37° C (bottom). The x-axis shows time in seconds and the y-axis shows response in response units (RU). Individual lines indicate response at different α-thrombin concentrations. The uppermost line is the response at 0 U/mL a- thrombin, and each subsequent line runs in order for α-thrombin concentrations of 0.005, 0.01 , 0.02, 0.04, 0.08, 0.16, 0.31 , 0.63, 1.3, 2.5, 5.0, 10, and 20 U/mL. (D). Thrombin-mediated release rate as a function of time for rFVIIIFc, SC rFVIIIFc, and rBDD FVIII at 25° C (top) and 37° C (bottom). The x-axis shows time in seconds and the y-axis shows response in response units (RU). Individual lines indicate response at different α-thrombin concentrations. The uppermost line is the response at 20 U/mL a-thrombin, and each subsequent line runs in order for a-thrombin concentrations of 10, 5, 2.5, 1.3, 0.63, 0.31, 0.16, 0.08, 0.04, 0.02, 0.01, and 0.005 U/mL. (E). Peak thrombin-mediated release rate as a function of thrombin concentration for rFVIIIFc, SC rFVIIIFc, and rBDD FVIII at 25° C (top) and 37° C (bottom). EC50 is half maximal effective concentration. The x-axis is α-thrombin concentration in U/mL and the y-axis is maximum release rate in RU/second.
DETAILED DESCRIPTION OF THE INVENTION
[0075] The present invention provides a method of treating Hemophilia A with Factor VIII
(processed, single chain, or a combination thereof) using a longer dosing interval and/or greater AUC than is possible with currently known Factor VIII products. The present invention also provides improved Factor VIII chimeric polypeptides and methods of production.
[0076] Treatment of hemophilia A is by replacement therapy targeting restoration of FVIII activity to 1 to 5 % of normal levels to prevent spontaneous bleeding (Mannucci, P.M. et al, N. Engl. J. Med. 344: 177 -9 (2001), herein incorporated by reference in its entirety). There are plasma-derived and recombinant FVIII products available to treat bleeding episodes on-demand or to prevent bleeding episodes from occurring by treating prophylactically. Based on the short half-life of these products (8-12 hr) (White G.C., et al, Thromb. Haemost. 77:660-7 (1997); Morfini, M., Haemophilia 9 (suppl l):94-99; discussion 100 (2003)), treatment regimens require frequent intravenous administration, commonly two to three times weekly for prophylaxis and one to three times daily for on-demand treatment (Manco-Johnson, M.J., et al, N. Engl. J. Med. 357:535-544 (2007)), each of which is incorporated herein by reference in its entirety. Such frequent administration is painful and inconvenient.
[0077] The present invention provides a method of administering Factor VIII to a human subject in need thereof (e.g., human patient), comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, or a hybrid of such a polypeptide at a dosing interval at least about one and one-half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion. The present invention is also directed to a method of increasing dosing interval of Factor VIII administration in a human subject in need thereof comprising administering the chimeric Factor VIII polypeptide.
[0078] The dosing interval may be at least about one and one -half to six times longer, one and one -half to five times longer, one and one -half to four times longer, one and one -half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion. The dosing interval may be at least about one and one -half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one -half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion. The dosing interval may be about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
[0079] The dosing interval may be at least about one and one-half to 5, one and one -half, 2, 3, 4, or 5 days or longer.
[0080] The present invention also provides a method of administering Factor VIII to a human subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide, or a hybrid of such a polypeptide to obtain an area under the plasma concentration versus time curve (AUC) at least about one and one-quarter times greater than the AUC obtained by an equivalent dose of said Factor VIII without non-Factor VIII portion (a polypeptide consisting of said Factor VIII portion), e.g., without the Fc portion. The present invention thus includes a method of increasing or extending AUC of Factor VIII activity in a human patient in need thereof comprising administering the chimeric Factor VIII polypeptide.
[0081] The present invention also provides a method of administering Factor VIII to a subject in need thereof, comprising administering to the subject a therapeutic dose of a polypeptide comprising a Factor VIII and an Fc or a hybrid of such a polypeptide at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
[0082] The methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
[0083] "Administering," as used herein, means to give a pharmaceutically acceptable Factor VIII polypeptide of the invention to a subject via a pharmaceutically acceptable route. Routes of administration can be intravenous, e.g., intravenous injection and intravenous infusion. Additional routes of administration include, e.g., subcutaneous, intramuscular, oral, nasal, and pulmonary administration. Chimeric polypeptides and hybrid proteins may be administered as part of a pharmaceutical composition comprising at least one excipient.
[0084] "Area under the plasma concentration versus time curve (AUC)," as used herein, is the same as the term of art in pharmacology, and is based upon the rate and extent of absorption of Factor VIII following administration. AUC is determined over a specified time period, such as 12, 18, 24, 36, 48, or 72 hours, or for infinity using extrapolation based on the slope of the curve. Unless otherwise specified herein, AUC is determined for infinity. The determination of AUC may be carried out in a single subject, or in a population of subjects for which the average is calculated.
[0085] A "B domain" of Factor VIII, as used herein, is the same as the B domain known in the art that is defined by internal amino acid sequence identity and sites of proteolytic cleavage by thrombin, e.g., residues Ser741-Argl648 of full length human factor VIII. The other human factor VIII domains are defined by the following amino acid residues: Al, residues Alal-Arg372; A2, residues Ser373-Arg740; A3, residues Serl690-Ile2032; CI, residues Arg2033-Asn2172; C2, residues Ser2173-Tyr2332. The A3-C1-C2 sequence includes residues Serl690-Tyr2332. The remaining sequence, residues Glul649-Argl689, is usually referred to as the factor VIII light chain activation peptide. The locations of the boundaries for all of the domains, including the B domains, for porcine, mouse and canine factor VIII are also known in the art. In one embodiment, the B domain of Factor VIII is deleted ("B domain deleted factor VIII" or "BDD FVIII"). An example of a BDD FVIII is REFACTO® (recombinant BDD FVIII), which has the same sequence as the Factor VIII portion of the sequence in Table 2A(i) (amino acids 1 to 1457 or 20 to 1457 of SEQ ID NO:2). In another embodiment, the B domain deleted Factor VIII contains an intact intracellular processing site, which corresponds to Arginine at residue 754 of B domain deleted Factor VIII, which corresponds to Arginine residue 773 of SEQ ID NO: 2, or residue 1648 of full- length Factor VIII, which corresponds to Arginine residue 1667 of SEQ ID NO: 6. The sequence residue numbers used herein without referring to any SEQ ID Numbers correspond to the Factor VIII sequence without the signal peptide sequence (19 amino acids) unless otherwise indicated. For example, S743/Q1638 of full-length Factor VIII corresponds to S762/Q1657 of SEQ ID NO: 6 due to the 19 amino acid signal peptide sequence. In other embodiments, the B domain deleted FVIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII. In some embodiments, the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648. In certain embodiments, the substitution or mutation is an amino acid other than arginine, e.g., alanine.
[0086] A "B domain deleted factor VIII" may have the full or partial deletions disclosed in U.S.
Patent Nos. 6,316,226, 6,346,513, 7,041,635, 5,789,203, 6,060,447, 5,595,886, 6,228,620, 5,972,885, 6,048,720, 5,543,502, 5,610,278, 5,171,844, 5,112,950, 4,868,112, and 6,458,563, each of which is incorporated herein by reference in its entirety. In some embodiments, a B domain deleted factor VIII sequence of the present invention comprises any one of the deletions disclosed at col. 4, line 4 to col. 5, line 28 and examples 1-5 of U.S. Patent No. 6,316,226 (also in US 6,346,513). In some embodiments, a B domain deleted factor VIII of the present invention has a deletion disclosed at col. 2, lines 26-51 and examples 5-8 of U.S. Patent No. 5,789,203 (also US 6,060,447, US 5,595,886, and US 6,228,620). In some embodiments, a B domain deleted factor VIII has a deletion described in col. 1, lines 25 to col. 2, line 40 of US Patent No. 5,972,885; col. 6, lines 1-22 and example 1 of U.S. Patent no. 6,048,720; col. 2, lines 17-46 of U.S. Patent No. 5,543,502; col. 4, line 22 to col. 5, line 36 of U.S. Patent no. 5,171,844; col. 2, lines 55-68, figure 2, and example 1 of U.S. Patent No. 5,112,950; col. 2, line 2 to col. 19, line 21 and table 2 of U.S. Patent No. 4,868,112; col. 2, line 1 to col. 3, line 19, col. 3, line 40 to col. 4, line 67, col. 7, line 43 to col. 8, line 26, and col. 11, line 5 to col. 13, line 39 of U.S. Patent no. 7,041,635; or col. 4, lines 25-53, of U.S. Patent No. 6,458,563. In some embodiments, a B domain deleted factor VIII has a deletion of most of the B domain, but still contains amino- terminal sequences of the B domain that are essential for in vivo proteolytic processing of the primary translation product into two polypeptide chain (i.e., intracellular processing site), as disclosed in WO 91/09122, which is incorporated herein by reference in its entirety. In some embodiments, a B domain deleted factor VIII is constructed with a deletion of amino acids 747- 1638, i.e., virtually a complete deletion of the B domain. Hoeben R.C., et al. J. Biol. Chem. 265 (13): 7318-7323 (1990), incorporated herein by reference in its entirety. A B domain deleted factor VIII may also contain a deletion of amino acids 771-1666 or amino acids 868-1562 of factor VIII. Meulien P., et al. Protein Eng. 2(4): 301-6 (1988), incorporated herein by reference in its entirety. Additional B domain deletions that are part of the invention include, e.g.,: deletion of amino acids 982 through 1562 or 760 through 1639 (Toole et al., Proc. Natl. Acad. Sci. U.S.A. 53:5939-5942 (1986)), 797 through 1562 (Eaton et al, Biochemistry 25:8343-8347 (1986)), 741 through 1646 (Kaufman (PCT published application No. WO 87/04187)), 747-1560 (Sarver et al, DNA 6:553-564 (1987)), 741 through 1648 (Pasek (PCT application No.88/00831)), 816 through 1598 or 741 through 1689 (Lagner (Behring Inst. Mitt. (1988) No 82: 16-25, EP 295597)), each of which is incorporated herein by reference in its entirety. Each of the foregoing deletions may be made in any Factor VIII sequence.
[0087] In one embodiment, the B domain deleted Factor VIII portion in the chimeric polypeptide is processed into two chains connected (or associated) by a metal bond, the first chain comprising a heavy chain (Al -A2 -partial B) and a second chain comprising a light chain (A3-C1 -C2) . In another embodiment, the B domain deleted Factor VIII portion is a single chain Factor VIII. The single chain Factor VIII can comprise an intracellular processing site, which corresponds to Arginine at residue 754 of B domain deleted Factor VIII (residue 773 of SEQ ID NO: 2) or at residue 1648 of full-length Factor VIII (residue 1657 of SEQ ID NO: 6).
[0088] The metal bond between the heavy chain and the light chain can be any metal known in the art. For example, the metals useful for the invention can be a divalent metal ion. The metals that can be used to associate the heavy chain and light chain include, but not limited to, Ca2+, Mn , or Cu . Fatouros et ah, Intern. J. Pharm. 155(1): 121-131 (1997); Wakabayashi et ah, JBC. 279(13): 12677-12684 (2004).
[0089] "Chimeric polypeptide," as used herein, means a polypeptide that includes within it at least two polypeptides (or subsequences or peptides) from different sources. Chimeric polypeptides may include, e.g., two, three, four, five, six, seven, or more polypeptides from different sources, such as different genes, different cDNAs, or different animal or other species. Chimeric polypeptides may include, e.g., one or more linkers joining the different subsequences. Thus, the subsequences may be joined directly or they may be joined indirectly, via linkers, or both, within a single chimeric polypeptide. Chimeric polypeptides may include, e.g., additional peptides such as signal sequences and sequences such as 6His and FLAG that aid in protein purification or detection. In addition, chimeric polypeptides may have amino acid or peptide additions to the N- and/or C-termini.
[0090] In some embodiments, the chimeric polypeptide comprises a Factor VIII portion and a non-Factor VIII portion. Exemplary non-Factor VIII portions include, e.g., Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of human chorionic gonadotropin (hCG) with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, and albumin-binding small molecules. Exemplary chimeric polypeptides of the invention include, e.g., chimeric Factor VIII-Fc polypeptides, chimeric Factor VIII-XTEN polypeptides, chimeric Factor Vlll-albumin polypeptides, chimeric Factor VIII-PAS polypeptides, chimeric Factor Vlll-transferrin polypeptides, chimeric Factor VIII-CTP polypeptides, chimeric Factor VIII-PEG polypeptides, chimeric Factor VIII-HES polypeptides, chimeric Facotr Vlll-albumbin binding polypeptide polypeptides, and chimeric Factor VIII.- albumin-binding small molecule polypeptides.
[0091] Exemplary chimeric Factor VIII-Fc polypeptides include, e.g., SEQ ID NO:2 or 6 (Table
2), with or without their signal sequences and the chimeric Fc polypeptide of SEQ ID NO:4 (Table 2).
[0092] The chimeric polypeptide may comprise a sequence at least 90% or 95% identical to the
Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2), wherein the sequence has Factor VIII activity. The Factor VIII activity can be measured by activated Partial Thromboplastin Time (aPPT) assay, chromogenic assay, or other known methods. The chimeric polypeptide may comprise a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2). [0093] As discussed above, exemplary chimeric polypeptides include Factor VIII fused to one or more XTEN polypeptides. Schellenburger et al, Nat. Biotech. 27: 1186-90 (2009), which is incorporated herein by reference in its entirety. The XTEN polypeptide can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII. A protease site may be included between the XTEN portion and the Factor VIII portion to allow such processing. XTEN polypeptides include, e.g., those disclosed in WO 2009/023270, WO 2010/091122, WO 2007/103515, US 2010/0189682, and US 2009/0092582, each of which is incorporated herein by reference in its entirety.
[0094] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to one or more albumin polypeptides, albumin binding polypeptides, or albumin-binding small molecules. In one embodiment, the albumin is human albumin. The albumin or albumin binding protein can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII. Examples of albumin, e.g., fragments thereof, that may be used in the present invention are known, e.g., U.S. Patent No. 7,592,010; U.S. Patent No. 6,686,179; and Schulte, Thrombosis Res. 124 Suppl. 2:S6-S8 (2009), each of which is incorporated herein by reference in its entirety.
[0095] The albumin binding polypeptides can compromise, without limitation, bacterial albumin- binding domains, albumin-binding peptides, or albumin-binding antibody fragments that can bind to albumin. Domain 3 from streptococcal protein G, as disclosed by Kraulis et al, FEBS Lett. 378: 190-194 (1996) and Linhult et al, Protein Sci. 11 :206-213 (2002) is an example of a bacterial albumin-binding domain. Examples of albumin-binding peptides include a series of peptides having the core sequence DICLPRWGCLW (SEQ ID NO: 7). See, e,g., Dennis et al., J. Biol. Chem. 2002, 277: 35035-35043 (2002). Examples of albumin-binding antibody fragments are disclosed in Muller and Kontermann, Curr. Opin. Mol. Ther. 9:319-326 (2007); Rooverset et al, Cancer Immunol. Immunother. 56:303-317 (2007), and Holt et al., Prot. Eng. Design Sci., 21 :283-288 (2008), which are incorporated herein by reference in their entireties..
[0096] In certain aspects, a recombinant FVIII polypeptide of the invention comprises at least one attachment site for a non-polypeptide small molecule, variant, or derivative that can bind to albumin thereof. An example of such albumin binding moieties is 2-(3-maleimidopropanamido)- 6-(4-(4-iodophenyl)butanamido)hexanoate ("Albu" tag) as disclosed by Trusselet et al, Bioconjugate Chem. 20:2286-2292 (2009).
[0097] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to at least one β subunit of the C-terminal peptide (CTP) of human chorionic gonadotropin or fragment, variant, or derivative thereof. The CTP can be fused to Factor VIII either the N- terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII. One or more CTP peptides fused to or inserted into a recombinant protein is known to increase the in vivo half-life of that protein. See, e.g., U.S. Patent No. 5,712,122, incorporated by reference herein in its entirety. Exemplary CTP peptides include
DPRFQDSSSSKAPPPSLPSPSRLPGPSDTPIL (SEQ ID NO: 8) or
SSSSKAPPPSLPSPSRLPGPSDTPILPQ. (SEQ ID NO: 9). See, e.g., U.S. Patent Application Publication No. US 2009/0087411 Al, incorporated by reference.
[0098] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to at least one PAS sequence or fragment, variant, or derivative thereof. The PAS sequence can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII. A PAS peptide or PAS sequence, as used herein, means an amino acid sequence comprising mainly alanine and serine residues or comprising mainly alanine, serine, and proline residues, the amino acid sequence forming random coil conformation under physiological conditions. Accordingly, the PAS sequence is a building block, an amino acid polymer, or a sequence cassette comprising, consisting essentially of, or consisting of alanine, serine, and proline which can be used as a part of the heterologous moiety in the chimeric protein. An amino acid polymer also can form random coil conformation when residues other than alanine, serine, and proline are added as a minor constituent in the PAS sequence. By "minor constituent" is meant that that amino acids other than alanine, serine, and proline can be added in the PAS sequence to a certain degree, e.g., up to about 12%, i.e., about 12 of 100 amino acids of the PAS sequence, up to about 10%, up to about 9%>, up to about 8%>, about 6%, about 5%, about 4%>, about 3%), i.e. about 2%>, or about 1%>, of the amino acids. The amino acids different from alanine, serine and proline cab be selected from the group consisting of Arg, Asn, Asp, Cys, Gin, Glu, Gly, His, He, Leu, Lys, Met, Phe, Thr, Trp, Tyr, and Val. Under physiological conditions, a PAS peptide forms a random coil conformation and thereby can mediate an increased in vivo and/or in vitro stability to a recombinant protein of the invention, and has procoagulant activity.
[0099] Non-limiting examples of the PAS peptides include ASPAAPAPASPAAPAPSAPA
(SEQ ID NO: 10), AAPASPAPAAPSAPAPAAPS (SEQ ID NO: 11), APSSPSPSAPSSPSPASPSS (SEQ ID NO: 12), APSSPSPSAPSSPSPASPS (SEQ ID NO: 13), SSPSAPSPSSPASPSPSSPA (SEQ ID NO: 14), AASPAAPSAPPAAASPAAPSAPPA (SEQ ID NO: 15), ASAAAPAAASAAASAPSAAA (SEQ ID NO: 16) or any variants, derivatives, fragments, or combinations thereof. Additional examples of PAS sequences are known from, e.g., US Pat. Publ. No. 2010/0292130 Al and PCT Appl. Publ. No. WO 2008/155134 Al . European issued patent EP2173890.
[00100] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to at least one transferrin peptide or fragment, variant, or derivative thereof. At least one transferrin peptide can be fused to either the N-terminal end of FVIII or to the C-terminal end of FVIII or inserted between two amino acids in FVIII. Any transferrin can be fused to or inserted into a recombinant FVIII protein of the invention. As an example, wild-type human Tf (Tf) is a 679 amino acid protein, of approximately 75 KDa (not accounting for glycosylation), with two main domains, N (about 330 amino acids) and C (about 340 amino acids), which appear to originate from a gene duplication. See GenBank accession numbers NM001063, XM002793, M12530, XM039845, XM 039847 and S95936 (www.ncbi.nlm.nih.gov), all of which are herein incorporated by reference in their entirety.
[00101] Transferrin transports iron through transferrin receptor (Tf ) -mediated endocytosis.
After the iron is released into an endosomal compartment and Tf-TfR complex is recycled to cell surface, the Tf is released back extracellular space for next cycle of iron transporting. Tf possesses a long half-life that is in excess of 14-17 days (Li et al, Trends Pharmacol. Sci. 23:206- 209 (2002)).Transferrin fusion proteins have been studied for half-life extension, targeted deliver for cancer therapies, oral delivery and sustained activation of proinsulin (Brandsma et al, Biotechnol. Adv., 29: 230-238 (2011); Bai et al, Proc. Natl. Acad. Sci. USA 102:7292-7296 (2005); Kim et al., J. Pharmacol. Exp. Ther., 334:682-692 (2010); Wang et al., J. Controlled Release 155:386-392 (2011)).
[00102] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to at least one polyethylene glycol (PEG) moieties.
[00103] PEGylated FVIII can refer to a conjugate formed between FVIII and at least one polyethylene glycol (PEG) molecule. PEG is commercially available in a large variety of molecular weights and average molecular weight ranges. Typical examples of PEG average molecular weight ranges include, but are not limited to, about 200, about 300, about 400, about 600, about 1000, about 1300-1600, about 1450, about 2000, about 3000, about 3000-3750, about 3350, about 3000-7000, about 3500-4500, about 5000-7000, about 7000-9000, about 8000, about 10000, about 8500-11500, about 16000-24000, about 35000, about 40000, about 60000, and about 80000 daltons. These average molecular weights are provided merely as examples and are not meant to be limiting in any way.
[00104] A recombinant FVIII protein of the invention can be PEGylated to include mono- or poly-
(e.g., 2-4) PEG moieties. PEGylation can be carried out by any of the PEGylation reactions known in the art. Methods for preparing a PEGylated protein product will generally include (i) reacting a polypeptide with polyethylene glycol (such as a reactive ester or aldehyde derivative of PEG) under conditions whereby the peptide of the invention becomes attached to one or more PEG groups; and (ii) obtaining the reaction product(s). In general, the optimal reaction conditions for the reactions will be determined case by case based on known parameters and the desired result.
[00105] There are a number of PEG attachment methods available to those skilled in the art, for example Malik F et al., Exp. Hematol. 20: 1028-35 (1992); Francis, Focus on Growth Factors 3(2):4-10 (1992); European Pat. Pub. Nos. EP0401384, EP0154316, and EP0401384; and International Pat. Appl. Pub. Nos. W092/16221 and W095/34326. As a non-limiting example, FVIII variants can contain cysteine substitutions in one or more insertion sites in FVIII, and the cysteines can be further conjugated to PEG polymer. See Mei et al, Blood 116:270-279 (2010) and U.S. Patent No. 7,632,921, which are incorporated herein by reference in their entireties.
[00106] As discussed above, exemplary chimeric polypeptides also include Factor VIII fused to at least one hydroxyethyl starch (HES) polymer. HES is a derivative of naturally occurring amylopectin and is degraded by alpha-amylase in the body. HES exhibits advantageous biological properties and is used as a blood volume replacement agent and in hemodilution therapy in the clinics. See, e.g., Sommermeyer et al., Krankenhauspharmazie 8:271-278 (1987); and Weidler et al., Arzneim.-Forschung/Drug Res. 41 : 494-498 (1991).
[00107] HES is mainly characterized by the molecular weight distribution and the degree of substitution. HES has a mean molecular weight (weight mean) of from 1 to 300 kD, from 2 to 200kD, from 3 to 100 kD, or from 4 to 70kD. Hydroxyethyl starch can further exhibit a molar degree of substitution of from 0.1 to 3, from 0.1 to 2,from 0.1 to 0.9, or from 0.1 to 0.8, and a ratio between C2:C6 substitution in the range of from 2 to 20 with respect to the hydroxyethyl groups. HES with a mean molecular weight of about 130 kD is Voluven® from Fresenius. Voluven® is an artificial colloid, employed, e.g. , for volume replacement used in the therapeutic indication for therapy and prophylaxis of hypovolaemia. There are a number of HES attachment methods available to those skilled in the art, e.g., the same PEG attachment methods described above.
[00108] In some embodiments, a chimeric polypeptide comprising a Factor VIII portion has an increased half-life (tl/2) over a polypeptide consisting of the same Factor VIII portion without the non Factor VIII portion. A chimeric Factor VIII polypeptide with an increased tl/2 may be referred to herein as a long-acting Factor VIII. Long-acting chimeric Factor VIII polypeptides include, e.g., Factor VIII fused to Fc (including, e.g., chimeric Factor VIII polypeptides in the form of a hybrid such as a FVIIIFc monomer dimer hybrid; see Example 1 , Fig. 1 , and Table 2A; and US Patent Nos. 7,404,956 and 7,348,004), Factor VIII fused to XTEN, and Factor VIII fused to albumin.
[00109] "Culture," "to culture" and "culturing," as used herein, means to incubate cells under in vitro conditions that allow for cell growth or division or to maintain cells in a living state. "Cultured cells," as used herein, means cells that are propagated in vitro.
[00110] "Factor VIII," as used herein, means functional factor VIII polypeptide in its normal role in coagulation, unless otherwise specified. Thus, the term Factor VIII includes variant polypeptides that are functional. Factor VIII proteins can be the human, porcine, canine, and murine factor VIII proteins. As described in the Background Art section, the full length polypeptide and polynucleotide sequences are known, as are many functional fragments, mutants and modified versions. Examples of human factor VIII sequences are shown as subsequences in SEQ ID NOs:2 or 6 (Table 2). Factor VIII polypeptides include, e.g., full-length factor VIII, full- length factor VIII minus Met at the N-terminus, mature factor VIII (minus the signal sequence), mature factor VIII with an additional Met at the N-terminus, and/or factor VIII with a full or partial deletion of the B domain. Factor VIII variants include B domain deletions, whether partial or full deletions.
[00111] A great many functional factor VIII variants are known, as is discussed above and below.
In addition, hundreds of nonfunctional mutations in factor VIII have been identified in hemophilia patients, and it has been determined that the effect of these mutations on factor VIII function is due more to where they lie within the 3 -dimensional structure of factor VIII than on the nature of the substitution (Cutler et al, Hum. Mutat. 79:274-8 (2002)), incorporated herein by reference in its entirety. In addition, comparisons between factor VIII from humans and other species have identified conserved residues that are likely to be required for function (Cameron et al., Thromb. Haemost. 79:317-22 (1998); US 6,251,632), incorporated herein by reference in its entirety.
[00112] The human factor VIII gene was isolated and expressed in mammalian cells (Toole, J. J., et al, Nature 372:342-347 (1984); Gitschier, J., et al, Nature 372:326-330 (1984); Wood, W. I., et al, Nature 372:330-337 (1984); Vehar, G. A., et al, Nature 372:337-342 (1984); WO 87/04187; WO 88/08035; WO 88/03558; U.S. Pat. No. 4,757,006), each of which is incorporated herein by reference in its entirety, and the amino acid sequence was deduced from cDNA. Capon et al., U.S. Pat. No. 4,965,199, incorporated herein by reference in its entirety, discloses a recombinant DNA method for producing factor VIII in mammalian host cells and purification of human factor VIII. Human factor VIII expression in CHO (Chinese hamster ovary) cells and BHKC (baby hamster kidney cells) has been reported. Human factor VIII has been modified to delete part or all of the B domain (U.S. Pat. Nos. 4,994,371 and 4,868,112, each of which is incorporated herein by reference in its entirety), and replacement of the human factor VIII B domain with the human factor V B domain has been performed (U.S. Pat. No. 5,004,803, incorporated herein by reference in its entirety). The cDNA sequence encoding human factor VIII and predicted amino acid sequence are shown in SEQ ID NOs: l and 2, respectively, of US Application Publ. No. 2005/0100990, incorporated herein by reference in its entirety.
[00113] U.S. Pat. No. 5,859,204, Lollar, J. S., incorporated herein by reference in its entirety, reports functional mutants of factor VIII having reduced antigenicity and reduced immunoreactivity. U.S. Pat. No. 6,376,463, Lollar, J. S., incorporated herein by reference in its entirety, also reports mutants of factor VIII having reduced immunoreactivity. US Application Publ. No. 2005/0100990, Saenko et al., incorporated herein by reference in its entirety, reports functional mutations in the A2 domain of factor VIII. [00114] A number of functional factor VIII molecules, including B-domain deletions, are disclosed in the following patents US 6,316,226 and US 6,346,513, both assigned to Baxter; US 7,041,635 assigned to In2Gen; US 5,789,203, US 6,060,447, US 5,595,886, and US 6,228,620 assigned to Chiron; US 5,972,885 and US 6,048,720 assigned to Biovitrum, US 5,543,502 and US 5,610,278 assigned to Novo Nordisk; US 5,171,844 assigned to Immuno Ag; US 5,112,950 assigned to Transgene S.A.; US 4,868,112 assigned to Genetics Institute, each of which is incorporated herein by reference in its entirety.
[00115] The porcine factor VIII sequence is published, (Toole, J. J., et al, Proc. Natl. Acad. Sci.
USA 53:5939-5942 (1986)), incorporated herein by reference in its entirety, and the complete porcine cDNA sequence obtained from PCR amplification of factor VIII sequences from a pig spleen cDNA library has been reported (Healey, J. F. et al, Blood 55:4209-4214 (1996), incorporated herein by reference in its entirety). Hybrid human/porcine factor VIII having substitutions of all domains, all subunits, and specific amino acid sequences were disclosed in U.S. Pat. No. 5,364,771 by Lollar and Runge, and in WO 93/20093, incorporated herein by reference in its entirety. More recently, the nucleotide and corresponding amino acid sequences of the Al and A2 domains of porcine factor VIII and a chimeric factor VIII with porcine Al and/or A2 domains substituted for the corresponding human domains were reported in WO 94/11503, incorporated herein by reference in its entirety. U.S. Pat. No. 5,859,204, Lollar, J. S., also discloses the porcine cDNA and deduced amino acid sequences. US Pat. No. 6,458,563, incorporated herein by reference in its entirety assigned to Emory discloses a B-domain deleted porcine Factor VIII.
[00116] The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90% or
95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2; and amino acids 20 to 2351 of SEQ ID NO:6), wherein said Factor VIII portion has Factor VIII activity. The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2; and amino acids 20 to 2351 of SEQ ID NO:6).
[00117] The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 90%> or
95%) identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 and amino acids 1 to 2351 of SEQ ID NO:6), wherein said Factor VIII portion has Factor VIII activity. The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 and amino acids 1 to 2351 of SEQ ID NO: 6). [00118] "Equivalent dose," as used herein, means the same dose of Factor VIII activity as expressed in International Units, which is independent of molecular weight of the polypeptide in question. One International Unit (IU) of factor VIII activity corresponds approximately to the quantity of factor VIII in one milliliter of normal human plasma. Several assays are available for measuring Factor VIII activity, including the European Pharmacopoeia chromogenic substrate assay and a one stage clotting assay.
[00119] "Fc," as used herein, means functional neonatal Fc receptor (FcRn) binding partners, unless otherwise specified. An FcRn binding partner is any molecule that can be specifically bound by the FcRn receptor with consequent active transport by the FcRn receptor of the FcRn binding partner. Thus, the term Fc includes any variants of IgG Fc that are functional. The region of the Fc portion of IgG that binds to the FcRn receptor has been described based on X-ray crystallography (Burmeister et al. , Nature 372:379 (1994), incorporated herein by reference in its entirety). The major contact area of the Fc with the FcRn is near the junction of the CH2 and CH3 domains. Fc-FcRn contacts are all within a single Ig heavy chain. The FcRn binding partners include, e.g., whole IgG, the Fc fragment of IgG, and other fragments of IgG that include the complete binding region of FcRn. The major contact sites include amino acid residues 248, 250-257, 272, 285, 288, 290-291, 308-311, and 314 of the CH2 domain and amino acid residues 385-387, 428, and 433-436 of the CH3 domain. References made to amino acid numbering of immunoglobulins or immunoglobulin fragments, or regions, are all based on Kabat et al. 1991, Sequences of Proteins of Immunological Interest, U. S. Department of Public Health, Bethesda; MD, incorporated herein by reference in its entirety. (The FcRn receptor has been isolated from several mammalian species including humans. The sequences of the human FcRn, rat FcRn, and mouse FcRn are known (Story et al., J. Exp. Med. 180: 2377 (1994), incorporated herein by reference in its entirety.) An Fc may comprise the CH2 and CH3 domains of an immunoglobulin with or without the hinge region of the immunoglobulin. Exemplary Fc variants are provided in WO 2004/101740 and WO 2006/074199, incorporated herein by reference in its entirety.
[00120] Fc (or Fc portion of a chimeric polypeptide) may contain one or more mutations, and combinations of mutations.
[00121] Fc (or Fc portion of a chimeric polypeptide) may contain mutations conferring increased half-life such as M252Y, S254T, T256E, and combinations thereof, as disclosed in Oganesyan et al., Mol. Immunol. 46: 1750 (2009), which is incorporated herein by reference in its entirety; H433K, N434F, and combinations thereof, as disclosed in Vaccaro et al, Nat. Biotechnol. 23: 1283 (2005), which is incorporated herein by reference in its entirety; the mutants disclosed at pages 1-2, paragraph [0012], and Examples 9 and 10 of US 2009/0264627 Al, which is incorporated herein by reference in its entirety; and the mutants disclosed at page 2, paragraphs [0014] to [0021] of US 20090163699 Al, which is incorporated herein by reference in its entirety. Fc (or Fc portion of a chimeric polypeptide) may also include, e.g., the following mutations: The Fc region of IgG can be modified according to well recognized procedures such as site directed mutagenesis and the like to yield modified IgG or Fc fragments or portions thereof that will be bound by FcRn. Such modifications include, e.g., modifications remote from the FcRn contact sites as well as modifications within the contact sites that preserve or even enhance binding to the FcRn. For example the following single amino acid residues in human IgGl Fc (Fcyl) can be substituted without significant loss of Fc binding affinity for FcRn: P238A, S239A, K246A, K248A, D249A, M252A, T256A, E258A, T260A, D265A, S267A, H268A, E269A, D270A, E272A, L274A, N276A, Y278A, D280A, V282A, E283A, H285A, N286A, T289A, K290A, R292A, E293A, E294A, Q295A, Y296F, N297A, S298A, Y300F, R301A, V303A, V305A, T307A, L309A, Q31 1A, D312A, N315A, K317A, E318A, K320A, K322A, S324A, K326A, A327Q, P329A, A330Q, A330S, P331A, P331 S, E333A, K334A, T335A, S337A, K338A, K340A, Q342A, R344A, E345A, Q347A, R355A, E356A, M358A, T359A, K360A, N361A, Q362A, Y373A, S375A D376A, A378Q, E380A, E382A, S383A, N384A, Q386A, E388A, N389A, N390A, Y391F, K392A, L398A, S400A, D401A, D413A, K414A, R416A, Q418A, Q419A, N421A, V422A, S424A, E430A, N434A, T437A, Q438A, K439A, S440A, S444A, and K447A, where for example P238A represents wildtype proline substituted by alanine at position number 238. In addition to alanine other amino acids may be substituted for the wildtype amino acids at the positions specified above. Mutations may be introduced singly into Fc giving rise to more than one hundred FcRn binding partners distinct from native Fc. Additionally, combinations of two, three, or more of these individual mutations may be introduced together, giving rise to hundreds more FcRn binding partners. Certain of these mutations may confer new functionality upon the FcRn binding partner. For example, one embodiment incorporates N297A, removing a highly conserved N-glycosylation site. The effect of this mutation is to reduce immunogenicity, thereby enhancing circulating half-life of the FcRn binding partner, and to render the FcRn binding partner incapable of binding to FcyRI, FcyRIIA, FcyRIIB, and FcyRIIIA, without compromising affinity for FcRn (Routledge et al. 1995, Transplantation 60:847, which is incorporated herein by reference in its entirety; Friend et al. 1999, Transplantation 68: 1632, which is incorporated herein by reference in its entirety; Shields et al. 1995, J. Biol. Chem. 276:6591 , which is incorporated herein by reference in its entirety). Additionally, at least three human Fc gamma receptors appear to recognize a binding site on IgG within the lower hinge region, generally amino acids 234-237. Therefore, another example of new functionality and potential decreased immunogenicity may arise from mutations of this region, as for example by replacing amino acids 233-236 of human IgGl "ELLG" to the corresponding sequence from IgG2 "PVA" (with one amino acid deletion). It has been shown that FcyRI, FcyRII, and FcyRIII which mediate various effector functions will not bind to IgGl when such mutations have been introduced (Ward and Ghetie, Therapeutic Immunology 2:11 (1995), which is incorporated herein by reference in its entirety; and Armour et al., Eur. J. Immunol. 29:2613 (1999), which is incorporated herein by reference in its entirety). As a further example of new functionality arising from mutations described above affinity for FcRn may be increased beyond that of wild type in some instances. This increased affinity may reflect an increased "on" rate, a decreased "off rate or both an increased "on" rate and a decreased "off rate. Mutations believed to impart an increased affinity for FcRn include, e.g., T256A, T307A, E380A, and N434A (Shields et al. , J. Biol. Chem. 276:6591 (2001), which is incorporated herein by reference in its entirety).
[00123] The Fc (or Fc portion of a chimeric polypeptide) may be at least 90% or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6). The Fc (or Fc portion of a chimeric polypeptide) may be identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 and amino acids 2352 to 2578 of SEQ ID NO:6).
[00124] "Hybrid" polypeptides and proteins, as used herein, means a combination of a chimeric polypeptide with a second polypeptide. The chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via protein-protein interactions, such as charge-charge or hydrophobic interactions. The chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via disulfide or other covalent bond(s). Hybrids are described in WO 2004/101740 and WO 2006/074199, each of which is incorporated herein by reference in its entirety. See also U.S. Patent Nos. 7,404,956 and 7,348,004, each of which is incorporated herein by reference in its entirety. The second polypeptide may be a second copy of the same chimeric polypeptide or it may be a non-identical chimeric polypeptide. See, e.g., Figure 1, Example 1, and Table 2. In one embodiment, the second polypeptide is a polypeptide comprising an Fc. In another embodiment, the chimeric polypeptide is a chimeric Factor VIII-Fc polypeptide and the second polypeptide consists essentially of Fc, e.g., the hybrid polypeptide of Example 1, which is a rFVIIIFc recombinant fusion protein consisting of a single molecule of recombinant B- domain deleted human FVIII (BDD-rFVIII) fused to the dimeric Fc domain of the human IgGl, with no intervening linker sequence. This hybrid polypeptide is referred to herein as FVIIIFc monomelic Fc fusion protein, FVIIIFc monomer hybrid, monomeric FVIIIIFc hybrid, and FVIIIFc monomer-dimer. See Example 1, Fig. 1, and Table 2A. The Examples provide preclinical and clinical data for this hybrid polypeptide.
[00125] The second polypeptide in a hybrid may comprise or consist essentially of a sequence at least 90%) or 95%> identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95%> identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO-:4). The second polypeptide may comprise or consist essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
[00126] Figure 1 is a schematic showing the structure of a B domain deleted factor VIII-Fc chimeric polypeptide, and its association with a second polypeptide that is an Fc polypeptide. To obtain this hybrid, the coding sequence of human recombinant B-domain deleted FVIII was obtained by reverse transcription-polymerase chain reaction (RT-PCR) from human liver poly A RNA (Clontech) using FVIII-specific primers. The FVIII sequence includes the native signal sequence for FVIII. The B-domain deletion was from serine 743 (S743; 2287 bp) to glutamine 1638 (Q1638; 4969 bp) for a total deletion of 2682 bp. Then, the coding sequence for human recombinant Fc was obtained by RT-PCR from a human leukocyte cDNA library (Clontech) using Fc specific primers. Primers were designed such that the B-domain deleted FVIII sequence was fused directly to the N-terminus of the Fc sequence with no intervening linker. The FVIIIFc DNA sequence was cloned into the mammalian dual expression vector pBUDCE4.1 (Invitrogen) under control of the CMV promoter. A second identical Fc sequence including the mouse Igk signal sequence was obtained by RT-PCR and cloned downstream of the second promoter, EFla, in the expression vector pBUDCE4.1.
[00127] The rFVIIIFc expression vector was transfected into human embryonic kidney 293 cells
(HEK293H; Invitrogen) using Lipofectamine 2000 transfection reagent (Invitrogen). Stable clonal cell lines were generated by selection with Zeocin (Invitrogen). One clonal cell line, 3C4- 22 was used to generate FVIIIFc for characterization in vivo. Recombinant FVIIIFc was produced and purified (McCue et al. 2009) at Biogen Idee (Cambridge, MA). The transfection strategy described above was expected to yield three products, i.e., monomelic rFVIIIFc hybrids, dimeric rFVIIIFc hybrids and dimeric Fc. However, there was essentially no dimeric rFVIIIFc detected in the conditioned medium from these cells. Rather, the conditioned medium contained Fc and monomelic rFVIIIFc. It is possible that the size of dimeric rFVIIIFc was too great and prevented efficient secretion from the cell. This result was beneficial since it rendered the purification of the monomer less complicated than if all three proteins had been present. The material used in these studies had a specific activity of approximately 9000 IU/mg.
[00128] In one embodiment, the polypeptides of the invention are administered to a patient who expresses high level of von Willebrand factor (VWF). "Subject" or "patient" as used herein means a human individual. A subject can be a patient who is currently suffering from a bleeding disorder or is expected to be in need of such a treatment. "Subject" can include an adult or a pediatric subject. The pediatric subject can be a pediatric patient under the age of 12. The term "pediatrics" as used herein is the branch of medicine that deals with the care of infants and children and the treatment of their diseases. In one embodiment, the subject is a pediatric patient who has a diagnosis of severe hemophilia A. In certain embodiments, pediatric subjects are treated with a long-acting Factor VIII polypeptide of the invention.
[00129] VWF is a plasma protein having a multimer structure in which the molecular weight of the various forms varies between approximately 230 kDa for each monomer subunit and up to more than 20 million Da in the multimer forms of greater molecular weight, thus forming the largest known soluble protein. Its plasma concentration is approximately around 5-10 μg/ml (Siedlecki et ah, Blood, vol 88: 2939-2950 (1996)) and the plasma form of smaller size is that corresponding to the dimer, with an approximate size of 500 kDa.
[00130] VWF has an essential role to play in primary haemostasis, being responsible for the adhesion of platelets to damaged vascular surfaces and therefore formation of the platelet plug on which the mechanisms for formation of the fibrin coagulate develop. It is suggested that the higher molecular weight multimers support platelet adhesion mechanisms to the sub-endothelium with greater efficiency and the clinical efficacy of VWF concentrates has been related to the concentration of these multimers of higher molecular weight (Metzner et al, Haemophilia 4:25- 32 (1998).
[00131] Therefore, subjects expressing high levels of VWF would require less frequent dosing of
FVIII compared to a subject who expresses lower or normal levels of VWF. The average range of VWF in plasma is between about 50 IU/dL and about 200 IU/dL. In one embodiment, the average level of VWF in plasma is about 50 IU/dL. In another embodiment, a VWF level in plasma of at least about 100 IU/dL is considered a high VWF level. In another embodiment, a high level of VWF in plasma is between about 100 IU/dL and about 200 IU/dL. In another embodiment a high level of VWF in plasma is at least about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
[00132] Therefore, in one embodiment, subjects expressing at least about 100 IU/dL of plasma
VWF are administered a long-acting FVIII polypeptide of the invention at a long interval dosing regimen. In one embodiment, the long-acting FVIII polypeptide is administered at a dosing interval of at least about 3 days. In another embodiment, the long-acting FVIII polypeptide is administered at a dosing interval of at least about once every week, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
[00133] In one embodiment, the subjects were previously identified as having high levels of
VWF. In certain embodiments, subjects having a blood serotype other than O {i.e., A, B, or AB) require less frequent dosing of long-acting FVIII because the long-acting FVIII has a longer half- life in these subjects. In these subjects, the increased half-life is due to their elevated VWF levels. [00134] Moreover, pharmacokinetic data, defined as the study of the time course of drug absorption, distribution, metabolism, and excretion, can be used as an identifier of subjects eligible for longer or shorter dosing intervals using a long-acting FVIII polypeptide of the invention. Cinical pharmacokinetics is the application of pharmacokinetic principles to the safe and effective therapeutic management of drugs in an individual patient. The primary goals of clinical pharmacokinetics include enhancing efficacy and decreasing toxicity of a patient's drug therapy. The development of strong correlations between drug concentrations and their pharmacologic responses has enabled clinicians to apply pharmacokinetic principles to actual patient situations.
[00135] Thus, in one embodiment, the half-life of a FVIII-Fc polypeptide of the invention is used to identify patients who express high levels of VWF. The range of half-life of FVIII-Fc is between about 10 and about 40 hours, depending at least in part on the levels of VWF also present. On average however, the half- life of FVIII-Fc is about 18 hours. Generally, FVIII-Fc exhibits an increased half-life of at least about 1.2-fold in patients having high levels of VWF compared to the half-life of FVIII-Fc when administered to individuals having average levels of VWF. In one embodiment, FVIII-Fc exhibits an increased half-life of at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold compared to the half-life of FVIII-Fc when administered to individuals having average levels of VWF. In one embodiment, in subjects expressing high levels of VWF, the half-life of FVIII-Fc is between at least about 20 hours and about 40 hours. In another embodiment, the half-life of FVIII-Fc is at least about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours. In one embodiment the half- life of FVIII-Fc is between about 20 and about 27 hours in subjects having high levels of VWF. Thus, in one embodiment, an increased half-life of FVIII-Fc compared to average values is indicative of a subject that is eligible for a longer dosing interval with a long-acting FVIII polypeptide of the invention.
[00136] In another embodiment, the half-life of a short-acting FVIII polypeptide is used to identify patients who express high levels of VWF. As used herein, the term "short-acting FVIII" refers to a FVIII polypeptide in which no extenders of half-life have been added. In one embodiment, short-acting FVIII polypeptides consist of full-length or B domain-deleted FVIII. Examples of short-acting FVIII polypeptides are Advate® and ReFacto®.
[00137] Since the half-life of short-acting FVIII also varies depending at least in part on VWF levels, short-acting FVIII polypeptides can also be used to identify patients that are eligible for a longer dosing interval of a long-acting FVIII polypeptide of the invention. In one embodiment, the short-acting FVIII exhibits an increased half-life of at least about 1.2-fold in individuals expressing high levels of VWF compared to the half-life of the short-acting FVIII when administered to individuals having average levels of VWF. In another embodiment, the short- acting FVIII exhibits an increased half-life of at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold in individuals expressing high levels of VWF compared to the half- life of the short-acting FVIII when administered to individuals having average levels of VWF. Thus, individuals that demonstrate an increased half- life of at least about 1.2-fold when they are administered a short-acting FVIII are eligible for a longer dosing interval with a long-acting FVIII polypeptide of the invention.
[00138] "Dosing interval," as used herein, means the dose of time that elapses between multiple doses being administered to a subject. The comparison of dosing interval may be carried out in a single subject or in a population of subjects and then the average obtained in the population may be calculated.
[00139] The dosing interval when administering a chimeric Factor VIII polypeptide, e.g., a chimeric Factor VIII-Fc polypeptide (a polypeptide comprising a Factor VIII or a hybrid) of the invention may be at least about one and one-half times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII). The dosing interval may be at least about one and one -half to six times longer, one and one-half to five times longer, one and one-half to four times longer, one and one -half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII). The dosing interval may be at least about one and one-half, two, two and one -half, three, three and one-half, four, four and one-half, five, five and one-half or six times longer than the dosing interval required for an equivalent dose of said Factor VIII without the non-Factor VIII portion, e.g., without the Fc portion (a polypeptide consisting of said Factor VIII).. The dosing interval may be about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer. The dosing interval may be at least about one and one-half to 5, one and one-half, 2, 3, 4, or 5 days or longer. For on-demand treatment, the dosing interval of said chimeric polypeptide or hybrid is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
[00140] In one embodiment, the effective dose is 25-80 IU/kg (25, 26, 27, 28, 29, 30, 31, 32, 33,
34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 62, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 IU/kg) and the dosing interval is once every 3-5, 3-6, 3-7, 3, 4, 5, 6, 7, or 8 or more days, or three times per week, or no more than three times per week. In one embodiment, the effective dose is 80 IU/kg and the dosing interval is once every 3 days. In a further embodiment, the effective dose of 80 IU/kg given at a dosing interval of every 3 days is administered to a pediatric subject. In another embodiment, the effective dose is 65 IU/kg and the dosing interval is once weekly, or once every 6-7 days. The doses can be administered repeatedly as long as they are necessary (e.g., at least 10, 20, 28, 30, 40, 50, 52, or 57 weeks, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 years).
[00141] In certain embodiments, the effective dose for on-demand treatment is 20-50IU/Kg (20,
21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 IU/kg). The on-demand treatment can be one time dosing or repeated dosing. For repeated dosing, the dosing interval can be every 12-24 hours, every 24-36 hours, every 24- 48 hours, every 36-48 hours, or every 48-72 hours.
[00142] "Long-acting Factor VIII" is a Factor VIII having an increased half-life (also referred to herein as tl/2, tl/2 beta, elimination half-life and HL) over a reference Factor VIII. The increased half-life of a long-acting Factor VIII may be due to fusion to one or more non-Factor VIII polypeptides such as, e.g., Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof. The increased half-life may be due to one or more modification, such as, e.g., pegylation. Exemplary long-acting Factor VIII polypeptides include, e.g., chimeric Factor VIII polypeptides comprising Fc, chimeric Factor VIII polypeptides comprising XTEN and chimeric Factor VIII polypeptides comprising albumin. Additional exemplary long-acting Factor VIII polypeptides include, e.g., pegylated Factor VIII.
[00143] The "reference" polypeptide, in the case of a long-acting chimeric Factor VIII polypeptide, is a polypeptide consisting essentially of the Factor VIII portion of the chimeric polypeptide, e.g., the same Factor VIII portion without the Fc portion, without the XTEN portion, or without the albumin portion. Likewise, the reference polypeptide in the case of a modified Factor VIII is the same Factor VIII without the modification, e.g., a Factor VIII without the pegylation.
[00144] In some embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a subject:
a mean residence time (MRT) (activity) in said subject of about 14-41.3 hours;
a clearance (CL) (activity) in said subject of about 1.22-5.19 mL/hour/kg or less;
a tl/2beta (activity) in said subject of about 11-26.4 hours;
an incremental recovery (K value) (activity; observed) in said subject of about 1.38-2.88 IU/dL per IU/kg;
a Vss (activity) in said subject of about 37.7-79.4 mL/kg; and
an AUC/dose in said subject of about 19.2-81.7 IU*h/dL per IU/kg. [00145] In some embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a mean incremental recovery (K- Value) (activity; observed) greater that 1.38 IU/dL per IU/kg; a mean incremental recovery (K- Value) (activity; observed) of at least about 1.5, at least about 1.85, or at least about 2.46 IU/dL per IU/kg.
a mean clearance (CL) (activity) in said patient population of about 2.33 ± 1.08 mL/hour/kg or less;
a mean clearance (CL) (activity) in said patient population of about 1.8-2.69 mL/hour/kg;
a mean clearance (CL) (activity) in said patient population that is about 65% of the clearance of a polypeptide comprising said Factor VIII without modification;
a mean mean residence time (MRT) (activity) in said patient population of at least about 26.3 ± 8.33 hours;
a mean MRT (activity) in said patient population of about 25.9 - 26.5 hours;
a mean MRT (activity) in said patent population that is about 1.5 fold longer than the mean MRT of a polypeptide comprising said Factor VIII without modification;
a mean tl/2beta (activity) in said patient population of about 18.3 ± 5.79 hours;
a mean tl/2beta (activity)in said patient population that is about 18 - 18.4 hours;
a mean tl/2beta (activity) in said patient population that is about 1.5 fold longer than the mean tl/2beta of a polypeptide comprising said Factor VIII without modification;
a mean incremental recovery (K value) (activity; observed) in said patient population of about 2.01 ± 0.44 IU/dL per IU/kg;
a mean incremental recovery (K value) (activity; observed) in said patient population of about 1.85 - 2.46 IU/dL per IU/kg;
a mean incremental recovery (K value) (activity; observed) in said patient population that is about 90 % of the mean incremental recovery of a polypeptide comprising said Factor VIII without modification;
a mean Vss (activity) in said patient population of about 55.1 ± 12.3 niL/kg;
a mean Vss (activity) in said patient population of about 45.3 - 56.1 niL/kg;
a mean AUC/dose (activity) in said patient population of about 49.9 ± 18.2 IU*h/dL per IU/kg; a mean AUC/dose (activity) in said patient population of about 44.8 - 57.6 IU*h/dL per IU/kg.
[00146] In other embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a Cmax OBS in said subject administered with the chimeric polypeptide is comparable to the Cmax OBS in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay; a Cmx_OBS in said subject of about 60.5 IU/dL, about 60.5 ± 1 IU/dL, about 60.5 ± 2 IU/dL, about 60.5 ± 3 IU/dL, about 60.5 ± 4 IU/dL, about 60.5 ± 5 IU/dL, about 60.5 ± 6 IU/dL, about 60.5 ± 7 IU/dL, about 60.5 ± 8 IU/dL, about 60.5 ± 9 IU/dL, or about 60.5 ± 10 IU/dL as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered; a Cmax OBS in said subject of about 53.1 - 69 IU/dL as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax_OBS in said subject of about 119 IU/dL, about 119 ± 1 IU/dL, about 119 ± 2 IU/dL, about 119 ± 3 IU/dL, about 119 ± 4 IU/dL, about 119 ± 5 IU/dL, about 119 ± 6 IU/dL, about 119 ± 7 IU/dL, about 119 ± 8 IU/dL, about 119 ± 9 IU/dL, about 119 ± 10 IU/dL, about 119 ± 11 IU/dL, about 119 ± 12 IU/dL, about 119 ± 13 IU/dL, about 119 ± 14 IU/dL, about 119 ± 15 IU/dL, about 119 ± 16 IU/dL, about 119 ± 17 IU/dL, or about 119 ± 18 IU/dL, as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 103 - 136 IU/dL as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a C^OBS in said subject of about 76.5 IU/dL, about 76.5 ± 1 IU/dL, about 76.5 ± 2 IU/dL, about 76.5 ± 3 IU/dL, about 76.5 ± 4 IU/dL, about 76.5 ± 5 IU/dL, about 76.5 ± 6 IU/dL, about 76.5 ± 7 IU/dL, about 76.5 ± 8 IU/dL, about 76.5 ± 9 IU/dL, about 76.5 ± 10 IU/dL, about 76.5 ± 11 IU/dL, about 76.5 ± 12 IU/dL, about 76.5 ± 13 IU/dL, about 76.5 ± 14 IU/dL, or about 76.5 ± 15 IU/dL, as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 64.9 - 90.1 IU/dL as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmx_OBS in said subject of about 182 IU/dL, about 182 ± 2 IU/dL, about 182 ± 4 IU/dL, about 182 ± 6 IU/dL, about 182 ± 8 IU/dL, about 182 ± 10 IU/dL, about 182 ± 12 IU/dL, about 182 ± 14 IU/dL, about 182 ± 16 IU/dL, about 182 ± 18 IU/dL, or about 182 ± 20 IU/dL as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; or
a Cmax_OBS in said subject of about 146 - 227 IU/dL, about 146 ± 5 IU/dL, about 146 ± 10 IU/dL, about 227 ± 5 IU/dL, or about 146 ± 10 IU/dL as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
[00147] In certain embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a tl/2beta (activity) in said subject that is at least 1.48, 1.49, 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, or 1.90 times higher than the tl/2beta (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 18.8 hours, 18.8 ± 1 hours, 18.8 ± 1 hours, 18.8 ± 2 hours, 18.8 ± 3 hours, 18.8 ± 4 hours, 18.8 ± 5 hours, 18.8 ± 6 hours, 18.8 ± 7 hours, 18.8 ± 8 hours, 18.8 ± 9 hours, 18.8 ± 10 hours, or 18.8 ± 11 hours as measured by a one stage (aPTT) assay; a tl/2beta (activity) in said subject of about 14.3 - 24.5 hours as measured by a one stage (aPTT) assay;
a tl/2beta (activity) in said subject of about 16.7 hours, 16.7 ± 1 hours, 16.7 ± 2 hours, 16.7 ± 3 hours, 16.7 ± 4 hours, 16.7 ± 5 hours, 16.7 ± 6 hours, 16.7 ± 7 hours, 16.7 ± 8 hours, 16.7 ± 9 hours, 16.7 ± 10 hours, or 16.7 ± 11 hours as measured by a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 13.8 - 20.1 hours as measured by a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 19.8 hours, 19.8 ± 1 hours, 19.8 ± 2 hours, 19.8 ± 3 hours, 19.8 ± 4 hours, 19.8 ± 5 hours, 19.8 ± 6 hours, 19.8 ± 7 hours, 19.8 ± 8 hours, 19.8 ± 9 hours, 19.8 ± 10 hours, or 19.8 ± 11 hours as measured by a two stage (chromogenic) assay; or
a tl/2beta (activity) in said subject of about 14.3 - 27.5 hours as measured by a two stage (chromogenic) assay.
[00148] In certain embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a clearance (CL) (activity) in said subject is 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, or 0.70 times lower than the clearance in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a clearance (CL) (activity) in said subject of about 1.68 mL/hour/kg, 1.68 ± 0.1 mL/hour/kg, 1.68 ± 0.2 mL/hour/kg, 1.68 ± 0.3 mL/hour/kg, 1.68 ± 0.4 mL/hour/kg, 1.68 ± 0.5 mL/hour/kg, 1.68 ± 0.6 mL/hour/kg, or 1.68 ± 0.7 mL/hour/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.31 - 2.15 mL/hour/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 2.32 mL/hour/kg, 2.32 ± 0.1 mL/hour/kg, 2.32 ± 0.2 mL/hour/kg, 2.32 ± 0.3 mL/hour/kg, 2.32 ± 0.4 mL/hour/kg, 2.32 ± 0.5 mL/hour/kg, 2.32 ± 0.6 mL/hour/kg, or 2.32 ± 0.7 mL/hour/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.64 - 3.29 mL/hour/kg as measured by oa ne stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.49 mL/hour/kg, 1.49 ± 0.1 mL/hour/kg, 1.49 ± 0.2 mL/hour/kg, 1.49 ± 0.3 mL/hour/kg, 1.49 ± 0.4 mL/hour/kg, 1.49 ± 0.5 mL/hour/kg, 1.49 ± 0.6 mL/hour/kg, or 1.49 ± 0.7 mL/hour/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.16 - 1.92 mL/hour/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; a clearance (CL) (activity) in said subject of about 1.52 mL/hour/kg, 1.52 ± 0.1 mL/hour/kg, 1.52 ± 0.2 mL/hour/kg, 1.52 ± 0.3 mL/hour/kg, 1.52 ± 0.4 mL/hour/kg, 1.52 ± 0.5 mL/hour/kg, 1.52 ± 0.6 mL/hour/kg, or 1.52 ± 0.7 mL/hour/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; or
a clearance (CL) (activity) in said subject of about 1.05-2.20 mL/hour/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
[00149] In some embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a MRT in said subject is at least 1.46, 1.47, 1.48, 1.49, 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.90, 1.91, 1.92, or 1.93 times higher than the MRT in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a MRT (activity) in said subject of about 27 hours, 27 ± 1 hours, 27 ± 2 hours, 27 ± 3 hours, 27 ± 4 hours, 27 ± 5 hours, 27 ± 6 hours, 27 ± 7 hours, 27 ± 8 hours, 27 ± 9 hours, or 27 ± 10 hours as meansured by a one stage (aPTT) assay;
a MRT (activity) in said subject of about 20.6 - 35.3 hours as meansured by a one stage (aPTT) assay;
a MRT (activity) in said subject of about 23.9 - 28.5 hours as measured by a two stage (chromogenic) assay;
a MRT (activity) in said subject of about 19.8 - 28.9 hours as measured by a two stage (chromogenic) assay; or
a MRT (activity) in said subject of about 20.5 - 39.6 hours as measured by a two stage (chromogenic) assay.
[00150] In other embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
an incremental recovery in said subject that is comparable to the Incremental Recovery in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
an incremental recovery in said subject of about 2.44 IU/dL per IU/kg, 2.44 ± 0.1 IU/dL per IU/kg, 2.44 ± 0.2 IU/dL per IU/kg, 2.44 ± 0.3 IU/dL per IU/kg, 2.44 ± 0.4 IU/dL per IU/kg, 2.44 ± 0.5 IU/dL per IU/kg, 2.44 ± 0.6 IU/dL per IU/kg, 2.44 ± 0.7 IU/dL per IU/kg, 2.44 ± 0.8 IU/dL per IU/kg, 2.44 ± 0.9 IU/dL per IU/kg, 2.44 ± 1.0 IU/dL per IU/kg, 2.44 ± 1.1 IU/dL per IU/kg, or 2.44 ± 1.2 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered; an incremental recovery in said subject of about 2.12 - 2.81 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 1.83 IU/dL per IU/kg, 1.83 ± 0.1 IU/dL per IU/kg, 1.83 ± 0.2 IU/dL per IU/kg, 1.83 ± 0.3 IU/dL per IU/kg, 1.83 ± 0.4 IU/dL per IU/kg, 1.83 ± 0.5 IU/dL per IU/kg, 1.83 ± 0.6 IU/dL per IU/kg, 1.83 ± 0.7 IU/dL per IU/kg, 1.83 ± 0.8 IU/dL per IU/kg, 1.83 ± 0.9 IU/dL per IU/kg, 1.83 ± 1.0 IU/dL per IU/kg, or 1.83 ± 1.1 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 1.59 - 2.10 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 3.09 IU/dL per IU/kg, 3.09 ± 0.1 IU/dL per IU/kg, 3.09 ± 0.2 IU/dL per IU/kg, 3.09 ± 0.3 IU/dL per IU/kg, 3.09 ± 0.4 IU/dL per IU/kg, 3.09 ± 0.5 IU/dL per IU/kg, 3.09 ± 0.6 IU/dL per IU/kg, 3.09 ± 0.7 IU/dL per IU/kg, 3.09 ± 0.8 IU/dL per IU/kg, 3.09 ± 0.9 IU/dL per IU/kg, 3.09 ± 1.0 IU/dL per IU/kg, 3.09 ± 1.1 IU/dL per IU/kg, 3.09 ± 1.2 IU/dL per IU/kg, or 3.09 ± 1.3 IU/dL per IU/kg, as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 2.80 IU/dL per IU/kg, 2.80 ± 0.1 IU/dL per IU/kg, 2.80 ± 0.2 IU/dL per IU/kg, 2.80 ± 0.3 IU/dL per IU/kg, 2.80 ± 0.4 IU/dL per IU/kg, 2.80 ± 0.5 IU/dL per IU/kg, 2.80 ± 0.6 IU/dL per IU/kg, 2.80 ± 0.7 IU/dL per IU/kg, 2.80 ± 0.8 IU/dL per IU/kg, 2.80 ± 0.9 IU/dL per IU/kg, 2.80 ± 1.0 IU/dL per IU/kg, 2.80 ± 1.1 IU/dL per IU/kg, or 2.80 ± 1.2 IU/dL per IU/kg, as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 2.61-3.66 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; or
an incremental recovery in said subject of about 2.24-3.50 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
[00151] In still other embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
a Vss (activity) in said subject that is comparable to the Vss (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a Vss (activity) in said subject of about 45.5 mL/kg, 45.5 ± 1 mL/kg, 45.5 ± 2 mL/kg, 45.5 ± 3 mL/kg, 45.5 ± 4 mL/kg, 45.5 ± 5 mL/kg, 45.5 ± 6 mL/kg, 45.5 ± 7 mL/kg, 45.5 ± 8 mL/kg, 45.5 ± 9 mL/kg, 45.5 ± 10 mL/kg, or 45.5 ± 11 mL/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 39.3 - 52.5 mL/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered; a Vss (activity) in said subject of about 62.8 mL/kg, 62.8 ± 1 mL/kg, 62.8 ± 2 mL/kg, 62.8 ± 3 mL/kg, 62.8 ± 4 mL/kg, 62.8 ± 5 mL/kg, 62.8 ± 6 mL/kg, 62.8 ± 7 mL/kg, 62.8 ± 8 mL/kg, 62.8 ± 9 mL/kg, 62.8 ± 10 mL/kg, 62.8 ± 1 1 mL/kg, 62.8 ± 12 mL/kg, 62.8 ± 13 mL/kg, 62.8 ± 14 mL/kg, 62.8 ± 15 mL/kg, or 62.8 ± 16 mL/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 55.2 - 71.5 mL/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 35.9 mL/kg, 35.9 ± 1 mL/kg, 35.9 ± 2 mL/kg, 35.9 ± 3 mL/kg, 35.9 ± 4 mL/kg, 35.9 ± 5 mL/kg, 35.9 ± 6 mL/kg, 35.9 ± 7 mL/kg, 35.9 ± 8 mL/kg, 35.9 ± 9 mL/kg, 35.9 ± 10 mL/kg, 35.9 ± 1 1 mL/kg, 35.9 ± 12 mL/kg, or 35.9 ± 13 mL/kg, as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 30.4-42.3 mL/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 43.4 mL/kg, 43.4 ± 1 mL/kg, 43.4 ± 2 mL/kg, 43.4 ± 3 mL/kg, 43.4 ± 4 mL/kg, 43.4 ± 5 mL/kg, 43.4 ± 6 mL/kg, 43.4 ± 7 mL/kg, 43.4 ± 8 mL/kg, 43.4 ± 9 mL/kg, 43.4 ± 10 mL/kg, 43.4 ± 1 1 mL/kg, 43.4 ± 12 mL/kg, 43.4 ± 13 mL/kg, 43.4 ± 14 mL/kg, 43.4 ± 15 mL/kg, or 43.4 ± 16 mL/kg, as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; or
a Vss (activity) in said subject of about 38.2-49.2 mL/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
[00152] In yet other embodiments, the long-acting Factor VIII has one or more of the following properties when administered to a patient population:
an AUCiNF in said subject that is at least 1.45 1.46, 1.47, 1.48, 1.49, 1.50, 1.51 , 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61 , 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71 , 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81 , 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.90 times higher than the AUC in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
an AUCiNF in said subject of about 1440 ± 316 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1 160 - 1880 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUC in said subject of about 1480 hr*IU/dL per IU/kg, 1480 ± 100 hr*IU/dL per IU/kg, 1480 ± 200 hr*IU/dL per IU/kg, 1480 ± 300 hr*IU/dL per IU/kg, 1480 ± 400 hr*IU/dL per IU/kg, 1480 ± 500 hr*IU/dL per IU/kg, 1480 ± 600 hr*IU/dL per IU/kg, 1480 ± 700 hr*IU/dL per IU/kg, 1480 ± 800 hr*IU/dL per IU/kg, 1480 ± 900 hr*IU/dL per IU/kg, or 1480 ± 1000 hr*IU/dL per IU/kg, as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCINP in said subject of about 2910 ± 1320 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1980 - 3970 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUC in said subject of about 2800 hr*IU/dL per IU/kg, 2800 ± 100 hr*IU/dL per IU/kg, 2800 ± 200 hr*IU/dL per IU/kg, 2800 ± 300 hr*IU/dL per IU/kg, 2800 ± 400 hr*IU/dL per IU/kg, 2800 ± 500 hr*IU/dL per IU/kg, 2800 ± 600 hr*IU/dL per IU/kg, 2800 ± 700 hr*IU/dL per IU/kg, 2800 ± 800 hr*IU/dL per IU/kg, 2800 ± 900 hr*IU/dL per IU/kg, or 2800 ± 1000 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCW in said subject of about 1660 hr*IU/dL per IU/kg, 1660 ± 100 hr*IU/dL per IU/kg, 1660 ± 200 hr*IU/dL per IU/kg, 1660 ± 300 hr*IU/dL per IU/kg, 1660 ± 400 hr*IU/dL per IU/kg, 1660 ± 500 hr*IU/dL per IU/kg, 1660 ± 600 hr*IU/dL per IU/kg, 1660 ± 700 hr*IU/dL per IU/kg, 1660 ± 800 hr*IU/dL per IU/kg, 1660 ± 900 hr*IU/dL per IU/kg, or 1660 ± 1000 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; an AUCiNF in said subject of about 1300 - 2120 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCW in said subject of about 4280 hr*IU/dL per IU/kg, 4280 ± 100 hr*IU/dL per IU/kg, 4280 ± 200 hr*IU/dL per IU/kg, 4280 ± 300 hr*IU/dL per IU/kg, 4280 ± 400 hr*IU/dL per IU/kg, 4280 ± 500 hr*IU/dL per IU/kg, 4280 ± 600 hr*IU/dL per IU/kg, 4280 ± 700 hr*IU/dL per IU/kg, 4280 ± 800 hr*IU/dL per IU/kg, 4280 ± 900 hr*IU/dL per IU/kg, 4280 ± 1000 hr*IU/dL per IU/kg, 4280 ± 1 100 hr*IU/dL per IU/kg, 4280 ± 1200 hr*IU/dL per IU/kg, 4280 ± 1300 hr*IU/dL per IU/kg, 4280 ± 1400 hr*IU/dL per IU/kg, 4280 ± 1500 hr*IU/dL per IU/kg, or 4280 ± 1600 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; or an AUCiNF in said subject of about 2960 - 6190 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
[00153] "On-demand treatment," as used herein, means treatment that is intended to take place over a short course of time and is in response to an existing condition, such as a bleeding episode, or a perceived need such as planned surgery. Conditions that may require on-demand treatment include, e.g., a bleeding episode, hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath. The subject may be in need of surgical prophylaxis, peri-operative management, or treatment for surgery. Such surgeries include, e.g., minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
[00154] In one embodiment, on-demand treatment resolves greater than 80% (greater than 80%>, greater than 81%>, greater than 82%, greater than 83%, greater than 84%, greater than 85%, greater than 86%, greater than 87%, greater than 88%, greater than 89%, greater than 90%, greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, greater than 99%, or 100%) or 80-100%, 80-90%, 85-90%, 90-100%, 90-95%, or 95-100% of bleeds (e.g., spontaneous bleeds) in a single dose. In another embodiment, greater than 80% (greater than 81%, greater than 82%, greater than 83%o, greater than 84%, greater than 85%, greater than 86%, greater than 87%, greater than 88%, greater than 89%, greater than 90%, greater than 91%, greater than 92%, greater than 93%, greater than 94%, greater than 95%, greater than 96%, greater than 97%, greater than 98%, or 100%) or 80-100%, 80-90%, 85-90%, 90-100%, 90-95%, or 95-100% of bleeding episodes are rated excellent or good by physicians after on-demand treatment. In other embodiments, greater than 5%o, (greater than 6%, greater than 7%, greater than 8%, greater than 9%, greater than 10%, greater than 11%, greater than 12%, greater than 13%, greater than 14%, greater than 15%, greater than 16%, greater than 17%, greater than 18%, greater than 19%, greater than 20%), or 5- 20%o, 5-15%), 5-10%), 10-20%), or 10-15% of bleeding episodes are rated as fair by physicians after on-demand treatment.
[00155] "Polypeptide," "peptide" and "protein" are used interchangeably and refer to a polymeric compound comprised of covalently linked amino acid residues.
[00156] "Polynucleotide" and "nucleic acid" are used interchangeably and refer to a polymeric compound comprised of covalently linked nucleotide residues. Polynucleotides may be DNA, cDNA, RNA, single stranded, or double stranded, vectors, plasmids, phage, or viruses. Polynucleotides include, e.g., those in Table 1, which encode the polypeptides of Table 2 (see Table 1). Polynucleotides also include, e.g., fragments of the polynucleotides of Table 1, e.g., those that encode fragments of the polypeptides of Table 2, such as the Factor VIII, Fc, signal sequence, 6His and other fragments of the polypeptides of Table 2.
[00157] "Prophylactic treatment," as used herein, means administering a Factor VIII polypeptide in multiple doses to a subject over a course of time to increase the level of Factor VIII activity in a subject's plasma. The increased level can be sufficient to decrease the incidence of spontaneous bleeding or to prevent bleeding, e.g., in the event of an unforeseen injury. During prophylactic treatment, the plasma protein level in the subject may not fall below the baseline level for that subject, or below the level of Factor VIII that characterizes severe hemophilia (<1 IU/dl [1%]). [00158] In one embodiment, the prophylaxis regimen is "tailored" to the individual patient, for example, by determining PK data for each patient and administering Factor VIII of the invention at a dosing interval that maintains a trough level of 1-3% FVIII activity. Adjustments may be made when a subject experiences unacceptable bleeding episodes defined as >2 spontaneous bleeding episodes over a rolling two-month period. In this case, adjustment will target trough levels of 3-5%. In another embodiment, prophylactic treatment results in prevention and control of bleeding, sustained control of bleeding, sustained protection from bleeding, and/or sustained benefit. Prophylaxis, e.g., sustained protection can be demonstrated by an increased AUC to last measured time point (AUC -LAST) and reduced clearance, resulting in increased terminal tl/2 compared to short acting FVIII. Prophylaxis can be demonstrated by better Cmax, better Tmax, and/or greater mean residence time versus short-acting FVIII. In some embodiments, prophylaxis results in no spontaneous bleeding episodes within about 24, 36, 48, 72, or 96 hours (e.g., 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 96, 87, 88, 89, 90, 91, 92, 93, 94, 95, or 96 hours), after injection (e.g., the last injection). In certain embodiments, prophylaxis results in greater than 30%> (e.g., greater than 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 96, 87, 88, 89, or 90%, for example, greater than 50%), mean reduction in annualized bleeding episodes with once weekly dosing (e.g., at 65 IU/kg). "Therapeutic dose," as used herein, means a dose that achieves a therapeutic goal, as described herein. The calculation of the required dosage of factor VIII is based upon the empirical finding that, on average, 1 IU of factor VIII per kg body weight raises the plasma factor VIII activity by approximately 2 IU/dL. The required dosage is determined using the following formula:
Required units = body weight (kg) x desired factor VIII rise (IU/dL or % of normal) x 0.5 (IU/kg per IU/dL)
[00159] The therapeutic doses that may be used in the methods of the invention are about 10-100
IU/kg, more specifically, 10-20, 20-30, 30-40, 40-50, 50-60, 60-70, 70-80, 80-90, or 90-100 IU/kg, and more specifically, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
[00160] Additional therapeutic doses that may be used in the methods of the invention are about
10 to about 150 IU/kg, more specifically, about 100-110, 110-120, 120-130, 130-140, 140-150 IU/kg, and more specifically, about 110, 115, 120, 125, 130, 135, 140, 145, or 150 IU/kg.
[00161] "Variant," as used herein, refers to a polynucleotide or polypeptide differing from the original polynucleotide or polypeptide, but retaining essential properties thereof, e.g., factor VIII coagulant activity or Fc (FcRn binding) activity. Generally, variants are overall closely similar, and, in many regions, identical to the original polynucleotide or polypeptide. Variants include, e.g., polypeptide and polynucleotide fragments, deletions, insertions, and modified versions of original polypeptides.
[00162] Variant polynucleotides may comprise, or alternatively consist of, a nucleotide sequence which is at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for example, the nucleotide coding sequence in SEQ ID NO: l, 3, or 5 (the factor VIII portion, the Fc portion, individually or together) or the complementary strand thereto, the nucleotide coding sequence of known mutant and recombinant factor VIII or Fc such as those disclosed in the publications and patents cited herein or the complementary strand thereto, a nucleotide sequence encoding the polypeptide of SEQ ID NO:2, 4, or 6 (the factor VIII portion, the Fc portion, individually or together), and/or polynucleotide fragments of any of these nucleic acid molecules (e.g., those fragments described herein). Polynucleotides which hybridize to these nucleic acid molecules under stringent hybridization conditions or lower stringency conditions are also included as variants, as are polypeptides encoded by these polynucleotides as long as they are functional.
[00163] Variant polypeptides may comprise, or alternatively consist of, an amino acid sequence which is at least 85%, 90%, 95%, 96%, 97%, 98%, 99% identical to, for example, the polypeptide sequence shown in SEQ ID NOS:2, 4, or 6 (the factor VIII portion, the Fc portion, individually or together), and/or polypeptide fragments of any of these polypeptides (e.g., those fragments described herein).
[00164] By a nucleic acid having a nucleotide sequence at least, for example, 95% "identical" to a reference nucleotide sequence, it is intended that the nucleotide sequence of the nucleic acid is identical to the reference sequence except that the nucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence. In other words, to obtain a nucleic acid having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. The query sequence may be, for example, the entire sequence shown in SEQ ID NO: l or 3, the ORF (open reading frame), or any fragment specified as described herein.
[00165] As a practical matter, whether any particular nucleic acid molecule or polypeptide is at least 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence or polypeptide of the present invention can be determined conventionally using known computer programs. In one embodiment, a method for determining the best overall match between a query sequence (reference or original sequence) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et ah, Comp. App. Biosci. 6:237-245 (1990), which is herein incorporated by reference in its entirety In a sequence alignment the query and subject sequences are both DNA sequences. An RNA sequence can be compared by converting U's to T's. The result of said global sequence alignment is in percent identity. In another embodiment, parameters used in a FASTDB alignment of DNA sequences to calculate percent identity are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=l, Joining Penalty=30, Randomization Group Length=0, Cutoff Score=l, Gap Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the length of the subject nucleotide sequence, whichever is shorter.
[00166] If the subject sequence is shorter than the query sequence because of 5' or 3' deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for 5' and 3' truncations of the subject sequence when calculating percent identity. For subject sequences truncated at the 5' or 3' ends, relative to the query sequence, the percent identity is corrected by calculating the number of bases of the query sequence that are 5' and 3' of the subject sequence, which are not matched/aligned, as a percent of the total bases of the query sequence. Whether a nucleotide is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
[00167] For example, a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity. The deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end. The 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%. In another example, a 90 base subject sequence is compared with a 100 base query sequence. This time the deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
[00168] By a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a query amino acid sequence of the present invention, it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a query amino acid sequence, up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, (indels) or substituted with another amino acid. These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
[00169] As a practical matter, whether any particular polypeptide is at least 85%, 90%>, 95%, 96%,
97%), 98%) or 99% identical to, for instance, the amino acid sequences of SEQ ID NO:2 (the factor VIII portion, the Fc portion, individually or together) or 4, or a known factor VIII or Fc polypeptide sequence, can be determined conventionally using known computer programs. In one embodiment, a method for determining the best overall match between a query sequence (reference or original sequence) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al., Comp. App. Biosci. 6:237-245(1990), incorporated herein by reference in its entirety. In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is in percent identity. In another embodiment, parameters used in a FASTDB amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=l, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=l, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
[00170] If the subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N- and C-terminal truncations of the subject sequence when calculating global percent identity. For subject sequences truncated at the N- and C-termini, relative to the query sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C- terminal residues of the subject sequence.
[00171] For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus. The 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C- termini of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
[00172] The polynucleotide variants may contain alterations in the coding regions, non-coding regions, or both. In one embodiment, the polynucleotide variants contain alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. In another embodiment, nucleotide variants are produced by silent substitutions due to the degeneracy of the genetic code. In other embodiments, variants in which 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to others, e.g., a bacterial host such as E. coli).
[00173] Naturally occurring variants are called "allelic variants," and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
[00174] Using known methods of protein engineering and recombinant DNA technology, variants may be generated to improve or alter the characteristics of the polypeptides. For instance, one or more amino acids can be deleted from the N-terminus or C-terminus of the secreted protein without substantial loss of biological function. Ron et al., J. Biol. Chem. 268: 2984-2988 (1993), incorporated herein by reference in its entirety, reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al, J. Biotechnology 7:199-216 (1988), incorporated herein by reference in its entirety.)
[00175] Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurring protein. For example, Gayle and coworkers (J. Biol. Chem 265:22105-22111 (1993), incorporated herein by reference in its entirety) conducted extensive mutational analysis of human cytokine IL-la. They used random mutagenesis to generate over 3,500 individual IL-la mutants that averaged 2.5 amino acid changes per variant over the entire length of the molecule. Multiple mutations were examined at every possible amino acid position. The investigators found that "[m]ost of the molecule could be altered with little effect on either [binding or biological activity]." (See Abstract.) In fact, only 23 unique amino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type.
[00176] As stated above, polypeptide variants include, e.g., modified polypeptides. Modifications include, e.g., acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation (Mei et al., Blood 776:270-79 (2010), which is incorporated herein by reference in its entirety), proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. In some embodiments, Factor VIII is modified, e.g., pegylated, at any convenient location. In some embodiments, Factor VIII is pegylated at a surface exposed amino acid of Factor VIII, e.g., a surface exposed cysteine, which may be an engineered cysteine. Id.. In some embodiments, modified Factor VIII, e.g., pegylated Factor VIII, is a long-acting Factor VIII.
[00177] "Volume of distribution at steady state (Vss)," as used herein, has the same meaning as the term used in pharmacology, which is the apparent space (volume) into which a drug distributes. Vss = the amount of drug in the body divided by the plasma concentration at steady state.
[00178] "About," as used herein for a range, modifies both ends of the range. Thus, "about 10-20" means "about 10 to about 20." [00179] The chimeric polypeptide used herein can comprise processed Factor VIII or single chain
Factor VIII or a combination thereof. "Processed Factor VIII," as used herein means Factor VIII that has been cleaved at Arginine 1648 (for full-length Factor VIII) or Arginine 754 (for B- domain deleted Factor VIII), i.e., intracellular processing site. Due to the cleavage at the intracellular processing site, processed Factor VIII comprises two polypeptide chains, the first chain being a heavy chain and the second chain being a light chain. For example, the processed Factor VIII-Fc fusion protein (i.e., Heavy chain and Light chain fused to Fc) run at approximately 90 kDa and 130 kDa on a non-reducing SDS-PAGE, respectively, and 90 kDa and 105 kDa on a reducing SDS-PAGE, respectively. Therefore, in one embodiment, at least about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII. In another embodiment, about 50%>, about 60%>, about 70%>, about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%> of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII. In a particular embodiment, the chimeric polypeptide comprising processed Factor VIII is purified (or isolated) from the chimeric polypeptide comprising single chain Factor VIII, and at least about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%) of the Factor VIII portion in the chimeric polypeptide is processed Factor VIII.
[00180] "Single chain Factor VIII," "SC Factor VIII," or "SCFVIII" as used herein means Factor
VIII that has not been cleaved at the Arginine site (residue 1648 for full-length Factor VIII (i.e., residue 1667 of SEQ ID NO: 6) or residue 754 for B-domain deleted Factor VIII (i.e., residue 773 of SEQ ID NO: 2). Therefore, single chain Factor VIII in the chimeric polypeptide used herein comprises a single chain. In one embodiment, the single chain Factor VIII contains an intact intracellular processing site. In another embodiment, the single chain Factor VIII of the invention comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645, a substitution or mutation at an amino acid position corresponding to Arginine 1648, or a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII. In other embodments, the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648. In certain embodiments, the substitution or mutation is an amino acid other than arginine, e.g., isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, histidine, ornithine, pyrrolysine, or taurine. The single chain Factor VIII-Fc fusion protein can run at approximately 220 kDa on a non reducing SDS-PAGE and at approximately 195 kDa on a reducing SDS-PAGE. [00181] In one embodiment, the chimeric polypeptide comprising single chain Factor VIII is purified (or isolated) from the chimeric polypeptide comprising processed Factor VIII, and at least about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 99%, or about 100% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII. In another embodiment, at least about 1%>, about 5%), about 10%>, about 15%>, about 20%>, about 25%>, about 30%>, or about 35%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII. In other embodiments, about 1%-about 10%, about 5%-about 15%, about 10%-about 20%, about 15%-about 25%, about 20%- about 30%, about 25%-about 35%, about 30%-about 40% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII. In a particular embodiment, about 1%, about 5%, about 10%, about 15%, about 20% about 25%, about 30%, about 35% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII. In other embodiments, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% of the Factor VIII portion of the chimeric polypeptide used herein is single chain Factor VIII. In some embodiments, the ratio of the single chain Factor VIII to the processed Factor VIII of the chimeric polypeptide is (a) about 25%> of single chain Factor VIII and about 75%> of processed Factor VIII; (b) about 20%) of single chain Factor VIII and about 80%> of processed Factor VIII; (c) about 15%> of single chain Factor VIII and about 85%> of processed Factor VIII; (d) about 10%> of single chain Factor VIII and about 90%> of processed Factor VIII; (e) about 5%> of single chain Factor VIII and about 95%) of processed Factor VIII; (f) about 1%> of single chain Factor VIII and about 99%> of processed Factor VIII; (g) about 100% of processed Factor VIII, (h) about 30%> of single chain Factor VIII and about 70%> of processed Factor VIII, (i) about 35%> of single chain Factor VIII and about 65%> of processed Factor VIII, or (j) about 40%> of single chain Factor VIII and about 60%) of processed Factor VIII. In other embodiments, the ratio of the single chain Factor VIII to the processed Factor VIII of the chimeric polypeptide is (a) about 30%> of single chain Factor VIII and about 70%> of processed Factor VIII; (b) about 40%> of single chain Factor VIII and about 60%) of processed Factor VIII; (c) about 50%> of single chain Factor VIII and about 50%> of processed Factor VIII; (d) about 60%> of single chain Factor VIII and about 40%> of processed Factor VIII; (e) about 70%> of single chain Factor VIII and about 30%> of processed Factor VIII; (f) about 80%) of single chain Factor VIII and about 20%> of processed Factor VIII; (g) about 90%> of single chain Factor VIII and about 10%> of processed Factor VIII; (h) about 95%> of single chain Factor VIII and about 5%> of processed Factor VIII; (i) about 99%> of single chain Factor VIII and about 1%) of processed Factor VIII; or (j) about 100% of single chain Factor VIII.
[00182] The Factor VIII portion in the chimeric polypeptide used herein has Factor VIII activity.
Factor VIII activity can be measured by any known methods in the art. For example, one of those methods can be a chromogenic assay. The chromogenic assay mechanism is based on the principles of the blood coagulation cascade, where activated Factor VIII accelerates the conversion of Factor X into Factor Xa in the presence of activated Factor IX, phospholipids and calcium ions. The Factor Xa activity is assessed by hydrolysis of a p-nitroanilide (pNA) substrate specific to Factor Xa. The initial rate of release of p-nitroaniline measured at 405 nM is directly proportional to the Factor Xa activity and thus to the Factor VIII activity in the sample. The chromogenic assay is recommended by the Factor VIII and Factor IX Subcommittee of the Scientific and Standardization Committee (SSC) of the International Society on Thrombosis and Hemostatsis (ISTH). Since 1994, the chromogenic assay has also been the reference method of the European Pharmacopoeia for the assignment of FVIII concentrate potency. Thus, in one embodiment, the chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions), when the Factor VIII activity is measured in vitro by a chromogenic assay.
[00183] In another embodiment, the chimeric polypeptide comprising single chain Factor VIII of this invention has a Factor Xa generation rate comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
[00184] In order to activate Factor X to Factor Xa, activated Factor IX (Factor IXa) hydrolyses one arginine-isoleucine bond in Factor X to form Factor Xa in the presence of Ca2+, membrane phospholipids, and a Factor VIII cofactor. Therefore, the interaction of Factor VIII with Factor IX is critical in coagulation pathway. In certain embodiments, the chimeric polypeptide comprising single chain factor VIII can interact with Factor IXa at a rate comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
[00185] In addition, Factor VIII is bound to von Willebrand Factor while inactive in circulation.
Factor VIII degrades rapidly when not bound to vWF and is released from vWF by the action of thrombin. In some embodiments, the chimeric polypeptide comprising single chain Factor VIII binds to von Willebrand Factor at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions). [00186] Factor VIII can be inactivated by activated protein C in the presence of calcium and phospholipids. Activated protein C cleaves Factor VIII heavy chain after Arginine 336 in the Al domain, which disrupts a Factor X substrate interaction site, and cleaves after Arginine 562 in the A2 domain, which enhances the dissociation of the A2 domain as well as disrupts an interaction site with the Factor IXa. This cleavage also bisects the A2 domain (43 kDa) and generates A2-N (18 kDa) and A2-C (25 kDa) domains. Thus, activated protein C can catalyze multiple cleavage sites in the heavy chain. In one embodiment, the chimeric polypeptide comprising single chain Factor VIII is inactivated by activated Protein C at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions).
[00187] In other embodiments, the chimeric polypeptide comprising single chain Factor VIII has
Factor VIII activity in vivo comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions). In a particular embodiment, the chimeric polypeptide comprising single chain Factor VIII is capable of protecting a HemA mouse at a level comparable to a chimeric polypeptide comprising processed Factor VIII (e.g., a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions) in a HemA mouse tail vein transection model.
[00188] The term "comparable" as used herein means a compared rate or level resulted from using the chimeric polypeptide is equal to, substantially equal to, or similar to the reference rate or level. The term "similar" as used herein means a compared rate or level has a difference of no more than 10% or no more than 15%> from the reference rate or level (e.g., FXa generation rate by a chimeric polypeptide consisting essentially of or consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions). The term "substantially equal" means a compared rate or level has a difference of no more than 0.01%, 0.5%) or 1%) from the reference rate or level.
[00189] The present invention further includes a composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%>, about 40%>, about 50%>, about 60%>, about 70%, about 80%, about 85%, about 90%, about 95%, or about 99% of the chimeric polypeptide comprises a Factor VIII portion, which is single chain Factor VIII and a second portion. In another embodiment, about 30%>, about 40%>, about 50%>, about 60%>, about 70%>, about 80%>, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% of the chimeric polypeptide in the composition is single chain Factor VIII. In other embodiments, the second portion is an Fc, XTEN, albumin, a PAS sequence, transferrin, CTP (28 amino acid C- terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or two or more combinations thereof. In still other embodiments, the composition of the present invention comprises a combination of a chimeric polypeptide comprising processed Factor VIII and a chimeric polypeptide comprising single chain Factor VIII, (a) wherein about 30% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 70%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (b) wherein about 40%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 60%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (c) wherein about 50%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII, and about 50%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (d) wherein about 60%) of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 40%o of the Factor VIII portion of the chimeric polypeptide being processed Factor VIII; (e) wherein about 70%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 30%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (f) wherein about 80%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 20%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (g) wherein about 90%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 10%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (h) wherein about 95% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 5% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; (i) wherein about 99% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 1%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII; or (j) wherein about 100% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
[00190] In certain embodiments, the composition of the present invention has Factor VIII activity comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one of the two Fc portions), when the Factor VIII activity is measured in vitro by a chromogenic assay.
[00191] In other embodiments, the composition of the invention has a Factor Xa generation rate comparable to a composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions). In still other embodiments, the composition comprising single chain factor VIII can interact with Factor IXa at a rate comparable to a composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc). In further embodiments, the single chain Factor VIII in the chimeric polypeptide of the present composition is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide of a composition (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions). In a particular embodiment, the composition comprising single chain Factor VIII has Factor VIII activity in vivo comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein the processed Factor VIII is fused to one Fc of the two Fc portions). In some embodiments, the composition comprising single chain Factor VIII of the invention is capable of protecting HemA mouse at a level comparable to the composition comprising processed Factor VIII (e.g., a composition comprising a chimeric polypeptide, which consists essentially of or consists of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions) in HemA mouse tail vein transection model.
[00192] The present invention further provides a method for treating a bleeding condition in a human subject using the composition of the invention. An exemplary method comprises administering to the subject in need thereof a therapeutically effective amount of a pharmaceutical composition/formulation comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 85%, about 90%), about 95%), or about 99%> of the chimeric polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion.
[00193] The bleeding condition can be caused by a blood coagulation disorder. A blood coagulation disorder can also be referred to as a coagulopathy. In one example, the blood coagulation disorder, which can be treated with a pharmaceutical composition of the current disclosure, is hemophilia or von Willebrand disease (vWD). In another example, the blood coagulation disorder, which can be treated with a pharmaceutical composition of the present disclosure is hemophilia A.
[00194] In some embodiments, the type of bleeding associated with the bleeding condition is selected from hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intraabdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, and bleeding in the illiopsoas sheath. [00195] In other embodiments, the subject suffering from bleeding condition is in need of treatment for surgery, including, e.g., surgical prophylaxis or peri-operative management. In one example, the surgery is selected from minor surgery and major surgery. Exemplary surgical procedures include tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, joint replacement surgery (e.g., total knee replacement, hip replacement, and the like), heart surgery, and caesarean section.
[00196] In another example, the subject is concomitantly treated with FIX. Because the compounds of the invention are capable of activating FIXa, they could be used to pre-activate the FIXa polypeptide before administration of the FIXa to the subject.
[00197] The methods of the invention may be practiced on a subject in need of prophylactic treatment or on-demand treatment.
[00198] The pharmaceutical compositions comprising at least 30% of single chain Factor VIII may be formulated for any appropriate manner of administration, including, for example, topical (e.g., transdermal or ocular), oral, buccal, nasal, vaginal, rectal or parenteral administration.
[00199] The term parenteral as used herein includes subcutaneous, intradermal, intravascular (e.g., intravenous), intramuscular, spinal, intracranial, intrathecal, intraocular, periocular, intraorbital, intrasynovial and intraperitoneal injection, as well as any similar injection or infusion techniqueThe composition can be also for example a suspension, emulsion, sustained release formulation, cream, gel or powder. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
[00200] In one example, the pharmaceutical formulation is a liquid formulation, e.g., a buffered, isotonic, aqueous solution. In another example, the pharmaceutical composition has a pH that is physiologic, or close to physiologic. In other examples, the aqueous formulation has a physiologic or close to physiologic osmolarity and salinity. It can contain sodium chloride and/or sodium acetate. In some examples, the composition of the present invention is lyophilized.
[00201] Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included herewith for purposes of illustration only and are not intended to be limiting of the invention. All patents and publications referred to herein are expressly incorporated by reference. Examples
Example 1
Cloning, expression and purification of rFVIIIFc
[00202] All molecular biology procedures were performed following standard techniques. The coding sequence of human FVIII (Genbank accession number NM 000132), including its native signal sequence, was obtained by reverse transcription-polymerase chain reactions (RT-PCR) from human liver polyA RNA. Due to the large size of FVIII, the coding sequence was obtained in several sections from separate RT-PCR reactions, and assembled through a series of PCR reactions, restriction digests and ligations into an intermediate cloning vector containing a B domain deleted (BDD) FVIII coding region with a fusion of serine 743 (S743) to glutamine 1638 (Q1638), eliminating 2682 bp from the B domain of full length FVIII. The human IgGl Fc sequence (e.g., GenBank accession number Y14735) was obtained by PCR from a leukocyte cDNA library, and the final expression cassette was made in such a way that the BDD FVIII sequence was fused directly to the N-terminus of the Fc sequence (hinge, CH2 and CH3 domains, beginning at D221 of the IgGl sequence, EU numbering) with no intervening linker. For expression of the Fc chain alone, the mouse IgK (kappa) light chain signal sequence was created with synthetic oligonucleotides and added to the Fc coding sequence using PCR to enable secretion of this protein product. The FVIIIFc and Fc chain coding sequences were cloned into a dual expression vector, pBudCE4.1 (Invitrogen, Carlsbad, CA).
[00203] HEK 293H cells (Invitrogen, Carlsbad, CA) were transfected with the pSYN-FVIII-013 plasmid using Lipofectamine transfection reagent (Invitrogen, Carlsbad, CA)), and a stable cell line was selected with zeocin. Cells were grown in serum free suspension culture, and rFVIIIFc protein purified from clarified harvest media using a four column purification process, including a FVIII-specific affinity purification step (McCue J. et ah, J. Chromatogr. A., 1216(45): 7824-30 (2009)), followed by a combination of anion exchange columns and a hydrophobic interaction column.
Example 2
Biochemical characterization
[00204] Processed recombinant FVIII-Fc (rFVIIIFc) is synthesized as two polypeptide chains, one chain consisting of BDD-FVIII (S743-Q1638 fusion, 1438 amino acids) fused to the Fc domain (hinge, CH2 and CH3 domains) of IgGl (226 amino acids, extending from D221 to G456, EU numbering), for a total chain length of 1664 amino acids, the other chain consisting of the same Fc region alone (226 amino acids). Though cells transfected with the FVIIIFc/Fc dual expression plasmid were expected to secrete three products (FVIIIFc dimer, FVIIIFc monomer, and Fc dimer), only the FVIIIFc monomer and Fc dimer were detected in conditioned media. Purified FVIIIFc was analyzed by non-reducing and reducing SDS-PAGE analysis (Figure 2A and, B). For the nonreduced SDS-PAGE, bands were found migrating at approximately 90 kDa and 130 kDa, consistent with the predicted molecular weights of the FVIIIFc heavy chain (HC) and light chain-dimeric Fc fusion (LCFc2) (Figure 2A, lane 3). A third band was also detected at approximately 220 kDa, consistent with the predicted molecular weight for single chain FVIIIFc (SC FVIIIFc; HC+LCFc2), in which the arginine residue at position 754 (1648 with respect to the full length sequence) is not cleaved during secretion. For the reduced SDS-PAGE analysis, major bands were seen migrating at approximately 25 kDa, 90 kDa, 105 kDa, and 195 kDa, consistent with the predicted molecular weights for the single chain Fc, HC, LCFc, and SC FVIIIFc (Figure 2B, lane 3). Cotransfection with human PC5, a member of the proprotein convertase subtlisin/kexin (PCSK) type proteases, resulted in full processing of the rFVIIIFc product (Figure 2A, B, lane 2).
[00205] Densitometry analysis of several batches of rFVIIIFc after SDS-PAGE indicated greater than 98% purity of the expected bands. Size exclusion chromatography (SEC) was also used to assess the degree of aggregation present, and all batches were found to have aggregate levels at 0.5% or less.
[00206] rFVIIIFc structure was further analyzed by thrombin cleavage, reduction, and analysis by
LC/UV and LC/MS. The four Factor VIII fragments generated by thrombin (by cleavages at three arginine residues, at positions 372, 740 and 795 (795 corresponds to 1689 with respect to the full length FVIII sequence), can be detected by UV absorbance (Figure 2C), corresponding to the following segments of the protein: Fc (peak 1), light-chain-Fc (peak 2); the Al domain from the heavy chain (peak 3) and the A2 domain from the heavy chain (peak 4). The 14 amino acid B domain linker and ~ 6 kDa a3 -related peptides are not detected by UV absorbance due to their small size.
[00207] Analysis of the thrombin digestion of rFVIIIFc by HPLC/MS provided further detailed information of the four main domains as well as the ~6 kDa a3 related peptides, and was compared to REFACTO®, a CHO-derived recombinant BDD-FVIII protein (rBDD FVIII), using the same methods. FVIII samples were passed through Detergent-OUTDTG-100X columns (GBioscience, Maryland Heights, MO) for the removal of Tween, fully digested with thrombin, reduced and analyzed either by RP-HPLC-UV (POROS Rl/10, Applied Biosystems) or RP- HPLC-MS (Agilent 1200 coupled to an Agilent 6210TOF mass spectrometer) using gradients of acetonitrile in water + 0.1 % formic acid . Peptide sequence was also confirmed with LysC peptide mapping, analyzed by RP-HPLC/MS (Thermo Finnigan LTQ-XL-ETD). [00208] As expected, the total ion current (TIC) chromatogram of rFVIIIFc (Figure 2D) appears similar to the UV chromatogram (Figure 2C). Five of the expected six products can be detected by LC/MS, including two forms of the a3 acidic region generated from the processed and single chain isoforms, as well as the thrombin used for the digestion. An additional truncated form of the a3 acidic region was also observed, and is described more fully below. rBDD FVIII yielded a similar TIC chromatogram, but without the free Fc chain and having a different mass for the LC compared to the rFVIIIFc LC-Fc, consistent with the lack of an Fc region (data not shown).
[00209] Due to the heterogeneity of glycosylation over much of the protein, the deconvoluted mass spectra for the Al, LC-Fc, and Fc regions are complex and therefore the identity of all of the molecular ions have not been established. However, the observed mass for the three major peaks from the Fc region were found to match the GO, Gl, and G2 isoforms found on IgG molecules, corresponding to biantennary oligosaccharides terminating in 0, 1 or 2 galactose residues. The deconvoluted mass spectra of the a3 -related peptides and the A2 domain provide the most definitive data, as there are no heterogeneity in the posttranslational modifications in these regions, allowing the expected masses to be identified unambiguously.
[00210] The 6 kDa N-terminal peptide released from the LC after cleavage of R1689 is predicted to comprise the a3 acidic region (amino acids El 649 to R1689) if derived from the processed isoform, and the 14 amino acid truncated B domain fused to the a3 acidic region if derived from the single chain isoform. Both rFVIIIFc and rBDD FVIII were found to contain both forms of the a3 region, proportional to the expected levels based on SDS-PAGE analysis. In addition, both proteins contained a truncated form of the a3 region corresponding to amino acids D1658-R1689, as has been reported for other FVIII products, though this was found in greater abundance in rBDD FVIII than in rFVIIIFc.
[00211] The A2 domain contains three potential tyrosine sulfation sites, but no glycosylation sites that could result in complex heterogeneity, and therefore the exact masses of this region can be calculated. In addition to the primary expected peak in the deconvoluted mass spectrum of rFVIIIFc correlating to the mass of the S373 to R740 sequence (Figure 2E), two additional forms were identified corresponding to known truncations of the FVIII HC, correlating with an A2 domain truncated at E720 and Y729. These reported truncated forms were also observed directly in the deconvoluted spectrum of rBDD FVIII A2 (Figure 2E). Both rFVIIIFc and rBDD FVIII A2 contained similar relative amounts of the form truncated at Y729 while the rBDD FVIII A2 domain contained a notably greater level of the form truncated at E720 as compared to the same form in rFVIIIFc (Figure 2E).
[00212] The primary sequence of rFVIIIFc was confirmed by peptide mapping with lysyl endopeptidase (Lys-C) digests followed by UV and mass spectrometric detection. Of the 99 theoretical peptides produced from rFVIIIFc, 81 were detected, corresponding to 98% of the total sequence. The posttranslational modifications of rFVIIIFc were also characterized by this method. FVIII contains 6 potential tyrosine sulfation sites, corresponding to positions 346, 718, 719, 723, 1664, and 1680. Fully sulfated peptides corresponding to these six sites were found, with trace amounts of non-sulfated peptide corresponding to position 1680 as assessed by integration of the total ion chromatogram in the mass spectra, and no detectable non-sulfated peptides corresponding to the other positions. BDD FVIII also contains 6 potential N- glycosylation sites, four of which have been reported to be glycosylated in recombinant FVIII products. Consistent with this, rFVIIIFc was found to have the same 4 sites glycosylated; N239 and N2118 were found to contain high mannose structures, while N41 and N1810 were found to contain more complex carbohydrates, similar to those found on rBDD FVIII. The Fc region N- linked glycosylation was found to match the GO, Gl , and G2 isoforms found with the thrombin map by LC/MS. FVIII has been reported to have O-glycosylation sites at Ser 741 and 743 that are partially occupied, and this was found to be the case with rFVIIIFc as well.
[00213] The rFVIIIFc polypeptide produced without cotransfected processing enzymes exhibited
15-25% single chain FVIIIFc (SC FVIIIFc), which differs from processed rFVIIIFc by a single peptide bond between R754 and E755 (R1648/E1649 with respect to the full length FVIII). This isoform was purified and characterized in all of the biochemical assays described above, and found to be comparable to rFVIIIFc as shown below. The activity of purified single chain FVIIIFc was found to be similar to rFVIIIFc in a chromogenic assay as well as by the various functional assays described below.
Measurement of FVIII Activity by Chromogenic and One-Stage aPTT Assays
[00214] FVIII activity was measured by a FVIII chromogenic assay. The average specific activity from four separate batches of rFVIIIFc was found to be 9762 ± 449 IU/mg by the chromogenic assay, corresponding to 2148±99 IU/nmol. The average specific activity from fourteen separate batches of rFVIIIFc was found to be 8460 ± 699 IU/mg by the aPTT assay, and 9348 ± 1353 IU/mg by the chromogenic assay, corresponding to 1861±154 and 2057±298 IU/nmol, respectively. FVIII activity of single chain FVIILFc was also measured by the chromogenic assay and compared to the completely processed rFVllLFc or rFVllLFc DS (containing about 25% single chain rFVllLFc). As Table 3A shows, single chain rFVIIIFc showed no significant difference in FVIII activity compared to the Factor VIII activity of completely processed FVIIIFc or rFVIIIFc DS by the chromogenic assay, both in the presence and the absence of von Willebrand Factor (VWF). Table 3B shows that full activity of SCrFVIIIFc, as measured by one- stage activated partial thromboplastin time (aPTT) assay, was observed in the absence of VWF. TABLE _3A_FVIII Activity by Chromogenic Assay
Figure imgf000059_0001
CV=coefficient of variation
TABLE _3B_FVIII Activity by aPTT assay
Figure imgf000059_0002
FVIII/VWF- rFVIIIFc 7742 7.4 depleted
plasma SC rFVIIIFc 3133 4.9 supplemented
with human Completely-processed rFVIIIFc
8495 4.0 VWF
[00215] In one-stage clotting assay (APTT), SC rFVIIIFc demonstrated a 60% decrease in activity when the plasma has normal VWF level, suggesting the potential role of VWF in the activation of SC rFVIIIFc. This observation was further confirmed by addition of human VWF back to the FVIII/VWF-depleted plasma (Table 3), where the coagulant activity of SC rFVIIIFc was reduced to the same level as in congenital FVIII-deficient plasma.
Activity in Xase Complex
[00216] FVIII activity was also measured in the context of the Xase complex, by incubating activated FIX and thrombin-activated REFACTO® or rFVIIIFc protein on a phospholipid surface in the presence of calcium, and monitoring the conversion of FX to FXa as measured by cleavage of a chromogenic or fluorogenic substrate, from which FXa generation rates were determined. This assay was then modified by varying one component of the assay while keeping the others constant in order to examine the interactions with each individual component.
[00217] The FXa generation rate was determined as a function of varying phospholipid concentrations for rFVIIIFc DS, rBDD FVIII, and single chain rFVIIIFc (Figure 3A), using synthetic phospholipid vesicles (25% phosphotadyl serine/75%) phosphotadyl choline). Both proteins were found to have a similar activity profile, with peak activity at approximately 156 μΜ phospholipids.
[00218] The FXa generation rate was then determined as a function of varying FX concentrations, and Km and Vmax values calculated (Figure 3B). The activity profiles for rFVIIIFc DS, rBDD FVIII, and single chain rFVIIIFc were found to be similar, with similar Km and Vmax (Table 4). Finally, the FXa generation rate was determined as a function of varying FIX concentrations (Figure 3C). The activity profiles appeared similar, with similar Kd and Vmax (Table 4). Similar results were obtained using platelets as a phospholipid source (unpublished data, June 2009).
TABLE 4. FXa Generation Parameters for FVIII Proteins on Phospholipids
Figure imgf000060_0001
TABLE 5. FlXa Interactions with FVIII Proteins
Figure imgf000061_0001
Inactivation by APC
[00219] Once active, FVIII is inactivated by cleavage by activated protein C (APC), as well as by dissociation of the A2 domain. rFVIIIFc and rBDD FVIII were both activated by thrombin, then incubated with APC for different times and activity determined in a FXa generation assay (Figure 4). In the absence of thrombin activation, little FXa generation was detected, and this was increased significantly with thrombin digestion. Treatment with APC for 90 min led to a significant decrease in FXa generation rates, similar to non-activated samples, and these results were similar for rFVIIIFc DS, rBDD FVIII, and single chain rFVIIIFc.
Affinity for vWF
[00220] FVIII interactions with von Willebrand factor (vWF) were measured by real-time biomolecular interaction analysis (BIAcore), based on surface Plasmon resonance (SPR) technology, to determine the kinetics of binding of rFVIIIFc and rBDD FVIII towards vWF (Table 6). Kinetic rate parameters of Ka (on-rate) and Kd (off-rate), and the affinity KD (Kd/Ka), were determined for each FVIII interaction under identical conditions. Both rFVIIIFc and rBDD FVIII were found to have a low nM binding affinity (KD) for vWF, of 1.64±0.37 and 0.846±0.181 nM, respectively. The proteins had similar off-rates, with a two fold difference in on-rate resulting in a two fold difference in the affinity.
TABLE 6. Biocore Binding Analysis of FVIII Proteins to vWF
Figure imgf000061_0002
[00221] As shown in Table 6, the affinity of rFVIILFc DS or single chain rFVIIIFc with vWF was found to be in the low nM range, approximately two fold greater than that of BDD FVIII alone. At physiological concentrations, this would result in a slight decrease in the percentage of rFVIIIFc (processed or single chain) complexed with vWF as compared to free FVIII, however in vivo studies have indicated that the half life of rFVIIIFc is significantly prolonged over full length or BDD FVIII despite this slightly lower affinity, and therefore this does not appear to compromise the half life of the molecule. It may be possible that the free rFVIIIFc is more efficiently recycled through the FcRn pathway and therefore this may contribute to a greater prolongation of half life.
Affinity for vWF and Thrombin-mediated Release from vWF
[00222] Recombinant B-domain deleted Factor VIIIFc (rFVIIIFc) was expressed in HEK293 cells. During biosynthesis in HEK293 cells, most of the rFVIIIFc is processed by limited proteolysis to generate a FVIII heavy chain (HC) and a FVIII light chain (LC) to which the Fc moiety is attached. Spontaneous disassociation of the HC and LC in plasma, and during storage of FVIII drug products, is thought to contribute to a loss of FVIII activity. The remaining portion of the biosynthesized rFVIIIFc, which is not processed, forms a single chain isoform of rFVIIIFc (SC rFVIIIFc), which may provide enhanced manufacturability and stability compared to the processed rFVIIIFc.
[00223] This example describes an assay comparing the interaction of SC rFVIIIFc with von
Willebrand factor (vWF) in relation to the interaction of rFVIIIFc with vWF. Interactions with vWF were measured by real-time biomolecular interaction analysis (BIAcore), based on surface Plasmon resonance (SPR) technology, to determine the kinetics of binding of rFVIIIFc and SC rFVIIIFc towards vWF (Table 11 and Figure 16A). FVIII-free human plasma derived vWF was immobilized by amine coupling on the surface of a biosensor at levels low enough to prevent mass transport limitation. rFVIIIFc and SC rFVIIIFc were sequentially injected in single-cycle kinetics mode at concentrations ranging from 0.13 to 5.0 nM. Sensorgram data was fit to a 1 : 1 interaction model.
[00224] Kinetic rate parameters of Ka (on-rate) and Kd (off-rate), and the affinity KD (Kd/Ka), were determined for each FVIII interaction under identical conditions. Both rFVIIIFc and SC rFVIIIFc were found to have a low nM binding affinity (KD) for vWF, of 0.34±0.1 and 0.31±0.1 nM, respectively. Both isoforms also had similar on-rates and off-rates.
TABLE 11. Biocore Binding Analysis of FVIII Proteins to vWF
Figure imgf000062_0001
[00225] Next, thrombin-mediated release of rFVIIIFc, SC rFVIIIFc, and B-domain deleted FVIII lacking Fc moieties (rBDD FVIII) was measured at both 25° C and 37° C. Human vWF was immobilized by amine coupling at similar levels on three flow cells on the biosensor surface. The remaining flow cell served as a blank for reference purposes. The rFVIIIFc and SC rFVIIIFc proteins were captured by vWF and allowed to slowly disassociate, and the concentrations of rFVIIIFc and SC rFVIIIFc were adjusted to obtain equimolar capture levels by the end of the dissociation phase. Human a-thrombin solutions were prepared by 2-fold serial dilution and applied at concentrations that ranged from 0.005 to 20 U/mL, resulting in proteolytic release of FVIIIa species from vWF. Thrombin mediated release of activated rFVIIIFc, SC rFVIIIFc, and rBDD FVIII from vWF was monitored in real-time using an SPR-based optical biosensor (Figure 16B). Following blank reference subtraction (Figure 16C), the release rate as a function of a- thrombin concentration was determined (Figure 16D). The thrombin half maximal effective concentration (EC50) for SC rFVIIIFc was 12±1 U/mL compared to 3.9±0.3 U/mL for rFVIIIFc at 25° C and was 15±1 U/mL for SC rFVIIIFc compared to 4.8±0.2 U/mL for rFVIIIFc at 37° C (Figure 16E). rFVIIIFc had a similar thrombin EC50 value compared to rBDD FVIII, having values of 3.9±0.3 U/mL and 3.3±0.3 U/mL, respectively at 25° C and values of 4.8±0.2 U/mL and 4.0±0.2 U/mL, respectively at 37° C. SC rFVIIIFc had a thrombin EC50 value that was approximately 3-fold higher than rFVIIIFc. This impairment of thrombin mediated release from vWF may underlie the specific reduction in specific activity for SC rFVIIIFc observed in the aPTT assay in which vWF was present (Table 3B).
Example 3
[00226] A Phase I/IIa, open-label, crossover, dose-escalation, multi-center, and first-in-human study was designed to evaluate the safety, tolerability, and pharmacokinetics of a single dose of rFVIIIFc in subjects with severe (defined as <1 IU/dL [1 %] endogenous factor VIII [FVIII]) hemophilia A. A total of approximately 12 previously treated patients were enrolled and dosed with rFVIIIFc at 25 or 65 IU/kg. After the screening (scheduled within 28 days prior to the first dose of the AD V ATE® [rFVIII], the reference comparator agent) and a minimum of 4-days (96 hours) elapsing with no FVIII treatment prior to the first injection, approximately 6 subjects received a single 25 IU/kg dose of AD V ATE® followed by a 3 -day (72 hours) pharmacokinetic (PK) profile then crossover and receive a 25 IU/kg single, open-label dose of rFVIIIFc for a 7-day (168 hours) PK profiling. The first 3 subjects were dosed sequentially. For the first three (3) subjects dosed with 25 IU/kg of rFVIIIFc, each subject underwent an inhibitor assessment at 14- days (336 hours) post-injection of rFVIIIFc. Dosing of the next subject (for the first three subjects only) occurred once the inhibitor testing is completed. After the 3rd subject completed the 14 day inhibitor assessment, the remaining three subjects at 25 IU/kg and the six subjects at 65 IU/kg began enrollment sequentially at least 1 day apart within each dose group.
[00227] One week after the last subject received the 25 IU/kg dose of the rFVIIIFc, approximately
6 unique subjects were recruited for the 65 IU/kg cohort. Each subject in the 65 IU/kg cohort received a single 65 IU/kg dose of ADVATE® followed by a 4-day (96 hours) PK profiling then crossover and receive a 65 IU/kg single, open-label dose of rFVIIIFc for a 10-day (240 hours) profiling. If a bleeding episode occurred before the first injection of rFVIIIFc in any cohort, subject's pre-study FVIII product was used for treatment and an interval of at least 4 days had to pass before receiving the first injection of rFVIIIFc for the PK profile.
[00228] All subjects were followed for a 14-day (336 hours) and 28 day safety evaluation period after administration of rFVIIIFc 25 IU/kg or 65 IU/kg for safety. All subjects underwent pharmacokinetic sampling pre- and post-dosing along with blood samples for analysis of FVIII activity at designated time points.
[00229] The pharmacokinetic data for the Phase I/IIa clinical trial demonstrated the following results for FVIIIFc. FVIIIFc had about a 50% increase in systemic exposure (AUCINF), about 50% reduction in clearance (CI), and about 50-70%) increase in elimination half-life and MRT compared to ADVATE® (full length rFVIII). In addition, FVIIIFc showed increased CI 68, TBLP1, TBLP3, and TBLP5 values compared to ADVATE®.
AUC^F Area under the concentration-time curve from zero to infinity
Beta HL Elimination phase half-life; also referred to as ti^p
CI 68 Estimated FVIIIFc activity above baseline at approximately 168 h after dose
CI Clearance
MRT Mean residence time
TBLP1 Model-predicted time after dose when FVIIIFc activity has declined to
approximately 1 IU/dL above baseline
TBLP3 Model-predicted time after dose when FVIIIFc activity has declined to
approximately 3 IU/dL above baseline
TBLP5 Model-predicted time after dose when FVIIIFc activity has declined to
approximately 5 IU/dL above baseline
Example 4
[00230] A recombinant B-domain-deleted factor VIII-Fc (rFVIIIFc) fusion protein has been created as an approach to extend the half-life of FVIII. The pharmacokinetics (PK) of rFVIIIFc were compared to rFVIII in hemophilia A mice. We found that the terminal half-life was twice as long for rFVIIIFc compared to rFVIII. In order to confirm that the underlying mechanism for the extension of half-life was due to the protection of rFVIIIFc by FcRn, the PK were evaluated in FcRn knockout and human FcRn transgenic mice. A single intravenous dose (125 IU/kg) was administered and the plasma concentration measured using a chromogenic activity assay. The Cmax was similar between rFVIIIFc and rFVIII (XYNTHA®) in both mouse strains. However, while the half-life for rFVIIIFc was comparable to that of rFVIII in the FcRn knockout mice, the half-life for rFVIIIFc was extended to approximately twice longer than that for rFVIII in the hFcRn transgenic mice. These results confirm that FcRn mediates or is responsible for the prolonged half-life of rFVIIIFc compared to rFVIII. Since hemostasis in whole blood measured by rotation thromboelastometry (ROTEM®) has been shown to correlate with the efficacy of coagulation factors in bleeding models of hemophilia mice as well as in clinical applications, we sought to evaluate the ex vivo efficacy of rFVIIIFc in the hemophilia A mice using ROTEM®. Hemophilia A mice were administered a single intravenous dose of 50 IU/kg rFVIIIFc, XYNTHA® (FVIII) or ADVATE® (FVIII). At 5 minutes post dose, clot formation was similar with respect to clotting time (CT), clot formation time (CFT) and a-angle. However, rFVIIIFc showed significantly improved CT at 72 and 96 hr post dose, and CFT and a-angle were also improved at 96 hrs compared to both XYNTHA® (FVIII) and ADVATE® (FVIII), consistent with prolonged PK of rFVIIIFc. Therefore construction of an Fc fusion of FVIII produces a molecule with a defined mechanism of action that has an increased half-life and the potential to provide prolonged protection from bleeding.
Example 5
[00231] This Example presents final analysis results for FVIII activity from 16 patients treated with 25 and 65 IU/kg FVIII products. See Example 3.
[00232] In this Example, rFVIIIFc is a recombinant fusion protein comprised of a single molecule of recombinant B-domain deleted human FVIII (BDD-rFVIII) fused to the dimeric Fc domain of the human IgGl, with no intervening linker sequence. This protein construct is also referred to herein as rFVIIIFc heterodimeric hybrid protein, FVIIIFc monomeric Fc fusion protein, FVIIIFc monomer hybrid, monomeric FVIIIIFc hybrid, and FVIIIFc monomer-dimer. See Example 1, Fig. 1 , and Table 2A.
[00233] Preclinical studies with rFVIIIFc have shown an approximately 2-fold prolongation of the half-life of rFVIII activity compared to commercially available rFVIII products. The rationale for this study was to evaluate the safety and tolerability of a single dose of rFVIIIFc in frozen liquid formulation and provide data on the PK in severe hemophilia A subjects. For this study, 16 evaluable subjects were available for PK evaluation. Single administration of two doses of both rFVIIIFc and ADVATE® at a nominal dose of 25 (n=6) and 65 IU/kg of body weight (n=10) were infused intravenously over approximately 10 minutes. Blood samples for plasma PK assessments were obtained before infusion, as well as up to 10 days after dosing. The PK of FVIII activity for both AD V ATE® and rFVIIIFc were characterized in this study using a model-dependent method.
OBJECTIVES
[00234] The primary objective of this study was to assess the safety and tolerability of single administration of two doses of rFVIIIFc (25 and 65 IU/kg) in previously treated patients (PTPs) aged 12 and above with severe hemophilia A.
[00235] The secondary objectives were to determine the pharmacokinetics (PK) parameters determined by pharmacodynamic (PD) activity of FVIII over time after a single administration of 25 or 65 IU/kg of rFVIIIFc compared to ADVATE® in one-stage clotting and chromogenic assays.
Study Design (See Example 3)
[00236] Blood samples were collected for FVIII activity PK evaluations at the screening visit
(within 28 days prior to dosing ADVATE®); on Day 0 (injection of ADVATE®) pre-injection and at 10 and 30 minutes and 1, 3, 6, and 9 hours post-injection; on Day 1 at 24 hours post-injection of ADVATE®; on Day 2 at 48 hours post-injection of ADVATE®; on Day 3 at 72 hours post- injection of ADVATE®; and on Day 4 at 96 hours post-injection of high dose of ADVATE® (Cohort B only).
[00237] Blood samples were collected for FVIII activity PK evaluations on the day of rFVIIIFc injection just prior to the administration of rFVIIIFc, at 10 and 30 minutes and 1, 3, 6, and 9 hours post-injection of rFVIIIFc; on Day 1 at 24 hours post-injection of rFVIIIFc; on Days 2 through 5 at 48, 72, 96, and 120 hours post-injection of rFVIIIFc; on Day 7 at 168 hours post-injection of rFVIIIFc; on Days 8, 9, and 10 at 192, 216, and 240 hours post-injection of high dose of rFVIIIFc (Cohort B only). FVIII activity was also measured at the final study visit (28 days post-injection of rFVIIIFc) at 672 hours post-injection of rFVIIIFc.
Pharmacokinetic Modeling and Calculations
Abbreviations
TBLP1 = Model-predicted time after dose when FVIII activity has declined to approximately 1 IU/dL above baseline.
TBLP3 = Model-predicted time after dose when FVIII activity has declined to approximately 3 IU/dL above baseline
KV_M = Cmax_M/Actual Dose (IU/kg)
KV OB = Cmax OB/Actual Dose (IU/kg) rVR M = 100 x Cmax M x Plasma Volume (dL) / Total Dose in IU; where plasma volume in mL = (23.7 x Ht in cm) + (9.0 x Wt in kg) - 1709.
rVR OB = 100 x Cmax OB x Plasma Volume (dL) / Total Dose in IU; where plasma volume in mL = (23.7 x Ht in cm) + (9.0 x Wt in kg) - 1709.
RESULTS
Single-Dose Pharmacokinetics (One-Stage Assay)
[00238] Observed FVIII activity increased sharply after the short IV infusion of either ADVATE® or rFVIIIFc, with mean (±SD) model-predicted Cmax values of 56.6 ± 4.74 and 121 ± 28.2 IU/dL for ADVATE® and 55.6 ± 8.18 and 108 ± 16.9 IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. All ADVATE®- and rFVIIIFc -treated patients had dose-related increases in FVIII activity. The observed increase in both Cmax and AUC was slightly less than proportional to dose over the dose range evaluated.
[00239] After the end of the infusion, the decline of the observed FVIII activity exhibited monoexponential decay characteristics until the baseline level was reached. The rate of decline in FVIII activity was slower for rFVIIIFc than for ADVATE® with mean (±SD) model-predicted elimination half-life values of 11.9 ± 2.98 and 10.4 ± 3.03 hr for ADVATE® and 18.0 ± 3.88 and 18.4 ± 6.99 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Elimination half- life values appeared to be dose-independent over the dose range evaluated for both FVIII products.
[00240] Total systemic FVIII exposure (assessed by AUC ) was ~ 48% and 61% greater following rFVIIIFc administration than ADVATE® at 25 and 65 IU/kg dose levels, respectively. Mean (±SD) model-predicted AUC values were 974 ± 259 and 1810 ± 606 hr*IU/dL for ADVATE® and 1440 ± 316 and 2910 ± 1320 hr*IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
[00241] Similar to elimination half-life, the MRT was prolonged for rFVIIIFc relative to
ADVATE®. Mean (±SD) model-predicted MRT values were 17.1 ± 4.29 and 14.9 ± 4.38 hr for ADVATE® and 25.9 ± 5.60 and 26.5 ± 10.1 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. MRT values appeared to be dose-independent over the dose range evaluated for both FVIII products.
[00242] In addition, primary PK parameter values for CL and V were determined. CL values for rFVIIIFc only accounted for ~ 66% of those observed for ADVATE® at equivalent doses. Mean (±SD) model-predicted CL values were 2.70 ± 0.729 and 4.08 ± 1.69 mL/hr/kg for ADVATE® and 1.80 ± 0.409 and 2.69 ± 1.25 mL/hr/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. V values were comparable between ADVATE® and rFVIIIFc with mean (±SD) model-predicted V values of 43.9 ± 4.27 and 56.1 ± 13.4 mL/kg for ADVATE® and 45.3 ± 7.23 and 61.6 ± 10.6 mL/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Slight increases in mean CL and V values were noted with increasing dose of ADVATE® and rFVIIIFc; however, the increase in standard deviations at the 65 IU/kg dose coupled with limited dose levels confounded an assessment of the dose-dependency of these parameters. For example, the CV% geometric mean CL value for the rFVIIIFc treatment group increased from 23.0% (25 IU/kg) to 48.6% (65 IU/kg).
[00243] In addition to the primary PK parameters, secondary PK parameters (e.g. K-values, IVR, etc.) were determined to evaluate FVIII duration of effect. Evidence of PK difference was also observed with rFVIIIFc demonstrating increased TBLPland TBLP3 values compared to ADVATE® at equivalent doses. IVR and K-values for ADVATE® and rFVIIIFc appeared to be comparable. A slight increase in TBLP1 and TBLP3 values were observed with increasing dose of ADVATE® and rFVIIIFc. In contrast, slight decreases in mean IVR and K-values were noted with increasing dose of ADVATE® and rFVIIIFc. As previously indicated, an assessment of the dose dependency of these parameters is confounded by limited dose levels.
[00244] Mean (±SD) observed TBLP1 were 2.88 ± 0.733 and 2.93 ± 0.848 IU/dL per IU/kg for
ADVATE® and 4.28 ± 0.873 and 5.16 ± 2.02 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Mean (±SD) observed TBLP3 were 2.06 ± 0.527 and 2.26 ± 0.666 IU/dL per IU/kg for ADVATE® and 3.09 ± 0.623 and 3.93 ± 1.59 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
[00245] Mean IVR and K-values calculated using observed Cmax values (subtracted with baseline and residual drug within the model) were generally greater than values determined using model- predicted Cmax values; consistent with slight underestimation of the observed peak activity using the one-compartment model. Mean (±SD) observed K-values were 2.57 ± 0.198 and 2.13 ± 0.598 IU/dL per IU/kg for ADVATE® and 2.46 ± 0.330 and 1.85 ± 0.332 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Mean (±SD) observed IVR values were 94.1 ± 15.6 and 85.8 ± 16.5 % for ADVATE® and 89.5 ± 11.9 and 74.8 ± 6.72 % for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
Single-Dose Pharmacokinetics (Chromogenic Assay)
[00246] Observed FVIII activity increased sharply after the short IV infusion of either ADVATE® or rFVIIIFc, with mean (±SD) model-predicted Cmax values of 70.2 ± 9.60 and 157 ± 38.6 IU/dL for ADVATE® and 70.3 ± 10.0 and 158 ± 34.7 IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
[00247] All ADVATE®- and rFVIIIFc-treated patients had dose-related increases in FVIII activity. The observed increase in both Cmax and AUCINF was slightly less than proportional to dose over the dose range evaluated. [00248] After the end of the infusion, the decline of the observed FVIII activity exhibited monoexponential decay characteristics until the baseline level was reached. The rate of decline in FVIII activity was slower for rFVIIIFc than for ADVATE® with mean (±SD) model-predicted elimination half-life values of 10.7 ± 1.98 and 10.3 ± 3.27 hr for ADVATE® and 16.2 ± 2.92 and 19.0 ± 7.94 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Elimination half- life values appeared to be dose-independent over the dose range evaluated for both FVIII products.
[00249] Total systemic FVIII exposure (assessed by AUC ) was ~ 53% and 84% greater following rFVIIIFc administration than ADVATE® at 25 and 65 IU/kg dose levels, respectively. Mean (±SD) model-predicted AUC values were 1080 ± 236 and 2320 ± 784 hr*IU/dL for ADVATE® and 1650 ± 408 and 4280 ± 1860 hr*IU/dL for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
[00250] Similar to elimination half-life, the MRT was prolonged for rFVIIIFc relative to
ADVATE®. Mean (±SD) model-predicted MRT values were 15.3 ± 2.86 and 14.8 ± 4.72 hr for ADVATE® and 23.4 ± 4.22 and 27.3 ± 11.4 hr for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. MRT values appeared to be dose-independent over the dose range evaluated for both FVIII products.
[00251] In addition, primary PK parameter values for CL and V were determined. CL values for rFVIIIFc only accounted for ~ 58-66%) of those observed for ADVATE® at equivalent doses. Mean (±SD) model-predicted CL values were 2.39 ± 0.527 and 3.21 ± 1.40 mL/hr/kg for ADVATE® and 1.57 ± 0.349 and 1.86 ± 0.970 mL/hr/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. V values were comparable between ADVATE® and rFVIIIFc with mean (±SD) model-predicted V values of 35.8 ± 5.52 and 43.6 ± 11.2 mL/kg for ADVATE® and 35.9 ± 6.65 and 42.7 ± 8.91 mL/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Increases in mean CL and V values were noted with increasing dose of ADVATE® and rFVIIIFc; however, the increase in standard deviations at 65 IU/kg coupled with limited dose levels confounded an assessment of the dose-dependency of these parameters.
[00252] In addition to the primary PK parameters, secondary PK parameters (e.g. K-values, IVR, etc.) were determined to evaluate FVIII duration of effect. Evidence of PK difference was also observed with rFVIIIFc demonstrating increased TBLPland TBLP3 values compared to ADVATE® at equivalent doses. IVR and K-values for ADVATE® and rFVIIIFc appeared to be comparable.
[00253] A slight increase in TBLP1 and TBLP3 values were observed with increasing dose of
ADVATE® and rFVIIIFc. In contrast, slight decreases in mean IVR and K-values were noted with increasing dose of ADVATE® and rFVIIIFc. As previously indicated, an assessment of the dose dependency of these parameters is confounded by limited dose levels. [00254] Mean (±SD) observed TBLP1 were 2.70 ± 0.511 and 3.09 ± 0.978 IU/dL per IU/kg for
ADVATE® and 4.06 ± 0.798 and 5.66 ± 2.38 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Mean (±SD) observed TBLP3 were 1.98 ± 0.377 and 2.39 ± 0.718 IU/dL per IU/kg for ADVATE® and 3.04 ± 0.598 and 4.44 ± 1.84 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
[00255] Mean IVR and K- values calculated using observed Cmax values (subtracted with baseline and residual drug within the model) were generally greater than values determined using model- predicted Cmax values; consistent with slight underestimation of the observed peak activity using the one -compartment model. Mean (±SD) observed K-values were 3.08 ± 0.429 and 2.85 ± 0.721 IU/dL per IU/kg for ADVATE® and 3.12 ± 0.451 and 2.92 ± 0.985 IU/dL per IU/kg for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively. Mean (±SD) observed IVR values were 112 ± 14.5 and 116 ± 26.9 % for ADVATE® and 113 ± 16.3 and 117 ± 33.6 % for rFVIIIFc for the 25 and 65 IU/kg dose groups, respectively.
CONCLUSIONS
[00256] All ADVATE®- and rFVIIIFc -treated patients had comparable dose-related increases in
Cmax and AUCINF over the dose range evaluated. Peak plasma levels of ADVATE® and rFVIIIFc activity were generally observed within the first hour after the end of the infusion and remained detectable for several days after dosing. After the end of infusion, the decline in baseline corrected FVIII activity exhibited monoexponential decay until the baseline was reached for both products. Parameter values for elimination half-life and MRT appeared to be dose- independent over the dose range evaluated for both FVIII products. Slight increases in mean CL and V values were noted with increasing dose of ADVATE® and rFVIIIFc; however, increased intersubject variability at the 65 IU/kg coupled with limited dose levels confounded an assessment of the dose-dependency of these parameters.
[00257] Comparison of rFVIIIFc and ADVATE® activity PK revealed an approximate 48-61%
(One-Stage Assay) or 53-84% (Chromogenic Assay) increase in systemic exposure, approximate 30-40%) reduction in clearance, and an approximate 50-80%) increase in both elimination half-life and MRT for rFVIIIFc relative to ADVATE® at comparable doses. Evidence of PK difference was also observed with rFVIIIFc demonstrating increased TBLP1 and TBLP3 values compared to ADVATE® at equivalent doses. IVR and K-values for ADVATE® and rFVIIIFc appeared to be comparable.
[00258] The PK parameters obtained from the Chromogenic Assay results generally agreed with those from the One-Stage Assay, except that the Chomogenic Assay yielded a higher estimation of exposure parameters (e.g., Cmax, AUCINF, etc.). [00259] With the observed improvements in PK, rFVIIIFc may provide a prolonged protection from bleeding, allowing less frequent injections for individuals with Hemophilia A.
Example 6
[00260] On the basis of the interim PK analysis from the first in-human study of rFVIILFc
(Example 3), the A-LONG study was designed. A-LONG is an open label, multi-center evaluation of the safety, pharmacokinetics, and efficacy of recombinant Factor VIII Fc fusion (FVIILFc) in the prevention and treatment of bleeding in previously treated subjects with severe hemophilia A (defined as <1 IU/dL [<1%] endogenous FVIII).
[00261] Approximately 106 subjects will be enrolled into one of three regimens: a tailored prophylaxis regimen (arm 1), a weekly dosing regimen (arm 2), and an on-demand regimen (arm 3).
Arm 1 : Tailored Prophylaxis Regimen
[00262] Arm 1 will include an overall group and a PK subgroup. Approximately 66 subjects will be enrolled. The initial regimen will be twice weekly at 25 IU/kg on the first day, followed by 50 IU/kg on the fourth day of the week. Subjects will administer rFVIIIFc on this weekly prophylaxis regimen until PK results for rFVIIIFc are available. Based on these results, a tailored prophylaxis regimen will be established for each individual, in which the dose and interval will be determined to maintain a trough level of 1-3% FVIII activity. Each subject will then administer his individually tailored prophylaxis regimen throughout the study.
[00263] Subjects will be monitored throughout the study and ongoing dose and interval adjustments will be made. Adjustments will only be made when a subject experiences unacceptable bleeding episodes defined as >2 spontaneous bleeding episodes over a rolling two- month period. In this case, adjustment will target trough levels of 3-5%.
Arm 2: Weekly Dosing Regimen
[00264] Approximately 20 subjects will be enrolled/randomized and undergo abbreviated rFVIIIFc PK profiling as follows: Washout of at least 96 hours; a single dose of rFVIIIFc 65 IU/kg; Abbreviated sampling beginning on rFVIIIFc Day 0, including pre-injection and 10 (±2) minutes, 3 hours (± 15 minutes), 72 (± 2) hours [Day 3], and 96 (± 2) hours [Day 4] from the start of injection. Following the abbreviated PK profiling, subjects will then administer a fixed dose of 65 IU/kg rFVIIIFc every 7 days at least for 28 weeks and up to 52 weeks. Arm 3: On-demand Regimen
[00265] A minimum of 10 major surgeries in at least 5 subjects will be evaluated in the study.
Major surgery is defined as any surgical procedure (elective or emergent) that involves general anesthesia and/or respiratory assistance in which a major body cavity is penetrated and exposed, or for which a substantial impairment of physical or physiological functions is produced (e.g., laparotomy, thoracotomy, craniotomy, joint replacement, and limb amputation).
[00266] For prophylaxis during surgery, subjects will be treated with 20 to 50 IU/kg rFVIIIFc every 12 to 24 hours. Prior to surgery, the physician will review the subject's rFVIIIFc PK profile and assess the dose regimen of Factor VIII replacement generally required for the type of planned surgery and the clinical status of the subject. Recommendation for the appropriate dosing of rFVIIIFc in the surgical treatment period, including any rehabilitation time, will take these factors into consideration.
[00267] The primary objectives of this study are (a) to evaluate the safety and tolerability of rFVIIIFc administered as prophylaxis, on-demand, and surgical treatment regimens; and (b) to evaluate the efficacy of rFVIIIFc administered as prophylaxis, on-demand, and surgical treatment regimens. The secondary objectives of this study are (a) to characterize the PK profile of rFVIIIFc and compare the PK of FVIIIFc with the currently marketed product, AD V ATE®; (b) to evaluate individual responses with FVIIIFc; and (c) to evaluate FVIIIFc consumption.
Primary Objectives
• To evaluate safety and tolerability of rFVIIIFc administered as prophylaxis, weekly, on- demand, and surgical treatment regimens
• To evaluate the efficacy of rFVIIIFc administered as tailored prophylaxis, on-demand, and surgical treatment regimens
Secondary Objectives
• To characterize the PK profile of rFVIIIFc and compare the PK of rFVIIIFc with the currently marketed product, AD V ATE®
• To evaluate individual responses with rFVIIIFc
• To characterize the range of dose and schedules required to adequately prevent bleeding in a prophylaxis regimen; maintain homeostasis in a surgical setting; or to treat bleeding episodes in an on-demand, weekly treatment, or prophylaxis setting
• To evaluate rFVIIIFc consumption (e.g., total annualized rFVIIIFc consumption per subject) Example 7
Clinical ROTEM® Assessment
[00268] In the study in Example 7, in addition to the measurement of plasma FVIII activity by one-stage activated partial thromboplastin time (aPTT) assay, whole blood rotational thromboelastometry (ROTEM®) has also been explored to assess the improvement in global hemostasis by rFVIIIFc and AD V ATE® in 2 subjects, specifically, 1 in the low dose cohort and 1 in the high dose cohort.
[00269] rFVIIIFc and AD V ATE® appear to be comparably active in clot formation when spiked into subjects' blood prior to rFVIIIFc treatment. The clotting time (CT) was linear with respect to the dose of rFVIIIFc and AD V ATE® in the range of approximately 1% of 100% of normal, and the dose response was comparable between rFVIIIFc and ADVATE® in the same subject.
[00270] Following dosing with AD V ATE® and subsequently rFVIIIFc, citrated whole blood was sampled at various time points and the clot formation following recalciiication was monitored by ROTEM®. Despite the variable baseline CT due to residue FVIII levels prior to ADVATE® or rFVIIIFc dosing, both products effectively corrected the CT to comparable levels 30 minutes post-injection. In addition, the improvement in CT was better sustained at and after 3 hours post- injection of 25 IU/kg of rFVIIIFc relative to ADVATE® in the subject dosed at this low dose. However, the differential improvement of rFVIIIFc versus ADVATE® was much less appreciable at the 65 IU/kg dose.
Example 8
[00271] HemA mice were used for tail clip studies. The mice were first anesthetized and then injected with 4.6 μg/kg, 1.38 μg/kg, or 0.46 μg/kg of either processed rFVIIIFc (Drug Substance, which contain about 75%-85% processed rFVIIIFc) and purified single chain rFVIIIFc. After the injection, the tail was cut from the tip and immediately placed into a tube to collect blood. Percentage of protection on survival was measured for rFVIIIFc processed (drug substance) and single chain FVIIIFc as shown in Table 7 and Figure 7.
TABLE 7. In Vivo Efficacy of rFVIILFc DS and Single chain rFVIIIFc
Figure imgf000073_0001
[00272] As shown in Table 7 and Figure 7, the protection on survival by single chain rFVIIIFc is comparable to processed rFVIIIFc (DS).
Clotting activity by in vitro ROTEM
[00273] The clotting potency of rFVIIIFc was further explored in whole blood Rotational
Thromboelastometry (ROTEM) over a range of concentrations, and compared to both rBDD FVIII (Xyntha) and recombinant full length FVIII (rflFVIII; Advate). For in vitro ROTEM, rFVIII proteins were spiked in triplicate into citrated pooled blood collected from the vena cava of 5-6 male HemA mice to the final concentration of 0, 0.1, 1, 10, and 100% of normal plasma FVIII level. The clot was initiated by the addition of CaCl2 (NATEM) and clotting time (CT), clot formation time (CFT), alpha-angle and maximum clot firmness (MCF) were recorded on the ROTEM system (Pentapharm GmbH, Munich, Germany). The clotting time (CT), clot formation time (CFT), and alpha angle for the three proteins spiked in HemA mouse blood at escalating doses from 0.1 - 100 % of normal FVIII levels are shown in Figure 14. In the wide range of 0.1 to 100% of normal, the CT and CFT are comparable among rFVIIIFc, rBDD FVIII and rflFVIII. The alpha angle is only significantly different (p<0.05) between rFVIIIFc and rBDD FVIII at 10%.
Clotting activity by ex vivo ROTEM
[00274] The pharmacodynamics of rFVIIIFc, as measured by ROTEM, was compared to rBDD
FVIII and rflFVIII after a single intravenous injection into Hemophilia A mice. For ex vivo ROTEM, male HemA mice were injected intravenously with a single dose of 50 IU/kg rFVIIIFc, ADVATE®, or XYNTHA®, and 5 mice sacrificed at each time point (5 minutes, 24, 48, 72 and 96 hours post dosing). Individual citrated whole blood collected from the vena cava was immediately analyzed by NATEM on the ROTEM system and parameters measured as above. The CT, CFT, and alpha angle were determined for samples taken from 5 min to 96 hours after dosing, and shown in Figure 15. At 5 min, all are comparably effective resulting in similar CT, CFT and alpha angle (Figure 15A - C). However, rFVIIIFc demonstrated a significantly improved (p<0.05) CT at 72 and 96 hrs, CFT and alpha angle at 96 hrs (Figures 15A - C) relative to rBDD FVIII and rflFVIII .
Example 9
[00275] Recombinant factor VIIIFc (rFVIIIFc) is comprised of a B domain deleted (BDD) rFVIII protein genetically fused to the Fc domain of human immunoglobulin Gl (IgGl). Prior to secretion from HEK 293 cells, most of the rFVIIIFc is processed into a FVIII heavy chain (HC) and light chain (LC+Fc). In circulation, rFVIIIFc is complexed with von Willebrand factor (VWF) and released upon activation in a manner that is indistinguishable from native FVIII. Spontaneous dissociation of the HC and LC is thought to contribute to the loss of FVIII activity in plasma and during storage of FVIII drug products. Here we describe a single chain non-processed isoform of rFVIIIFc (SC rFVIIIFc), which may provide superior manufacturability and enhanced stability compared to native FVIII.
[00276] SC rFVIIIFc was purified from rFVIIIFc, which contains a fraction of the non-processed isoform. Compared to rFVIIIFc, SC rFVIIIFc showed equivalent chromogenic activity but approximately 60% reduced activity by the one stage (aPTT) assay, (Table 3A-B). Thrombin generation assay (TGA) was performed using calibrated automated thrombogram (from Thrombinoscope®). In a thrombin generation assay (TGA), SC rFVIIIFc also showed a reduced thrombin potential (Figure 13 A), and peak thrombin (Figure 13B) compared to rFVIIIFc. However, as shown in Table 3B, full activity of SC rFVIIIFc by aPTT was observed in the absence of vWF, suggesting release from vWF may be delayed due to covalent linkage of the a3 acidic region to the HC after Arg 1680 cleavage in SC rFVIIIFc, in contrast to a3 release and dissociation from fully processed FVIII. Delayed dissociation from vWF may explain the reduced activity observed in the aPTT assay and TGA, while full activity was observed in the two-stage chromogenic assay. A reduced rate of activation in the presence of vWF was confirmed in a modified chromogenic substrate assay with limiting thrombin as FVIII activator.
[00277] In vivo function of SC rFVIIIFc was assessed in the HemA mouse tail vein transection
(TVT) model. HemA male mice were treated with indicated doses of either rFVIIIFc drug product or SC rFVIIIFc 48 hours prior to TVT. Tail re -bleeding and survival were monitored hourly up to 12 hours post TVT with final observation performed at 24-hour post TVT. SC rFVIIIFc and the rFVIIIFc demonstrated equivalent in vivo efficacy in this model, with an ED50 of 1.17 μg/kg and 1.23 μg /kg respectively when TVT was performed at 48 hours post infusion (Figure 7(A)). Comparable 24 hour post TVT survival curves (p > 0.65) (Figure 7(B)) and re-bleed rates (Figure 7(C)) in HemA mice were observed for the SC rFVIIIFc and rFVIIIFc at each tested dose level, indicating that SC rFVIIIFc was equally effective as rFVIIIFc despite its lower apparent aPTT activity. The delayed in vitro activation of SC rFVIIIFc in the presence of vWF therefore appears to have no significant impact on its in vivo efficacy. Thus, SC rFVIIIFc represents a novel and efficacious isoform of rFVIIIFc with potential clinical applications. Further studies will be required to demonstrate enhanced product stability in the context of this Fc fusion protein.
Example 10
[00278] Current factor VIII (FVIII) products display a half- life (t1/2) of approximately 8-12 hours, requiring frequent intravenous injections for prophylaxis and treatment of hemophilia A patients. rFVIIIFc is a recombinant fusion protein composed of a single molecule of FVIII covalently linked to the Fc domain of human IgGi to extend circulating rFVIII half-life. This first-in-human study in previously-treated male subjects with severe hemophilia A investigated safety and pharmacokinetics of rFVIIIFc. Sixteen subjects received a single dose of ADVATE® at 25 or 65 IU/kg followed by an equal dose of rFVIIIFc. Most adverse events were unrelated to study drug. None of the study subjects developed anti-FVIIIFc antibodies or inhibitors. Across dose levels, as compared with ADVATE®, rFVIIIFc showed 1.54 to 1.71-fold longer elimination t and mean residence time, 1.49 to 1.56-fold lower clearance, and 1.48 to 1.56-fold higher total systemic exposure. ADVATE® and rFVIIIFc had comparable dose-dependent peak plasma concentrations and recoveries. Time to 1% FVIII activity above baseline was approximately 1.53 to 1.68-fold longer than ADVATE® across dose levels. Thus, rFVIIIFc may offer a viable therapeutic approach to achieve prolonged hemostatic protection and less frequent dosing in patients with hemophilia A.
[00279] Hemophilia A is an inherited bleeding disorder that results in frequent spontaneous and traumatic bleeding into the joints and soft tissues. Mannucci PM, Tuddenham EGD, N Engl J Med, 344:1773-1779 (2001). When inadequately treated, this bleeding leads to chronic arthropathy, disability, and increased risk of death. Soucie JM et al., Blood. 96(2):437-442 (2000).
[00280] Plasma-derived FVIII (pdFVIII) and recombinant human FVIII (rFVIII) products are utilized for treatment (on-demand therapy) and prevention (prophylaxis therapy) of bleeding episodes. rFVIII was developed to reduce the risk of blood-borne pathogen transmission following the widespread contamination of plasma products with HIV and hepatitis viruses, and to secure an adequate supply of FVIII product. However, hemostatic protection with current FVIII products is temporally limited due to a short half-life (t1/2) of approximately 8-12 hours, requiring prophylactic injections three times per week or every other day for most patients in order to maintain FVIII levels above 1%, a level that has been established as protective against most spontaneous bleeding episodes. Manco-Johnson et al., New Engl J Med. 357(6):535-44 (2007).
[00281] Many studies have shown that, even at high doses, on-demand therapy is not effective in preventing arthropathy. Aledort L. et al., J Intern Med. 25(5:391-399 (1994); Petrini P. et al., Am J Pediatr Hematol Oncol.15:280-287 (1991). The benefits of prophylactic therapy have been demonstrated in numerous clinical studies 4' 6 15 and Manco-Johnson et al., supra, established that children started on primary prophylaxis after their first joint bleed had significantly fewer bleeds and less joint damage than children treated on-demand.
[00282] Compared to on-demand treatment, prophylactic therapy also decreases disability, hospitalization rate, and time lost from school or work;6'16 and improves quality of life for patients and their families.17 However, prophylactic therapy often requires use of central venous access devices in children, and their attendant risks of infection, sepsis, and thrombosis. In addition, despite the benefits, acceptance of and compliance with prophylaxis decreases with age, in part because of inconvenience and invasiveness. ' Thus, a rFVIII product with a prolonged plasma ti 2 would potentially be of benefit. Lillicrap D., Current Opinion in Hematology 77:393-397 (2010).
[00283] rFVIIIFc is a recombinant fusion protein composed of a single molecule of B-domain deleted rFVIII covalently linked to the human IgGi Fc domain. Potential advantages of Fc-fusion proteins include better tolerability and prolonged hemostatic protection, and the Fc domain represents a natural molecule with no known inherent toxicity.21'22 Attachment to the IgGi Fc domain permits binding to the neonatal Fc receptor (FcRn), which is expressed in many cell types, including endothelial cells. FcRn expression remains stable throughout life and is responsible for protecting IgGi and Fc-fusion proteins from lysosomal degradation, thus prolonging the t1/2 of the protein.21'23 Numerous proteins within the circulation are internalized into the cells lining the vasculature via nonspecific pinocytosis and are trafficked to endosomal and lysosomal degradation pathways.
[00284] Fc proteins interact with FcRn, resident within endosomes. Endosomes containing FcRn direct the Fc fusion proteins back to the plasma membrane, releasing them into circulation in a pH-dependent manner,24 thereby avoiding lysosomal degradation. This recycling approach has been used successfully to extend the t1/2 of therapeutic biologies; a number of Fc fusion-based drugs have been approved for clinical use (eg etanercept, romiplostim) and others are in development.25'26
[00285] Preclinical data for rFVIIIFc indicate that FVIII can be rescued from degradation by a natural protective pathway mediated by FcRn, thus extending t1/2. In Hemophilia A mice and dogs, terminal plasma t1/2 for rFVIIIFc was approximately 2 times longer than with rFVIII.27'28 Based on these data, we conducted a first-in-human clinical study to investigate the safety and PK of a long-lasting rFVIIIFc fusion protein in subjects with hemophilia A.
[00286] Study Design: This open-label, dose-escalation, multicenter Phase l/2a study in previously treated patients with severe hemophilia A investigated the safety of rFVIIIFc and its pharmacokinetics (PK) compared with AD V ATE® (antihemophilic factor [recombinant], plasma/albumin-free method, octocog alfa, Baxter Healthcare). This study was performed in accordance with the US CFR and ICH Guidelines on Good Clinical Practices. Prior to any testing, approval from participating Institutional Review Boards and written informed consents from all subjects were obtained. The study design was sequential; a single dose of ADVATE® was administered at 25 or 65 IU/kg followed by an equal dose of rFVIIIFc (FIG. 8). Both drugs were injected intravenously over approximately 10 minutes. The two dose levels were expected to bracket the typical therapeutic dose ranges. Subjects were followed for 28 days after receiving rFVIIIFc for safety analyses, including testing for anti-FVIII antibodies and inhibitors at 14 and 28 days post-injection. Plasma FVIII activity was measured in subjects before injection, 10 and 30 minutes, 1, 3, 6, 9, 24, 48, 72, 96, 120, and 168 hours (7 days) after rFVIIIFc injection, with additional samples at 192, 216, and 240 hours (10 days) for subjects dosed at 65 IU/kg of rFVIIIFc. Plasma FVIII activity was measured at the same time points after ADVATE® treatment, through 72 hours for the 25 IU/kg group and 96 hours for the 65 IU/kg group.
[00287] Subjects: Male subjects were at least 12 years of age with severe hemophilia A (defined as FVIII activity level < 1%) and had at least 100 documented prior exposure days to FVIII concentrates (pdFVIII or rFVIII). Subjects with known hypersensitivity to mouse or hamster protein, history of inhibitor or detectable inhibitor titer at screening, or who were taking any medications that could affect hemostasis or systemic immunosuppressive drugs, or who experienced an active bacterial or viral infection (other than hepatitis or HIV) within 30 days of screening were excluded. Subject's genotype was recorded at study entry, when known.
[00288] Treatment Product: The human rFVIIIFc and Fc transgenes were stably transfected into
HEK293 cells and the cell line was extensively tested for stability, sterility, and viral contamination to ensure safety. The purified drug product is composed of a monomeric B- domain-deleted FVIII covalently linked through its carboxy-terminus to the N-terminus of an Fc monomer, which forms a disulfide bond with a second Fc monomer during synthesis and secretion from the cells. rFVIIIFc was purified by chromatography and nanofiltration, and was fully active in one-stage and chromogenic clotting assays relative to commercially available rFVIII preparations. It was supplied as a frozen liquid containing 1000 IU per 2 niL of solution and formulated with L-histidine (pH 7), sodium chloride, calcium chloride, sucrose, mannitol, and Polysorbate 20. For injection, the product was diluted with saline solution (0.9% NaCl).
[00289] Outcome Measures: The primary objective of the study was safety, evaluated through physical examination, reporting of treatment-emergent adverse events (AEs), development of antibodies, and laboratory monitoring over time. The secondary objectives included parameters derived from PK analyses. Laboratory assessments included prothrombin time, activated partial thromboplastin time (aPTT), international normalized ratio, levels of D-dimer, von Willebrand factor (vWF) antigen, standard hematology and blood chemistry tests, and urinalysis.
[00290] FVIII activity was measured by the one-stage clotting (aPTT) assay on a Siemens
BCS-XP analyzer using commercial reagents (Dade Actin FSL) with calibration against a normal reference plasma (Precision Biologies CRYOcheck™) traceable to the World Health Organization (WHO) 5th International Standard (IS) for human plasma. In addition to the aPTT assay, FVIII activity was measured by a chromogenic substrate assay29 using a commercially available kit (Aniara BIOPHEN FVIILC) that complies with European Pharmacopoeia recommendations. The chromogenic assay was calibrated against normal human reference plasma (Instrumentation Laboratories ORKE45), which also had a potency assigned against the WHO 5th IS human plasma standard. [00291] The lower limit of quantification (LLOQ) for the one-stage and chromogenic assays was
0.5 IU/dL and 0.4 IU/dL, respectively. FVIII inhibitors were measured by the - *Nijmegen-modified Bethesda assay and less than 0.6 BU/mL was considered negative. Anti- rFVIIIFc antibodies were assessed using a specific bridging electrochemiluminescent immunoassay which uses biotin and sulfo-tagged rFVIIIFc. Assay sensitivity was determined to be 89 ng/mL using an anti-human FVIII monoclonal antibody as a surrogate control. Exploratory whole blood rotation thromboelastometry (ROTEM®) was performed in two subjects, one from each dose level, at various time points to assess the improvement in global hemostasis following injection with ADVATE® and rFVIIIFc.
[00292] Pharmacokinetic Analyses: A user-defined one -compartment disposition model, which automatically estimates the endogenous FVIII level and subsequent residual decay, was utilized in WinNonLin for analysis of the individual subject plasma FVIII activity- versus-time data following a single administration of ADVATE® or rFVIIIFc. Actual sampling times, doses, and duration of injection were used for calculations of parameters including maximum activity (Cmax), ti 2, clearance (CL), volume of distribution at steady-state (Vss), area under the curve (time zero extrapolated to infinity [AUC ]), mean residence time (MRT), and incremental recovery.
[00293] Monte Carlo Simulation of rFVIIIFc Activity-Versus-Time Profile - To construct FVIII activity-time profiles following dosing regimens of 25 IU/kg or 65 IU/kg, a Monte Carlo simulation was conducted using the population PK model of ADVATE® and rFVIIIFc. The mean estimates of model parameters (CL, volume of distribution) in the tested population, the inter- individual variance, and the residual variability were estimated based on the one-stage (aPTT) clotting assay activity data of ADVATE® and rFVIIIFc from 16 subjects in this Phase 1 /2a study. Five hundred subjects were simulated with 15 sampling points for each subject for each dosing regimen. The percentage of the population with FVIII activity above or equal to 1% and 3% at different times following different dosing regimens of ADVATE® or rFVIIIFc was estimated.
[00294] Statistical Analyses - Selected PK parameters for rFVIIIFc and ADVATE® were compared using an analysis of variance model. PK parameters were log-transformed for these analyses and estimated means, mean differences, and confidence intervals on the log-scale were transformed to obtain estimates for geometric means, geometric mean ratios (GMR), and confidence intervals, respectively, on the original scale. The GMR is the geometric mean of the intra-subject ratio of the rFVIIIFc PK parameter value to the ADVATE® PK parameter value. Results
[00295] Subject Disposition- Nineteen subjects were enrolled in the study; 16 underwent PK evaluation for both ADVATE® and rFVIIIFc. One subject self-administered his previous product prior to completing the wash-out period following the dose with ADVATE® and was thus excluded from the PK analysis, but was included in the safety analysis. Three subjects were discontinued from the study before receiving either study drug: one voluntarily withdrew; a second was withdrawn by the Investigator for non-compliance; and one was withdrawn at the Sponsor's request due to completion of study enrollment. Of the subjects dosed, six subjects received 25 IU/kg and 10 subjects received 65 IU/kg of both ADVATE® and rFVIIIFc. Mean age was 40.3 years (23 to 61 years). Genotypic identification was collected for seven subjects; inversion of intron 22 was reported in six subjects; and a frame-shift defect was reported in one subject. The genotype was unknown for nine subjects. Thirteen subjects had hepatitis C antibodies, four of whom were also positive for HIV.
[00296] Safety - Forty- four treatment-emergent AEs were reported by 11 (69%) subjects during the treatment and follow-up periods. This included the day of dosing with Advate or rFVIIIFc through a 28-day post-dosing observation period. The majority of events were considered mild and none led to withdrawal from the study. One event, dysgeusia, occurred transiently in one subject while receiving a 65 IU/kg dose of rFVIIIFc and was considered related to rFVIIIFc.
[00297] One subject experienced an anxiety attack after receiving 65 IU/kg of rFVIIIFc which resulted in 21 AEs, 19 of which were graded as mild, and two of which (headache and photophobia) were rated as moderate. Neither was deemed related to rFVIIIFc by the Investigator.
[00298] No serious bleeding episodes were reported. No evidence of allergic reactions to injection was detected. All plasma samples tested negative for FVIII inhibitors and anti-rFVIIIFc antibodies. No signs of injection site reactions were observed. No clinically meaningful changes in abnormal laboratory values were reported.
[00299] Pharmacokinetics: Correlation Between aPTT and Chromogenic Activity for rFVIIIFc in
Plasma - ADVATE® and rFVIIIFc activities were determined in the same assays using commercially available reagents and calibration against normal human plasma standards. There was a strong correlation between the results obtained by the one-stage clotting assay and the chromogenic assay in samples that had an activity above the LLOQ. Correlation coefficients (Pearson R2) of 0.94 and 0.95 were observed between the two assay results for 151 samples following ADVATE® dosing and 185 samples following rFVIIIFc dosing, respectively. Compared to the aPTT results, the chromogenic FVIII activities were, on average, 21 > higher for ADVATE® and 32% higher for rFVIIIFc, not statistically significant (Figure 9). This observation led to a slightly higher estimation of exposure parameters by the chromogenic assessment for both drugs. The apparent higher FVIII recoveries by the chromogenic assay are typical for recombinant FVIII products tested in clinical assays, and are in agreement with most other marketed FVIII products.30"32
[00300] Improved Pharmacokinetics for rFVIIIFc - The primary PK estimates were derived from one-stage (aPTT) clotting assay activity data. In subjects who received 25 or 65 IU/kg of ADVATE followed by an equal dose of rFVIIIFc, the plasma FVIII activity rose sharply and reached within the first hour following dosing. The subsequent decline of the observed FVIII activity exhibited monoexponential decay characteristics until the baseline FVIII activity was reached (Figure 10). The Cmax increased proportionally to the dose, but was comparable between equal doses of ADVATE® and rFVIIIFc (Table 8) The total exposure (AUCINF) also increased proportionally to the dose. However, the AUC of rFVIIIFc was 1.48 and 1.56-fold greater than that of ADVATE® at 25 IU/kg (p=0.002) and 65 IU/kg (p<0.001), respectively (Table 8).
Table 8. PK Parameters by One-Stage (aPTT) Assay for rFVIIIFc and ADVATE Per Dose Group
Figure imgf000082_0001
CI = Confidence Interval; Geom. Mean = Geometric Mean; OBS = observed. Estimated means, 95% CI for means, and mean differences were transformed to obtain estimated geometric means, 95% CI for geometric means, and geometric mean ratios, respectively.
[00301] The t1/2, MRT, CL, and Vss appeared to be independent of dose (Table 8). The geometric mean t1/2 of rFVIIIFc was 18.8 hours for both the 25 IU/kg and 65 IU/kg dose groups. This represents a 1.54 and 1.70-fold improvement over that of ADVATE® (12.2 hours and 1 1.0 hours) at equivalent doses (p<0.001), respectively (Table 8). The same intra-subject improvement was observed in the MRT of rFVIIIFc (27.0 hours for both dose groups) compared with ADVATE® (17.5 hours for the 25 IU/kg and 15.8 hours for the 65 IU/kg) (p<0.001). Consistent with improvement in the t1/2 and MRT was a corresponding 1.49 and 1.56-fold reduction in intra- subject CL at doses of 25 IU/kg (p=0.002) and 65 IU/kg (p<0.001), respectively. There were no significant differences in Vss and incremental recovery between ADVATE and rFVIIIFc. Therefore, within each subject, rFVIIIFc demonstrated an improved PK profile compared with ADVATE®. It was also observed that the patients with shorter half-life on ADVATE® had shorter half-life on rFVIIIFc, and patients with longer half-life on ADVATE® had longer half-life on rFVIIIFc.
[00302] The improved PK profile of rFVIIIFc resulted in increased time post-dosing to 1% FVIII activity which was 1.53 and 1.68-fold longer respectively, than with ADVATE® at 25 IU/kg (p<0.001) and 65 IU/kg (p<0.001) (data not shown), suggesting a potentially longer therapeutic duration for rFVIIIFc.
[00303] The favorable PK profile of rFVIIIFc relative to ADVATE® was also demonstrated by
FVIII activity measured in the chromogenic assay (Table 9), which was comparable to data derived from aPTT assays. The estimation of exposure, ie, and AUC , was slightly higher, however, based on the chromogenic assay than on the one-stage (aPTT) clotting assay for both ADVATE® and rFVIIIFc.
Table 9. PK Parameters by Two-Stage (Chromogenic) Assay for rFVIIIFc and ADVATE Per Dose Group
Figure imgf000084_0001
CI = Confidence Interval; Geom. Mean = Geometric Mean; OBS = observed. Estimated means, 95%> CI for means, and mean differences were transformed to obtain estimated geometric means, 95%> CI for geometric means, and geometric mean ratios, respectively.
[00304] Correlation Between von Willebrand Factor and Disposition of rFVIIIFc - Because the majority of FVIII in circulation is in complex with VWF33 and because the genome -wide association study has identified that the genetic determinants of FVIII levels are primarily dependent on VWF levels,34 we examined the association between VWF and rFVIIIFc. A strong correlation was observed between VWF levels and CL and t1/2 for both rFVIIIFc and ADVATE®. As shown in Figure 10, as the level of VWF increased, the CL of rFVIIIFc (p=0.0016) and of ADVATE® (p=0.0012) decreased.
[00305] The opposite relationship was observed between the level of VWF and t1/2. As the level of
VWF increased, the t1/2 of rFVIIIFc (p=0.0003) and of ADVATE® (p<0.0001) increased. This correlation suggests that the Fc moiety of rFVIIIFc does not alter the role of VWF in protecting FVIII from clearance.
[00306] Effects of Prolonged PK of rFVIIIFc on Whole Blood ROTEM® - Prior to administration of study drug, blood from one subject in each dose group was spiked with an equal dose of rFVIIIFc or ADVATE® and analyzed by whole blood ROTEM®. Clotting time (CT) was linear with respect to the dose in the range of approximately 1% to 100% of normal, and the dose response was comparable between rFVIIIFc and ADVATE® in the same subject (data not shown), indicating comparable potency of rFVIIIFc and ADVATE® in clot formation.
[00307] Despite the variable baseline CT due to residual FVIII levels prior to the administration of
ADVATE® or rFVIIIFc, both products effectively corrected the CT to comparable levels 30 minutes post-dosing (Figure 12). The improvement in CT was better sustained by rFVIIIFc than ADVATE® after 3 hours following a dose of 25 IU/kg (Figure 12A), and after 24 hours following a dose of 65 IU/kg (Figure 12B).
[00308] rFVIIIFc was well tolerated by subjects at both doses. There were no clinically significant changes observed in hematology, blood chemistry, or urinalysis parameters. The majority of AEs were mild, unrelated to rFVIIIFc, and resolved without sequelae. No serious AEs or deaths occurred during the study, and no subjects at either dose developed neutralizing or binding antibodies to rFVIIIFc.
[00309] rFVIIIFc demonstrated a significantly improved FVIII activity PK profile relative to
ADVATE®, with Ιυ2 and MRT across dose levels being 1.54 to 1.71-fold longer, as measured by the one-stage (aPTT) clotting assay and 1.59 to 1.84-fold longer by the two-stage chromogenic assay. The prolonged activity of rFVIIIFc predicts possible prolonged efficacy, allowing for a less frequent dosing regimen in the prophylactic treatment of patients with Hemophilia A.
[00310] Adopting the PK parameters derived from this study, the Monte Carlo simulation predicts that a higher percentage of patients receiving rFVIIIFc will sustain FVIII levels above 1%> or 3% as compared with patients receiving equal doses of ADVATE® (Table 10). For example, at a dose of 25 IU/kg, 12.2% of ADVATE® patients versus 71.2% of rFVIIIFc patients are predicted to have FVIII trough levels above 1% on Day 4; at a dose of 65 IU/kg, 11.0% of ADVATE® patients versus 66.4% of rFVIIIFc patients are predicted to have FVIII levels above 3% on Day 4. Clinical trials in larger numbers of patients are planned to confirm results from this Phase l/2a study and from the Monte Carlo simulation predictions. Table 10. Predicted Percentage of Subjects Achieving FVIII Trough Levels Above 1% and 3% of Normal at a Specified Dose Regimen of ADVATE® or rFVIIIFc
Figure imgf000086_0001
[00311] Despite the success of Fc fusion technology in prolonging circulating t1/2 for a variety of protein therapeutics, rFVIII was considered too large to successfully produce dimeric Fc fusions. We thus created a monomeric Fc fusion protein whereby a single effector molecule was covalent linked to a dimeric Fc, enabling binding to intracellular FcRn and subsequent recycling.21'22 In vitro coagulation assays demonstrate no loss of specific activity for rFVIIIFc, compared to B- domain deleted or native FVIII, by either clotting or chromogenic assays, using commercially available reagents and commonly used FVIII reference standards (JAD, TL, SCL, et al., manuscript submitted August, 2011). In addition, these results indicate that rFVIIIFc can be reliably assayed in a clinic setting by either the one-stage assay or the chromogenic method.
[00312] In summary, this Phase l/2a clinical trial is the first trial to demonstrate the safety and prolonged t1/2 of rFVIIIFc in patients with severe hemophilia A. A pivotal Phase 3 study is ongoing with rFVIIIFc to establish effective prophylaxis dosing regimens for individuals with hemophilia A.
Example 11
[00313] A novel single-chain (SC) isoform of factor VIII (FVIII), resulting from incomplete proteolysis at residue R1648 during biosynthesis, may provide superior manufacturability and stability relative to native FVIII. A single recombinant B domain deleted factor VIII molecule fused to an immunoglobulin Fc domain (rFVIIIFc) and its purified SC counterpart (SC-rFVIIIFc) exhibited similar specific activity in one stage clotting assays using plasma depleted of von Willebrand factor (VWF), but SC-rFVIIIFc exhibited lower specific activity in the presence of VWF. This study was undertaken to determine if VWF -bound rFVIIIFc, SC-rFVIIIFc and rBDD- FVIII (XYNTHA®, REFACTO AF®) differ with respect to thrombin-mediated proteolytic release from VWF.
[00314] Equimolar amounts of rFVIIIFc, SC-rFVIIIFc, and rBDD-FVIII were captured on an optical biosensor chip on which human VWF had been immobilized by amine coupling. Human a-thrombin at a range of concentrations was infused over the chip surface, and the rates of FVIII release from immobilized VWF were monitored in real time. The half maximal effective concentration (EC50) of a-thrombin was determined for each FVIII species.
[00315] α-thrombin EC50 values for rFVIIIFc and rBDD-FVIII were comparable (3.7 ± 0.2 U/mL and 3.2 ± 0.3 U/mL, respectively), whereas the EC50 value for SC-rFVIIIFc was greater than 3- fold higher (11.7 ± 0.9 U/mL). This finding that SC-rFVIIIFc is released more slowly from VWF than are either rFVIIIFc or rBDD-FVIII is consistent with a previously observed finding regarding the activities of rFVIIIFc and SC-rFVIIIFc in a one-stage clotting assay (aPTT) in which SC-FVIIIFc had a lower apparent activity only when VWF was present in the assay plasma sample. However, all samples possessed equivalent activities in a mouse bleeding model, indicating that responsiveness of FVIII preparations to thrombin in the release of FVIII from VWF does not correlate with efficacy in vivo.
TABLE 1 : Polynucleotide Sequences A. B-Domain Deleted FVIIIFc
(i) B-Domain Deleted FVIIIFc Chain DNA Sequence (FVIII signal peptide underlined. Fc region in bold) (SEP ID NO: l. which encodes SEP ID NO:2
661 A TGCAAATAGA GCTCTCCACC TGCTTCTTTC
721 TGTGCCTTTT GCGATTCTGC TTTAGTGCCA CCAGAAGATA CTACCTGGGT GCAGTGGAAC
781 TGTCATGGGA CTATATGCAA AGTGATCTCG GTGAGCTGCC TGTGGACGCA AGATTTCCTC
841 CTAGAGTGCC AAAATCTTTT CCATTCAACA CCTCAGTCGT GTACAAAAAG ACTCTGTTTG
901 TAGAATTCAC GGATCACCTT TTCAACATCG CTAAGCCAAG GCCACCCTGG ATGGGTCTGC
961 TAGGTCCTAC CATCCAGGCT GAGGTTTATG ATACAGTGGT CATTACACTT AAGAACATGG
1021 CTTCCCATCC TGTCAGTCTT CATGCTGTTG GTGTATCCTA CTGGAAAGCT TCTGAGGGAG
1081 CTGAATATGA TGATCAGACC AGTCAAAGGG AGAAAGAAGA TGATAAAGTC TTCCCTGGTG
1141 GAAGCCATAC ATATGTCTGG CAGGTCCTGA AAGAGAATGG TCCAATGGCC TCTGACCCAC
1201 TGTGCCTTAC CTACTCATAT CTTTCTCATG TGGACCTGGT AAAAGACTTG AATTCAGGCC
1261 TCATTGGAGC CCTACTAGTA TGTAGAGAAG GGAGTCTGGC CAAGGAAAAG ACACAGACCT
1321 TGCACAAATT TATACTACTT TTTGCTGTAT TTGATGAAGG GAAAAGTTGG CACTCAGAAA
1381 CAAAGAACTC CTTGATGCAG GATAGGGATG CTGCATCTGC TCGGGCCTGG CCTAAAATGC
1441 ACACAGTCAA TGGTTATGTA AACAGGTCTC TGCCAGGTCT GATTGGATGC CACAGGAAAT
1501 CAGTCTATTG GCATGTGATT GGAATGGGCA CCACTCCTGA AGTGCACTCA ATATTCCTCG
1561 AAGGTCACAC ATTTCTTGTG AGGAACCATC GCCAGGCGTC CTTGGAAATC TCGCCAATAA
1621 CTTTCCTTAC TGCTCAAACA CTCTTGATGG ACCTTGGACA GTTTCTACTG TTTTGTCATA
1681 TCTCTTCCCA CCAACATGAT GGCATGGAAG CTTATGTCAA AGTAGACAGC TGTCCAGAGG
1741 AACCCCAACT ACGAATGAAA AATAATGAAG AAGCGGAAGA CTATGATGAT GATCTTACTG
1801 ATTCTGAAAT GGATGTGGTC AGGTTTGATG ATGACAACTC TCCTTCCTTT ATCCAAATTC
1861 GCTCAGTTGC CAAGAAGCAT CCTAAAACTT GGGTACATTA CATTGCTGCT GAAGAGGAGG
1921 ACTGGGACTA TGCTCCCTTA GTCCTCGCCC CCGATGACAG AAGTTATAAA AGTCAATATT
1981 TGAACAATGG CCCTCAGCGG ATTGGTAGGA AGTACAAAAA AGTCCGATTT ATGGCATACA
2041 CAGATGAAAC CTTTAAGACT CGTGAAGCTA TTCAGCATGA ATCAGGAATC TTGGGACCTT
2101 TACTTTATGG GGAAGTTGGA GACACACTGT TGATTATATT TAAGAATCAA GCAAGCAGAC
2161 CATATAACAT CTACCCTCAC GGAATCACTG ATGTCCGTCC TTTGTATTCA AGGAGATTAC
2221 CAAAAGGTGT AAAACATTTG AAGGATTTTC CAATTCTGCC AGGAGAAATA TTCAAATATA
2281 AATGGACAGT GACTGTAGAA GATGGGCCAA CTAAATCAGA TCCTCGGTGC CTGACCCGCT
2341 ATTACTCTAG TTTCGTTAAT ATGGAGAGAG ATCTAGCTTC AGGACTCATT GGCCCTCTCC
2401 TCATCTGCTA CAAAGAATCT GTAGATCAAA GAGGAAACCA GATAATGTCA GACAAGAGGA
2461 ATGTCATCCT GTTTTCTGTA TTTGATGAGA ACCGAAGCTG GTACCTCACA GAGAATATAC
2521 AACGCTTTCT CCCCAATCCA GCTGGAGTGC AGCTTGAGGA TCCAGAGTTC CAAGCCTCCA
2581 ACATCATGCA CAGCATCAAT GGCTATGTTT TTGATAGTTT GCAGTTGTCA GTTTGTTTGC
2641 ATGAGGTGGC ATACTGGTAC ATTCTAAGCA TTGGAGCACA GACTGACTTC CTTTCTGTCT
2701 TCTTCTCTGG ATATACCTTC AAACACAAAA TGGTCTATGA AGACACACTC ACCCTATTCC
2761 CATTCTCAGG AGAAACTGTC TTCATGTCGA TGGAAAACCC AGGTCTATGG ATTCTGGGGT
2821 GCCACAACTC AGACTTTCGG AACAGAGGCA TGACCGCCTT ACTGAAGGTT TCTAGTTGTG
2881 ACAAGAACAC TGGTGATTAT TACGAGGACA GTTATGAAGA TATTTCAGCA TACTTGCTGA
2941 GTAAAAACAA TGCCATTGAA CCAAGAAGCT TCTCTCAAAA CCCACCAGTC TTGAAACGCC
3001 ATCAACGGGA AATAACTCGT ACTACTCTTC AGTCAGATCA AGAGGAAATT GACTATGATG
3061 ATACCATATC AGTTGAAATG AAGAAGGAAG ATTTTGACAT TTATGATGAG GATGAAAATC
3121 AGAGCCCCCG CAGCTTTCAA AAGAAAACAC GACACTATTT TATTGCTGCA GTGGAGAGGC
3181 TCTGGGATTA TGGGATGAGT AGCTCCCCAC ATGTTCTAAG AAACAGGGCT CAGAGTGGCA
3241 GTGTCCCTCA GTTCAAGAAA GTTGTTTTCC AGGAATTTAC TGATGGCTCC TTTACTCAGC
3301 CCTTATACCG TGGAGAACTA AATGAACATT TGGGACTCCT GGGGCCATAT ATAAGAGCAG
3361 AAGTTGAAGA TAATATCATG GTAACTTTCA GAAATCAGGC CTCTCGTCCC TATTCCTTCT
3421 ATTCTAGCCT TATTTCTTAT GAGGAAGATC AGAGGCAAGG AGCAGAACCT AGAAAAAACT
3481 TTGTCAAGCC TAATGAAACC AAAACTTACT TTTGGAAAGT GCAACATCAT ATGGCACCCA
3541 CTAAAGATGA GTTTGACTGC AAAGCCTGGG CTTATTTCTC TGATGTTGAC CTGGAAAAAG
3601 ATGTGCACTC AGGCCTGATT GGACCCCTTC TGGTCTGCCA CACTAACACA CTGAACCCTG 3661 CTCATGGGAG ACAAGTGACA GTACAGGAAT TTGCTCTGTT TTTCACCATC TTTGATGAGA
3721 CCAAAAGCTG GTACTTCACT GAAAATATGG AAAGAAACTG CAGGGCTCCC TGCAATATCC
3781 AGATGGAAGA TCCCACTTTT AAAGAGAATT ATCGCTTCCA TGCAATCAAT GGCTACATAA
3841 TGGATACACT ACCTGGCTTA GTAATGGCTC AGGATCAAAG GATTCGATGG TATCTGCTCA
3901 GCATGGGCAG CAATGAAAAC ATCCATTCTA TTCATTTCAG TGGACATGTG TTCACTGTAC
3961 GAAAAAAAGA GGAGTATAAA ATGGCACTGT ACAATCTCTA TCCAGGTGTT TTTGAGACAG
4021 TGGAAATGTT ACCATCCAAA GCTGGAATTT GGCGGGTGGA ATGCCTTATT GGCGAGCATC
4081 TACATGCTGG GATGAGCACA CTTTTTCTGG TGTACAGCAA TAAGTGTCAG ACTCCCCTGG
4141 GAATGGCTTC TGGACACATT AGAGATTTTC AGATTACAGC TTCAGGACAA TATGGACAGT
4201 GGGCCCCAAA GCTGGCCAGA CTTCATTATT CCGGATCAAT CAATGCCTGG AGCACCAAGG
4261 AGCCCTTTTC TTGGATCAAG GTGGATCTGT TGGCACCAAT GATTATTCAC GGCATCAAGA
4321 CCCAGGGTGC CCGTCAGAAG TTCTCCAGCC TCTACATCTC TCAGTTTATC ATCATGTATA
4381 GTCTTGATGG GAAGAAGTGG CAGACTTATC GAGGAAATTC CACTGGAACC TTAATGGTCT
4441 TCTTTGGCAA TGTGGATTCA TCTGGGATAA AACACAATAT TTTTAACCCT CCAATTATTG
4501 CTCGATACAT CCGTTTGCAC CCAACTCATT ATAGCATTCG CAGCACTCTT CGCATGGAGT
4561 TGATGGGCTG TGATTTAAAT AGTTGCAGCA TGCCATTGGG AATGGAGAGT AAAGCAATAT
4621 CAGATGCACA GATTACTGCT TCATCCTACT TTACCAATAT GTTTGCCACC TGGTCTCCTT
4681 CAAAAGCTCG ACTTCACCTC CAAGGGAGGA GTAATGCCTG GAGACCTCAG GTGAATAATC
4741 CAAAAGAGTG GCTGCAAGTG GACTTCCAGA AGACAATGAA AGTCACAGGA GTAACTACTC
4801 AGGGAGTAAA ATCTCTGCTT ACCAGCATGT ATGTGAAGGA GTTCCTCATC TCCAGCAGTC
4861 AAGATGGCCA TCAGTGGACT CTCTTTTTTC AGAATGGCAA AGTAAAGGTT TTTCAGGGAA
4921 ATCAAGACTC CTTCACACCT GTGGTGAACT CTCTAGACCC ACCGTTACTG ACTCGCTACC
4981 TTCGAATTCA CCCCCAGAGT TGGGTGCACC AGATTGCCCT GAGGATGGAG GTTCTGGGCT
5041 GCGAGGCACA GGACCTCTAC GACAAAACTC ACACATGCCC ACCGTGCCCA GCTCCAGAAC
5101 TCCTGGGCGG ACCGTCAGTC TTCCTCTTCC CCCCAAAACC CAAGGACACC CTCATGATCT
5161 CCCGGACCCC TGAGGTCACA TGCGTGGTGG TGGACGTGAG CCACGAAGAC CCTGAGGTCA
5221 AGTTCAACTG GTACGTGGAC GGCGTGGAGG TGCATAATGC CAAGACAAAG CCGCGGGAGG
5281 AGCAGTACAA CAGCACGTAC CGTGTGGTCA GCGTCCTCAC CGTCCTGCAC CAGGACTGGC
5341 TGAATGGCAA GGAGTACAAG TGCAAGGTCT CCAACAAAGC CCTCCCAGCC CCCATCGAGA
5401 AAACCATCTC CAAAGCCAAA GGGCAGCCCC GAGAACCACA GGTGTACACC CTGCCCCCAT
5461 CCCGGGATGA GCTGACCAAG AACCAGGTCA GCCTGACCTG CCTGGTCAAA GGCTTCTATC
5521 CCAGCGACAT CGCCGTGGAG TGGGAGAGCA ATGGGCAGCC GGAGAACAAC TACAAGACCA
5581 CGCCTCCCGT GTTGGACTCC GACGGCTCCT TCTTCCTCTA CAGCAAGCTC ACCGTGGACA
5641 AGAGCAGGTG GCAGCAGGGG AACGTCTTCT CATGCTCCGT GATGCATGAG GCTCTGCACA
5701 ACCACTACAC GCAGAAGAGC CTCTCCCTGT CTCCGGGTAA A
(ii Fc DNA sequence (mouse IgK signal peptide underlined) (SEQ ID NO:3, which encodes SEQ ID NO:4
7981 ATGGA GACAGACACA
8041 CTCCTGCTAT GGGTACTGCT GCTCTGGGTT CCAGGTTCCA CTGGTGACAA AACTCACACA
8101 TGCCCACCGT GCCCAGCACC TGAACTCCTG GGAGGACCGT CAGTCTTCCT CTTCCCCCCA
8161 AAACCCAAGG ACACCCTCAT GATCTCCCGG ACCCCTGAGG TCACATGCGT GGTGGTGGAC
8221 GTGAGCCACG AAGACCCTGA GGTCAAGTTC AACTGGTACG TGGACGGCGT GGAGGTGCAT
8281 AATGCCAAGA CAAAGCCGCG GGAGGAGCAG TACAACAGCA CGTACCGTGT GGTCAGCGTC
8341 CTCACCGTCC TGCACCAGGA CTGGCTGAAT GGCAAGGAGT ACAAGTGCAA GGTCTCCAAC
8401 AAAGCCCTCC CAGCCCCCAT CGAGAAAACC ATCTCCAAAG CCAAAGGGCA GCCCCGAGAA
8461 CCACAGGTGT ACACCCTGCC CCCATCCCGC GATGAGCTGA CCAAGAACCA GGTCAGCCTG
8521 ACCTGCCTGG TCAAAGGCTT CTATCCCAGC GACATCGCCG TGGAGTGGGA GAGCAATGGG
8581 CAGCCGGAGA ACAACTACAA GACCACGCCT CCCGTGTTGG ACTCCGACGG CTCCTTCTTC
8641 CTCTACAGCA AGCTCACCGT GGACAAGAGC AGGTGGCAGC AGGGGAACGT CTTCTCATGC
8701 TCCGTGATGC ATGAGGCTCT GCACAACCAC TACACGCAGA AGAGCCTCTC CCTGTCTCCG
8761 GGTAAA B. Full Length FVIIIFc
(Ϊ) Full Length FVIIIFc DNA Sequence (FVIII signal peptide underlined. Fc region in bold) (SEP ID NO:5. which encodes SEP ID NO:6
661 ATG CAAATAGAGC TCTCCACCTG
721 CTTCTTTCTG TGCCTTTTGC GATTCTGCTT TAGTGCCACC AGAAGATACT ACCTGGGTGC
781 AGTGGAACTG TCATGGGACT ATATGCAAAG TGATCTCGGT GAGCTGCCTG TGGACGCAAG
841 ATTTCCTCCT AGAGTGCCAA AATCTTTTCC ATTCAACACC TCAGTCGTGT ACAAAAAGAC
901 TCTGTTTGTA GAATTCACGG ATCACCTTTT CAACATCGCT AAGCCAAGGC CACCCTGGAT
961 GGGTCTGCTA GGTCCTACCA TCCAGGCTGA GGTTTATGAT ACAGTGGTCA TTACACTTAA
1021 GAACATGGCT TCCCATCCTG TCAGTCTTCA TGCTGTTGGT GTATCCTACT GGAAAGCTTC
1081 TGAGGGAGCT GAATATGATG ATCAGACCAG TCAAAGGGAG AAAGAAGATG ATAAAGTCTT
1141 CCCTGGTGGA AGCCATACAT ATGTCTGGCA GGTCCTGAAA GAGAATGGTC CAATGGCCTC
1201 TGACCCACTG TGCCTTACCT ACTCATATCT TTCTCATGTG GACCTGGTAA AAGACTTGAA
1261 TTCAGGCCTC ATTGGAGCCC TACTAGTATG TAGAGAAGGG AGTCTGGCCA AGGAAAAGAC
1321 ACAGACCTTG CACAAATTTA TACTACTTTT TGCTGTATTT GATGAAGGGA AAAGTTGGCA
1381 CTCAGAAACA AAGAACTCCT TGATGCAGGA TAGGGATGCT GCATCTGCTC GGGCCTGGCC
1441 TAAAATGCAC ACAGTCAATG GTTATGTAAA CAGGTCTCTG CCAGGTCTGA TTGGATGCCA
1501 CAGGAAATCA GTCTATTGGC ATGTGATTGG AATGGGCACC ACTCCTGAAG TGCACTCAAT
1561 ATTCCTCGAA GGTCACACAT TTCTTGTGAG GAACCATCGC CAGGCGTCCT TGGAAATCTC-
1621 GCCAATAACT TTCCTTACTG CTCAAACACT CTTGATGGAC CTTGGACAGT TTCTACTGTT
1681 TTGTCATATC TCTTCCCACC AACATGATGG CATGGAAGCT TATGTCAAAG TAGACAGCTG
1741 TCCAGAGGAA CCCCAACTAC GAATGAAAAA TAATGAAGAA GCGGAAGACT ATGATGATGA
1801 TCTTACTGAT TCTGAAATGG ATGTGGTCAG GTTTGATGAT GACAACTCTC CTTCCTTTAT
1861 CCAAATTCGC TCAGTTGCCA AGAAGCATCC TAAAACTTGG GTACATTACA TTGCTGCTGA
1921 AGAGGAGGAC TGGGACTATG CTCCCTTAGT CCTCGCCCCC GATGACAGAA GTTATAAAAG
1981 TCAATATTTG AACAATGGCC CTCAGCGGAT TGGTAGGAAG TACAAAAAAG TCCGATTTAT
2041 GGCATACACA GATGAAACCT TTAAGACTCG TGAAGCTATT CAGCATGAAT CAGGAATCTT
2101 GGGACCTTTA CTTTATGGGG AAGTTGGAGA CACACTGTTG ATTATATTTA AGAATCAAGC
2161 AAGCAGACCA TATAACATCT ACCCTCACGG AATCACTGAT GTCCGTCCTT TGTATTCAAG
2221 GAGATTACCA AAAGGTGTAA AACATTTGAA GGATTTTCCA ATTCTGCCAG GAGAAATATT
2281 CAAATATAAA TGGACAGTGA CTGTAGAAGA TGGGCCAACT AAATCAGATC CTCGGTGCCT
2341 GACCCGCTAT TACTCTAGTT TCGTTAATAT GGAGAGAGAT CTAGCTTCAG GACTCATTGG
2401 CCCTCTCCTC ATCTGCTACA AAGAATCTGT AGATCAAAGA GGAAACCAGA TAATGTCAGA
2461 CAAGAGGAAT GTCATCCTGT TTTCTGTATT TGATGAGAAC CGAAGCTGGT ACCTCACAGA
2521 GAATATACAA CGCTTTCTCC CCAATCCAGC TGGAGTGCAG CTTGAGGATC CAGAGTTCCA
2581 AGCCTCCAAC ATCATGCACA GCATCAATGG CTATGTTTTT GATAGTTTGC AGTTGTCAGT
2641 TTGTTTGCAT GAGGTGGCAT ACTGGTACAT TCTAAGCATT GGAGCACAGA CTGACTTCCT
2701 TTCTGTCTTC TTCTCTGGAT ATACCTTCAA ACACAAAATG GTCTATGAAG ACACACTCAC
2761 CCTATTCCCA TTCTCAGGAG AAACTGTCTT CATGTCGATG GAAAACCCAG GTCTATGGAT
2821 TCTGGGGTGC CACAACTCAG ACTTTCGGAA CAGAGGCATG ACCGCCTTAC TGAAGGTTTC
2881 TAGTTGTGAC AAGAACACTG GTGATTATTA CGAGGACAGT TATGAAGATA TTTCAGCATA
2941 CTTGCTGAGT AAAAACAATG CCATTGAACC AAGAAGCTTC TCCCAGAATT CAAGACACCC
3001 TAGCACTAGG CAAAAGCAAT TTAATGCCAC CACAATTCCA GAAAATGACA TAGAGAAGAC
3061 TGACCCTTGG TTTGCACACA GAACACCTAT GCCTAAAATA CAAAATGTCT CCTCTAGTGA
3121 TTTGTTGATG CTCTTGCGAC AGAGTCCTAC TCCACATGGG CTATCCTTAT CTGATCTCCA
3181 AGAAGCCAAA TATGAGACTT TTTCTGATGA TCCATCACCT GGAGCAATAG ACAGTAATAA
3241 CAGCCTGTCT GAAATGACAC ACTTCAGGCC ACAGCTCCAT CACAGTGGGG ACATGGTATT
3301 TACCCCTGAG TCAGGCCTCC AATTAAGATT AAATGAGAAA CTGGGGACAA CTGCAGCAAC
3361 AGAGTTGAAG AAACTTGATT TCAAAGTTTC TAGTACATCA AATAATCTGA TTTCAACAAT
3421 TCCATCAGAC AATTTGGCAG CAGGTACTGA TAATACAAGT TCCTTAGGAC CCCCAAGTAT
3481 GCCAGTTCAT TATGATAGTC AATTAGATAC CACTCTATTT GGCAAAAAGT CATCTCCCCT
3541 TACTGAGTCT GGTGGACCTC TGAGCTTGAG TGAAGAAAAT AATGATTCAA AGTTGTTAGA
3601 ATCAGGTTTA ATGAATAGCC AAGAAAGTTC ATGGGGAAAA AATGTATCGT CAACAGAGAG
3661 TGGTAGGTTA TTTAAAGGGA AAAGAGCTCA TGGACCTGCT TTGTTGACTA AAGATAATGC
3721 CTTATTCAAA GTTAGCATCT CTTTGTTAAA GACAAACAAA ACTTCCAATA ATTCAGCAAC
3781 TAATAGAAAG ACTCACATTG ATGGCCCATC ATTATTAATT GAGAATAGTC CATCAGTCTG
3841 GCAAAATATA TTAGAAAGTG ACACTGAGTT TAAAAAAGTG ACACCTTTGA TTCATGACAG
3901 AATGCTTATG GACAAAAATG CTACAGCTTT GAGGCTAAAT CATATGTCAA ATAAAACTAC
3961 TTCATCAAAA AACATGGAAA TGGTCCAACA GAAAAAAGAG GGCCCCATTC CACCAGATGC
4021 ACAAAATCCA GATATGTCGT TCTTTAAGAT GCTATTCTTG CCAGAATCAG CAAGGTGGAT
4081 ACAAAGGACT CATGGAAAGA ACTCTCTGAA CTCTGGGCAA GGCCCCAGTC CAAAGCAATT
4141 AGTATCCTTA GGACCAGAAA AATCTGTGGA AGGTCAGAAT TTCTTGTCTG AGAAAAACAA
4201 AGTGGTAGTA GGAAAGGGTG AATTTACAAA GGACGTAGGA CTCAAAGAGA TGGTTTTTCC 4261 AAGCAGCAGA AACCTATTTC TTACTAACTT GGATAATTTA CATGAAAATA ATACACACAA
4321 TCAAGAAAAA AAAATTCAGG AAGAAATAGA AAAGAAGGAA ACATTAATCC AAGAGAATGT
4381 AGTTTTGCCT CAGATACATA CAGTGACTGG CACTAAGAAT TTCATGAAGA ACCTTTTCTT
4441 ACTGAGCACT AGGCAAAATG TAGAAGGTTC ATATGACGGG GCATATGCTC CAGTACTTCA
4501 AGATTTTAGG TCATTAAATG ATTCAACAAA TAGAACAAAG AAACACACAG CTCATTTCTC
4561 AAAAAAAGGG GAGGAAGAAA ACTTGGAAGG CTTGGGAAAT CAAACCAAGC AAATTGTAGA
4621 GAAATATGCA TGCACCACAA GGATATCTCC TAATACAAGC CAGCAGAATT TTGTCACGCA
4681 ACGTAGTAAG AGAGCTTTGA AACAATTCAG ACTCCCACTA GAAGAAACAG AACTTGAAAA
4741 AAGGATAATT GTGGATGACA CCTCAACCCA GTGGTCCAAA AACATGAAAC ATTTGACCCC
4801 GAGCACCCTC ACACAGATAG ACTACAATGA GAAGGAGAAA GGGGCCATTA CTCAGTCTCC
4861 CTTATCAGAT TGCCTTACGA GGAGTCATAG CATCCCTCAA GCAAATAGAT CTCCATTACC
4921 CATTGCAAAG GTATCATCAT TTCCATCTAT TAGACCTATA TATCTGACCA GGGTCCTATT
4981 CCAAGACAAC TCTTCTCATC TTCCAGCAGC ATCTTATAGA AAGAAAGATT CTGGGGTCCA 5041 AGAAAGCAGT CATTTCTTAC AAGGAGCCAA AAAAAATAAC CTTTCTTTAG CCATTCTAAC 5101 CTTGGAGATG ACTGGTGATC AAAGAGAGGT TGGCTCCCTG GGGACAAGTG CCACAAATTC 5161 AGTCACATAC AAGAAAGTTG AGAACACTGT TCTCCCGAAA CCAGACTTGC CCAAAACATC 5221 TGGCAAAGTT GAATTGCTTC CAAAAGTTCA CATTTATCAG AAGGACCTAT TCCCTACGGA 5281 AACTAGCAAT GGGTCTCCTG GCCATCTGGA TCTCGTGGAA GGGAGCCTTC TTCAGGGAAC 5341 AGAGGGAGCG ATTAAGTGGA ATGAAGCAAA CAGACCTGGA AAAGTTCCCT TTCTGAGAGT 5401 AGCAACAGAA AGCTCTGCAA AGACTCCCTC CAAGCTATTG GATCCTCTTG CTTGGGATAA 5461 CCACTATGGT ACTCAGATAC CAAAAGAAGA GTGGAAATCC CAAGAGAAGT CACCAGAAAA 5521 AACAGCTTTT AAGAAAAAGG ATACCATTTT GTCCCTGAAC GCTTGTGAAA GCAATCATGC 5581 AATAGCAGCA ATAAATGAGG GACAAAATAA GCCCGAAATA GAAGTCACCT GGGCAAAGCA
5641 AGGTAGGACT GAAAGGCTGT GCTCTCAAAA CCCACCAGTC TTGAAACGCC ATCAACGGGA 5701 AATAACTCGT ACTACTCTTC AGTCAGATCA AGAGGAAATT GACTATGATG ATACCATATC 5761 AGTTGAAATG AAGAAGGAAG ATTTTGACAT TTATGATGAG GATGAAAATC AGAGCCCCCG 5821 CAGCTTTCAA AAGAAAACAC GACACTATTT TATTGCTGCA GTGGAGAGGC TCTGGGATTA 5881 TGGGATGAGT AGCTCCCCAC ATGTTCTAAG AAACAGGGCT CAGAGTGGCA GTGTCCCTCA
5941 GTTCAAGAAA GTTGTTTTCC AGGAATTTAC TGATGGCTCC TTTACTCAGC CCTTATACCG 6001 TGGAGAACTA AATGAACATT TGGGACTCCT GGGGCCATAT ATAAGAGCAG AAGTTGAAGA 6061 TAATATCATG GTAACTTTCA GAAATCAGGC CTCTCGTCCC TATTCCTTCT ATTCTAGCCT 6121 TATTTCTTAT GAGGAAGATC AGAGGCAAGG AGCAGAACCT AGAAAAAACT TTGTCAAGCC 6181 TAATGAAACC AAAACTTACT TTTGGAAAGT GCAACATCAT ATGGCACCCA CTAAAGATGA 6241 GTTTGACTGC AAAGCCTGGG CTTATTTCTC TGATGTTGAC CTGGAAAAAG ATGTGCACTC 6301 AGGCCTGATT GGACCCCTTC TGGTCTGCCA CACTAACACA CTGAACCCTG CTCATGGGAG 6361 ACAAGTGACA GTACAGGAAT TTGCTCTGTT TTTCACCATC TTTGATGAGA CCAAAAGCTG 6421 GTACTTCACT GAAAATATGG AAAGAAACTG CAGGGCTCCC TGCAATATCC AGATGGAAGA 6481 TCCCACTTTT AAAGAGAATT ATCGCTTCCA TGCAATCAAT GGCTACATAA TGGATACACT 6541 ACCTGGCTTA GTAATGGCTC AGGATCAAAG GATTCGATGG TATCTGCTCA GCATGGGCAG
6601 CAATGAAAAC ATCCATTCTA TTCATTTCAG TGGACATGTG TTCACTGTAC GAAAAAAAGA 6661 GGAGTATAAA ATGGCACTGT ACAATCTCTA TCCAGGTGTT TTTGAGACAG TGGAAATGTT 6721 ACCATCCAAA GCTGGAATTT GGCGGGTGGA ATGCCTTATT GGCGAGCATC TACATGCTGG 6781 GATGAGCACA CTTTTTCTGG TGTACAGCAA TAAGTGTCAG ACTCCCCTGG GAATGGCTTC 6841 TGGACACATT AGAGATTTTC AGATTACAGC TTCAGGACAA TATGGACAGT GGGCCCCAAA 6901 GCTGGCCAGA CTTCATTATT CCGGATCAAT CAATGCCTGG AGCACCAAGG AGCCCTTTTC
6961 TTGGATCAAG GTGGATCTGT TGGCACCAAT GATTATTCAC GGCATCAAGA CCCAGGGTGC
7021 CCGTCAGAAG TTCTCCAGCC TCTACATCTC TCAGTTTATC ATCATGTATA GTCTTGATGG 7081 GAAGAAGTGG CAGACTTATC GAGGAAATTC CACTGGAACC TTAATGGTCT TCTTTGGCAA 7141 TGTGGATTCA TCTGGGATAA AACACAATAT TTTTAACCCT CCAATTATTG CTCGATACAT 7201 CCGTTTGCAC CCAACTCATT ATAGCATTCG CAGCACTCTT CGCATGGAGT TGATGGGCTG 7261 TGATTTAAAT AGTTGCAGCA TGCCATTGGG AATGGAGAGT AAAGCAATAT CAGATGCACA 7321 GATTACTGCT TCATCCTACT TTACCAATAT GTTTGCCACC TGGTCTCCTT CAAAAGCTCG 7381 ACTTCACCTC CAAGGGAGGA GTAATGCCTG GAGACCTCAG GTGAATAATC CAAAAGAGTG 7441 GCTGCAAGTG GACTTCCAGA AGACAATGAA AGTCACAGGA GTAACTACTC AGGGAGTAAA 7501 ATCTCTGCTT ACCAGCATGT ATGTGAAGGA GTTCCTCATC TCCAGCAGTC AAGATGGCCA 7561 TCAGTGGACT CTCTTTTTTC AGAATGGCAA AGTAAAGGTT TTTCAGGGAA ATCAAGACTC 7621 CTTCACACCT GTGGTGAACT CTCTAGACCC ACCGTTACTG ACTCGCTACC TTCGAATTCA 7681 CCCCCAGAGT TGGGTGCACC AGATTGCCCT GAGGATGGAG GTTCTGGGCT GCGAGGCACA 7741 GGACCTCTAC GACAAAACTC ACACATGCCC ACCGTGCCCA GCTCCAGAAC TCCTGGGCGG 7801 ACCGTCAGTC TTCCTCTTCC CCCCAAAACC CAAGGACACC CTCATGATCT CCCGGACCCC 7861 TGAGGTCACA TGCGTGGTGG TGGACGTGAG CCACGAAGAC CCTGAGGTCA AGTTCAACTG 7921 GTACGTGGAC GGCGTGGAGG TGCATAATGC CAAGACAAAG CCGCGGGAGG AGCAGTACAA 7981 CAGCACGTAC CGTGTGGTCA GCGTCCTCAC CGTCCTGCAC CAGGACTGGC TGAATGGCAA 8041 GGAGTACAAG TGCAAGGTCT CCAACAAAGC CCTCCCAGCC CCCATCGAGA AAACCATCTC 8101 CAAAGCCAAA GGGCAGCCCC GAGAACCACA GGTGTACACC CTGCCCCCAT CCCGGGATGA 8161 GCTGACCAAG AACCAGGTCA GCCTGACCTG CCTGGTCAAA GGCTTCTATC CCAGCGACAT 8221 CGCCGTGGAG TGGGAGAGCA ATGGGCAGCC GGAGAACAAC TACAAGACCA CGCCTCCCGT 8281 GTTGGACTCC GACGGCTCCT TCTTCCTCTA CAGCAAGCTC ACCGTGGACA AGAGCAGGTG 8341 GCAGCAGGGG AACGTCTTCT CATGCTCCGT GATGCATGAG GCTCTGCACA ACCACTACAC 8401 GCAGAAGAGC CTCTCCCTGT CTCCGGGTAA A
(ii) Fc (same sequence as A (ii) (SEQ ID NO:3))l
Table 2: Polypeptide Sequences
A. B-Domain Deleted FVIII-Fc Monomer Hybrid (BDD FVIIIFc monomer dimer): created by coexpressing BDD FVIIIFc and Fc chains.
Construct = HC-LC-Fc fusion. An Fc expression cassette is cotransfected with BDDFVIII-Fc to generate the BDD FVIIIFc monomer-. For the BDD FVIIIFc chain, the Fc sequence is shown in bold; HC sequence is shown in double underline; remaining B domain sequence is shown in italics. Signal peptides are underlined.
i) B domain deleted FVIII-Fc chain (19 amino acid signal sequence underlined) (SEP ID NP:2)
MQIELSTCFFLCLLRFCFS
ATRRYYLGAVELSWDYMQSDLGELPVDARFPPRVPKSFPFNTSWYKKTLFVEFTDHLFNIAKPRPPWMGL LGPTIQAEVYDTVVITLKNMASHPVSLHAVGVSYWKASEGAEYDDQTSQREKEDDKVFPGGSHTYVWQVLK ENGPMASDPLCLTYSYLSHVDLVKDLNSGLIGALLVCREGSLAKEKTQTLHKFILLFAVFDEGKSWHSETK NSLMQDRDAASARAWPKMHTVNGYVNRSLPGLIGCHRKSVYWHVIGMGTTPEVHSIFLEGHTFLVRNHRQA SLEISPITFLTAQTLLMDLGQFLLFCHISSHQHDGMEAYVKVDSCPEEPQLRMKNNEEAEDYDDDLTDSEM DWRFDDDNSPSFIQIRSVAKKHPKTWVHYIAAEEEDWDYAPLVLAPDDRSYKSQYLNNGPQRIGRKYKKV RFMAYTDETFKTREAIQHESGILGPLLYGEVGDTLLI IFKNQASRPYNIYPHGITDVRPLYSRRLPKGVKH LKDFPILPGEI FKYKWTVTVEDGPTKSDPRCLTRYYSSFVNMERDLASGLIGPLLICYKESVDQRGNQIMS DKRNVILFSVFDENRSWYLTENIQRFLPNPAGVQLEDPEFQASNIMHSINGYVFDSLQLSVCLHEVAYWYI LSIGAQTDFLSVFFSGYTFKHKMVYEDTLTLFPFSGETVFMSMENPGLWILGCHNSDFRNRGMTALLKVSS CDKNTGDYYEDSYEDISAYLLSKNNAIEPRgFgQ-VPPVLK Q^EITRTTLQSDQEEIDYDDTI SVEMKKE DFDIYDEDENQSPRSFQKKTRHYFIAAVERLWDYGMSSSPHVLRNRAQSGSVPQFKKWFQEFTDGSFTQP LYRGELNEHLGLLGPYIRAEVEDNIMVTFRNQASRPYSFYSSLISYEEDQRQGAEPRKNFVKPNETKTYFW KVQHHMAPTKDEFDCKAWAYFSDVDLEKDVHSGLIGPLLVCHTNTLNPAHGRQVTVQEFALFFTIFDETKS WYFTENMERNCRAPCNIQMEDPTFKENYRFHAINGYIMDTLPGLVMAQDQRIRWYLLSMGSNENIHSIHFS GHVFTVRKKEEYKMALYNLYPGVFETVEMLPSKAGIWRVECLIGEHLHAGMSTLFLVYSNKCQTPLGMASG HIRDFQITASGQYGQWAPKLARLHYSGSI AWSTKEPFSWIKVDLLAPMI IHGIKTQGARQKFSSLYI SQF I IMYSLDGKKWQTYRGNSTGTLMVFFGNVDSSGIKH IFNPPI IARYIRLHPTHYSIRSTLRMELMGCDLN SCSMPLGMESKAI SDAQITASSYFTNMFATWSPSKARLHLQGRSNAWRPQVNNPKEWLQVDFQKTMKVTGV TTQGVKSLLTSMYVKEFLISSSQDGHQWTLFFQNGKVKVFQGNQDSFTPWNSLDPPLLTRYLRIHPQSWV HQIALRMEVLGCEAQDLYDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVWDVSHEDPEV KFNWYVDGVEVHNAKTKPREEQYNSTYRWSVL LHQDWLNGBEYKCBVSNI^ALPAPIEKTISBCAKGQPR EPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFSCSV HEALHNHYTQKSLSLSPGK ii) Fc chain (20 amino acid heterologous signal peptide from mouse IgK chain underlined) ( SEP ID NP:4)
METDTLLLWVLLLWVPGSTG
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKP REEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSRDELTKNQ VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALH NHYTQKSLSLSPGK B. Full length FVIIIFc monomer hybrid (Full length FVIIIFc monomer dimer): created by coexpressing FVIIIFc and Fc chains.
Construct = HC-B-LC-Fc fusion. An Fc expression cassette is cotransfected with full length FVIII-Fc to generate the full length FVIIIFc monomer. For the FVIIIFc chain, the Fc sequence is shown in bold; HC sequence is shown in double underline; B domain sequence is shown in italics. Signal peptides are underlined.
i) Full length FVIIIFc chain (FVIII signal peptide underlined (SEP ID NO: 6)
MQIELSTCFFLCLLRFCFS
ATRRYYLGAVELSWDYMQSDLGELPVDARFPPRVPKSFPFNTSWYKKTLFVEFTDHLFNIAKPRPPWMGL LGPTIQAEVYDTVVITLKNMASHPVSLHAVGVSYWKASEGAEYDDQTSQREKEDDKVFPGGSHTYVWQVLK ENGPMASDPLCLTYSYLSHVDLVKDLNSGLIGALLVCREGSLAKEKTQTLHKFILLFAVFDEGKSWHSETK NSLMQDRDAASARAWPKMHTVNGYVNRSLPGLIGCHRKSVYWHVIGMGTTPEVHSIFLEGHTFLVRNHRQA SLEISPITFLTAQTLLMDLGQFLLFCHISSHQHDGMEAYVKVDSCPEEPQLRMKNNEEAEDYDDDLTDSEM DWRFDDDNSPSFIQIRSVAKKHPKTWVHYIAAEEEDWDYAPLVLAPDDRSYKSQYLNNGPQRIGRKYKKV RFMAYTDETFKTREAIQHESGILGPLLYGEVGDTLLI IFKNQASRPYNIYPHGITDVRPLYSRRLPKGVKH LKDFPILPGEI FKYKWTVTVEDGPTKSDPRCLTRYYSSFVNMERDLASGLIGPLLICYKESVDQRGNQIMS DKRNVILFSVFDENRSWYLTENIQRFLPNPAGVQLEDPEFQASNIMHSINGYVFDSLQLSVCLHEVAYWYI LSIGAQTDFLSVFFSGYTFKHKMVYEDTLTLFPFSGETVFMSMENPGLWILGCHNSDFRNRGMTALLKVSS COKNTGOYYEOSYEOISAY SKNNAIEPRSFSQNSRHPSTRQKQFNATTIPENDIEKTDPWFAHRTPMPK IQNVSSSDLLMLLRQSPTPHGLSLSDLQEAKYETFSDDPSPGAIDSNNSLSEMTHFRPQLHHSGDMVFTPE SGLQLRLNEKLGTTAATELKKLDFKVSSTSNNLISTIPSDNLAAGTDNTSSLGPPSMPVHYDSQLDTTLFG KKSSPLTESGGPLSLSEENNDSKLLESGLMNSQESSWGKNVSSTESGRLFKGKRAHGPALLTKDNALFKVS ISLLKTNKTSNNSATNRKTHIDGPSLLIENSPSVWQNILESDTEFKKVTPLIHDRMLMDKNATALRLNHMS NKTTSSKNMEMVQQKKEGPIPPDAQNPDMSFFKMLFLPESARWIQRTHGKNSLNSGQGPSPKQLVSLGPEK SVEGQNFLSEKNKWVGKGEFTKDVGLKEMVFPSSRNLFLTNLDNLHENNTHNQEKKIQEEIEKKETLIQE NVVLPQIHTVTGTKNFMKNLFLLSTRQNVEGSYDGAYAPVLQDFRSLNDSTNRTKKHTAHFSKKGEEENLE GLGNQTKQIVEKYACTTRISPNTSQQNFVTQRSKRALKQFRLPLEETELEKRIIVDDTSTQWSKNMKHLTP STLTQIDYNEKEKGAITQSPLSDCLTRSHSIPQANRSPLPIAKVSSFPSIRPIYLTRVLFQDNSSHLPAAS YRKKDSGVQESSHFLQGAKKNNLSLAILTLEMTGDQREVGSLGTSATNSVTYKKVENTVLPKPDLPKTSGK VELLPKVHIYQKDLFPTETSNGSPGHLDLVEGSLLQGTEGAIKWNEANRPGKVPFLRVATESSAKTPSKLL DPLAWDNHYGTQIPKEEWKSQEKSPEKTAFKKKDTILSLNACESNHAIAAINEGQNKPEIEVTWAKQGRTE
^LC5QiPPVLKRiiQ^EITRTTLQSDQEEI DYDDTI SVEMKKEDFDIYDEDENQSPRSFQKKTRHYFIAAVE RLWDYGMSSSPHVLRNRAQSGSVPQFKKVVFQEFTDGSFTQPLYRGELNEHLGLLGPYIRAEVEDNIMVTF RNQASRPYSFYSSLI SYEEDQRQGAEPRKNFVKPNETKTYFWKVQHHMAPTKDEFDCKAWAYFSDVDLEKD VHSGLIGPLLVCHTNTLNPAHGRQVTVQEFALFFTIFDETKSWYFTENMERNCRAPCNIQMEDPTFKENYR FHAINGYIMDTLPGLVMAQDQRIRWYLLSMGSNENIHSIHFSGHVFTVRKKEEYKMALYNLYPGVFETVEM LPSKAGIWRVECLIGEHLHAGMSTLFLVYSNKCQTPLGMASGHIRDFQITASGQYGQWAPKLARLHYSGSI NAWSTKEPFSWIKVDLLAPMI IHGIKTQGARQKFSSLYI SQFI IMYSLDGKKWQTYRGNSTGTLMVFFGNV DSSGIKHNI FNPPI IARYIRLHPTHYSIRSTLRMELMGCDLNSCSMPLGMESKAI SDAQITASSYFTNMFA TWSPSKARLHLQGRSNAWRPQVNNPKEWLQVDFQKTMKVTGVTTQGVKSLLTSMYVKEFLISSSQDGHQWT LFFQNGKVKVFQGNQDSFTPVVNSLDPPLLTRYLRIHPQSWVHQIALRMEVLGCEAQDLYDKTHTCPPCPA PELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKP EEQY STYRV VSVLTVLHQDWLNGKEYKCBVSNBCALPAPIEKTISBCAKGQPREPQVYTLPPSRDELTBNQVSLTCLVKGFY PSDIAVEWESNGQPEN YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSV HEALHNHYTQKSLSLS PGK if) Fc chain (20 amino acid heterologous signal peptide from mouse IgK chain underlined) ( SEP ID NO: 4) METDTLLLWVLLLWVPGSTG
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCWVDVSHEDPEVKFNWYVDGVEVHNAKTKP REEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSRDELTKNQ VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALH NHYTQKSLSLSPGK
Cited References
4. Aledort L. et al, J Intern Med.: 236:391-399 (1994)
5. Petrini P. et al., Am J Pediatr Hematol Oncol. 73:280-287 (1991).
6. Aznar J. et al, Haemophilia 6(3): \ 70-176 (2000).
7. Feldman B. et al, J Thromb Haemost. 4: 1228-1236 (2006).
8. Kreuz W. et al, Haemophilia 4:413-417 (1998).
9. Liesner R. et al., B JHaem. 92:973-978 (1996).
10. Ljung R., Haemophilia. 40:409-412 (1998).
11. Lofquist T, et al, J Intern Med 247:395-400 (1997).
12. Nilsson I, et al, B. J Int Med 252:25-32 (1992).
13. Risebrough N. et al, Haemophilia. 74:743-752 (2008).
14. Van Den Berg H. et al, Haemophilia 9 (Suppl. l):21-31 (2003).
15. Van Den Berg H. et al, Haematologica 89(6):645-650 (2004).
16. Molho P. et al, Haemophilia 60:23-32 (2000).
17. Coppola A. et al, Blood Transfus. 6(2): 4-1 1 (2008).
18. Geraghty S. et al, Haemophilia 72:75-81 (2006).
19. Hacker M. et al, Haemophilia 70:392-396 (2001).
20. Lillicrap D., Current Opinion in Hematology 77:393-397 (2010).
21. Dumont J.A. et al, BioDrugs 20(3): 151-60 (2006).
22. Dumont J.A. et al, "Monomeric Fc fusion technology: an approach to create long lasting clotting factors," in: Kontermann R., ed., Therapeutic Proteins - Strategies to Modulate Half-Life, Chapter 11, Wiley VCH publisher; in press.
3. Roopenian D.C. et al, Nat Rev Immunol. 70:715-25 (Epub 2007 Aug 17).
4. Lencer W.I. and Blumberg R.S., Trends Cell Biol. I5(l):5-9 (2005).
5. Huang C, Curr Opin Biotechnol. 20(6):692-9. (Epub 2009 Nov 4).
6. Schmidt S.R., Curr Opin Drug Discov Devel. 720:284-295 (2009).
7. Dumont J. et al, Blood. 116(21) Abstract 545 (2009).
8. Liu T. et al, J Thromb Haemost. 9(S2):561 (201 1).
9. Rosen S., Scand J Haematol Suppl. JJ(Suppl 40): 139-45 (1984).
0. Lee C.A. et al, Thromb Haemost. 520:1644-7 (Dec. 1999).
1. Mikaelsson M. and Oswaldsson U., Semin Thromb Hemost. 250:257-64 (June 2002). 2. Stroobants A. . et al, J Thromb Haemost. 9 (Suppl 2) (2011).
3. Lenting P.J. et al, J Thromb Haemost. 5: 1353-60 (2007).
4. Smith N.L. et al, Circulation 727: 1382-1392 (2010).

Claims

WHAT IS CLAIMED IS:
1. A method of administering Factor VIII to a human subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric polypeptide comprising a Factor VIII portion and a second portion at a dosing interval at least about one and one -half times longer than the dosing interval required for an equivalent dose of a polypeptide consisting of said Factor VIII portion.wherein said Factor VIII portion comprises processed Factor VIII, which has two chains, a first chain comprising a heavy chain and a second chain comprising a light chain, wherein said first chain and said second chain are associated by a metal bond.
2. The method of claim 1, wherein at least about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, or about 99% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
3. The method of claim 2, wherein about 100% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
4. The method of any one of claims 1 or 2, wherein said Factor VIII portion comprises single chain Factor VIII, which is a single chain.
5. The method of claim 4, wherein said single chain Factor VIII contains an intact intracellular processing site.
6. The method of claim 5, wherein said intracellular processing site corresponds to Arginine at residue 1648 of full-length Factor VIII (Arginine residue 1667 of amino acids 20-2351 of SEQ ID NO: 6).
7. The method of any one of claims 3-6, wherein at least about 1%, about 5%, about 10%, about 15%), about 20%), or about 25% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
8. The method of claim 7, wherein about 25% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 75% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
9. The method of claim 7, wherein about 20% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 80% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
10. The method of claim 7, wherein about 15% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 85% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
11. The method of claim 7, wherein about 10% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 90% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
12. The method of claim 7, wherein about 5% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 95% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
13. The method of claim 7, wherein about 1%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 99% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
14. The method of any one of claims 3-6, wherein at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, or about 99% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
15. The method of claim 14, wherein about 30% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 70% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
16. The method of claim 14, wherein about 40% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 60% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
17. The method of claim 14, wherein about 50% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 50% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
18. The method of claim 14, wherein about 60% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 40% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
19. The method of claim 14, wherein about 70% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 30% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
20. The method of claim 14, wherein about 80% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 20% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
21. The method of claim 14, wherein about 85% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 15% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
22. The method of claim 14, wherein about 90% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 10% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
23. The method of claim 14, wherein about 95% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 5% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
24. The method of claim 14, wherein about 99% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 1% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
25. The method of claim 14, wherein about 100% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
26. The method of any one of claims 4-25, wherein the chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity at a level comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions, when the Factor VIII activity is measured in vitro by a chromogenic assay.
27. The method of any one of claims 4-26, wherein said chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity in vivo comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
28. The method of any one of claims 4-27, wherein said chimeric polypeptide comprising single chain Factor VIII has a Factor Xa generation rate comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
29. The method of any one of claims 4-28, wherein said single chain Factor VIII in the chimeric polypeptide is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
30. The method of any one of claims 4-29, wherein said single chain Factor VIII in the chimeric polypeptide has a Factor IXa interaction rate comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
31. The method of any one of claims 4-30, wherein said single chain Factor VIII in the chimeric polypeptide binds to von Willebrand Factor at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
32. The method of any one of claims 1-31, wherein said chimeric polypeptide has one or more characteristics selected from the group consisting of:
an ability to interact with phospholipid vesicles comparable to the ability of a polypeptide consisting of said Factor VIII portion;
an ability to form a Xase complex that activates Factor X comparable to the ability of a polypeptide consisting of said Factor VIII portion;
an ability to be activated by alpha-thrombin within five minutes comparable to the ability of a polypeptide consisting of said Factor VIII portion; and
an ability to interact with Factor IXa comparable to the ability of a polypeptide consisting of said Factor VIII portion.
33. The method of claim 32, wherein said chimeric polypeptide has one or more characteristics selected from the group consisting of: forms a Xase complex that activates Factor X at a Km within about one, about one and one half, or about two standard deviations of the Km of a polypeptide consisting of said Factor VIII portion, wherein Km is measured as a function of Factor X concentration;
forms a Xase complex that activates Factor X at a Vmax within about one, about one and one half, or about two standard deviations of the Vmax of a polypeptide consisting of said Factor VIII portion, wherein Vmax is measured as a function of Factor X concentration;
forms a Xase complex that activates Factor X at a Kd within about one, about one and one half, or about two standard deviations of the Kd of a polypeptide consisting of said Factor VIII portion, wherein Kd is measured as a function of Factor IXa concentration; and
forms a Xase complex that activates Factor X at a Vmax within about one, about one and one half, or about two standard deviations of the Vmax of a polypeptide consisting of said Factor VIII portion, wherein Vmax is measured as a function of Factor IXa concentration.
34. The method of any of claims 1 -33, wherein said dose has a mean incremental recovery (K- Value) (activity; observed) greater that 1.38 IU/dL per IU/kg.
35. The method of any of claims 34, wherein said dose has a mean incremental recovery (K- Value) (activity; observed) of at least about 1.5, at least about 1.85, or at least about 2.46 IU/dL per IU/kg.
36. The method of any of claims 1 -35, wherein said chimeric polypeptide exhibits one or more pharmacokinetic parameters, in said patient population or in said subject, selected from the group consisting of:
a Cmax OBS in said subject administered with 25 IU/kg of the chimeric polypeptide is comparable to the Cmax OBS in a subject administered with 25 IU/kg of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay; a Cmax_OBS in said subject of about 60.5 IU/dL as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 53.1 - 69 IU/dL as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 119 IU/dL as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 103 - 136 IU/dL as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 76.5 IU/dL as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 64.9 - 90.1 IU/dL as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Cmax OBS in said subject of about 182 IU/dL as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered; a Cmax OBS in said subject of about 146 - 227 IU/dL as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a mean clearance (CL) (activity) in said patient population of about 2.33 ± 1.08 mL/hour/kg or less;
a mean clearance (CL) (activity) in said patient population of about 1.8-2.69 mL/hour/kg;
a mean clearance (CL) (activity) in said patient population that is about 65% of the clearance of a polypeptide consisting of said Factor VIII portion;
a clearance (CL) (activity) in said subject of about 1.22 - 5.19 mL/hour/kg;
a clearance (CL) (activity) in said subject is 0.51, 0.52, 0.53, 0.54, 0.55, 0.56, 0.57, 0.58, 0.59, 0.60, 0.61, 0.62, 0.63, 0.64, 0.65, 0.66, 0.67, 0.68, 0.69, or 0.70 times lower than the clearance in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a clearance (CL) (activity) in said subject of about 1.68 mL/hour/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.31 - 2.15 mL/hour/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 2.32 mL/hour/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.64 - 3.29 mL/hour/kg as measured by oa ne stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.49 mL/hour/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.16 - 1.92 mL/hour/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.52 mL/hour/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a clearance (CL) (activity) in said subject of about 1.05-2.20 mL/hour/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a mean mean residence time (MRT) (activity) in said patient population of at least about 26.3 ± 8.33 hours;
a mean MRT (activity) in said patient population of about 25.9 - 26.5 hours;
a mean MRT (activity) in said patent population that is about 1.5 fold longer than the mean MRT of a polypeptide consisting of said Factor VIII portion;
a mean residence time (MRT) (activity) in said subject of about 14 - 41.3 hours;
a MRT in said subject is at least 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79,
1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, or 1.90 times higher than the MRT in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a MRT (activity) in said subject of about 27 hours as meansured by a one stage (aPTT) assay; a MRT (activity) in said subject of about 20.6 - 35.3 hours as meansured by a one stage (aPTT) assay;
a MRT (activity) in said subject of about 23.9 - 28.5 hours as measured by a two stage (chromogenic) assay;
a MRT (activity) in said subject of about 19.8 - 28.9 hours as measured by a two stage (chromogenic) assay;
a MRT (activity) in said subject of about 20.5 - 39.6 hours as measured by a two stage (chromogenic) assay;
a mean tl/2beta (activity) in said patient population of about 18.3 ± 5.79 hours;
a mean tl/2beta (activity)in said patient population that is about 18 - 18.4 hours;
a mean tl/2beta (activity) in said patient population that is about 1.5 fold longer than the mean tl/2beta of a polypeptide consisting of said Factor VIII portion;
a tl/2beta (activity) in said subject of about 11 - 26.4 hours;
a tl/2beta (activity) in said subject that is at least 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, or 1.87 times higher than the tl/2beta (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 18.8 hours as measured by a one stage (aPTT) assay; a tl/2beta (activity) in said subject of about 14.3 - 24.5 hours as measured by a one stage (aPTT) assay;
a tl/2beta (activity) in said subject of about 16.7 hours as measured by a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 13.8 - 20.1 hours as measured by a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 19.8 hours as measured by a two stage (chromogenic) assay;
a tl/2beta (activity) in said subject of about 14.3 - 27.5 hours as measured by a two stage (chromogenic) assay;
a mean incremental recovery (K value) (activity; observed) in said patient population of about 2.01 ± 0.44 IU/dL per IU/kg; a mean incremental recovery (K value) (activity; observed) in said patient population of about 1.85 - 2.46 IU/dL per IU/kg;
a mean incremental recovery (K value) (activity; observed) in said patient population that is about 90 % of the mean incremental recovery of a polypeptide consisting of said Factor VIII portion;
an incremental recovery (K value) (activity; observed) in said subject of about 1.38 - 2.88 IU/dL per IU/kg;
an incremental recovery in said subject that is comparable to the Incremental Recovery in a subject administered with a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
an incremental recovery in said subject of about 2.44 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 2.12 - 2.81 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 1.83 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 1.59 - 2.10 IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 3.09 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 2.80 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an incremental recovery in said subject of about 2.61-3.66 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; and
an incremental recovery in said subject of about 2.24-3.50 IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a mean Vss (activity) in said patient population of about 55.1 ± 12.3 mL/kg;
a mean Vss (activity) in said patient population of about 45.3 - 56.1 mL/kg;
a Vss (activity) in said subject of about 37.7 - 79.4 mL/kg;
a Vss (activity) in said subject that is comparable to the Vss (activity) in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when measured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
a Vss (activity) in said subject of about 45.5 mL/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 39.3 - 52.5 mL/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered; a Vss (activity) in said subject of about 62.8 mL/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 55.2 - 71.5 mL/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 35.9 mL/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 30.4-42.3 mL/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 43.4 mL/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a Vss (activity) in said subject of about 38.2-49.2 mL/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
a mean AUC/dose (activity) in said patient population of about 49.9 ± 18.2 IU*h/dL per IU/kg; a mean AUC/dose (activity) in said patient population of about 44.8 - 57.6 IU*h/dL per IU/kg; and
an AUC/dose in said subject of about 19.2 - 81.7 IU*h/dL per IU/kg;
an AUCiNF in said subject that is at least 1.45 1.46, 1.47, 1.48, 1.49, 1.50, 1.51, 1.52, 1.53, 1.54, 1.55, 1.56, 1.57, 1.58, 1.59, 1.60, 1.61, 1.62, 1.63, 1.64, 1.65, 1.66, 1.67, 1.68, 1.69, 1.70, 1.71, 1.72, 1.73, 1.74, 1.75, 1.76, 1.77, 1.78, 1.79, 1.80, 1.81, 1.82, 1.83, 1.84, 1.85, 1.86, 1.87, 1.88, 1.89, 1.90 times higher than the AUCINF in a subject administered with the same amount of a polypeptide consisting of the full-length, mature Factor VIII when meansured by a one stage (aPTT) assay or a two stage (chromogenic) assay;
an AUCiNF in said subject of about 1440 ± 316 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1160 - 1880 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1480 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 2910 ± 1320 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1980 - 3970 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 2800 hr*IU/dL per IU/kg as measured by a one stage (aPTT) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 1660 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered; an AUCiNF in said subject of about 1300 - 2120 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 25 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 4280 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered;
an AUCiNF in said subject of about 2960 - 6190 hr*IU/dL per IU/kg as measured by a two stage (chromogenic) assay when about 65 IU/kg of the chimeric polypeptide is administered.
37. The method of any of claims 1-36, wherein the therapeutic dose is about 10 to about 150, 100-110, 110-120, 120-130, 130-140, 140-150, 110, 115, 120, 125, 130, 135, 140, 145, or 150 IU/kg.
38. The method of any of claims 1 -37, wherein the dosing interval is one and one -half to 5, one and one-half, 2, 3, 4, or 5 days or longer.
39. A composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 96%o, about 97%), about 98%, about 99%, or about 100%) of said polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion, wherein said single chain Factor VIII is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO: 6).
40. The composition of claim 39, wherein said single chain Factor VIII is identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
41. A composition comprising a chimeric polypeptide having Factor VIII activity, wherein at least about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, about 95%, about 96%), about 97%), about 98%, about 99%, or about 100%) of said polypeptide comprises a Factor VIII portion, which is single chain Factor VIII, and a second portion, wherein said single chain Factor VIII is at least 90%) or 95%) identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
42. The composition of claim 41, wherein said single chain Factor VIII is identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
43. The composition of any of claims 39 to 42, wherein said second portion is an Fc region, albumin, a PAS sequence, transferrin or CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or any combinations thereof.
44. The composition of any of claims 43, wherein said second portion is an Fc region.
45. The composition of claim 44, wherein said Fc is at least 90%) or 95%) identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6)
46. The composition of claim 45, wherein said Fc is identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
47. The composition of claim 39, wherein said polypeptide comprises a sequence at least 90% or 95%o identical to the Factor VIII and Fc amino acid sequences shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95%> identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
48. The composition of claim 39, wherein said polypeptide comprises a sequence identical to the Factor VIII and Fc amino acid sequences shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90%> or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
49. The composition of any of claims 39-48, wherein said polypeptide is in the form of a hybrid comprising a second polypeptide, wherein said second polypeptide consists essentially of an Fc.
50. The composition of claim 49, wherein said second polypeptide consists essentially of a sequence at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
51. The composition of claim 50, wherein said second polypeptide consists essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
52. The composition of any of claims 39-51, wherein said polypeptide has a half-life at least one and one -half to six times longer, one and one -half to five times longer, one and one -half to four times longer, one and one -half to three times longer, or one and one-half to two times longer to a polypeptide consisting of said Factor VIII.
53. The composition of any one of claims 39-52, which has one, about one and one half, or about two or more characteristics selected from the group consisting of:
an ability to interact with phospholipid vesicles comparable to the ability of a polypeptide consisting of said Factor VIII portion;
an ability to form a Xase complex that activates Factor X comparable to the ability of a polypeptide consisting of said Factor VIII portion;
an ability to be activated by alpha-thrombin within five minutes comparable to the ability of a polypeptide consisting of said Factor VIII portion; and
an ability to interact with Factor IXa comparable to the ability of a polypeptide consisting of said Factor VIII portion.
54. The composition of any one of claims 39-53, wherein about 70%, about 60%>, about 50%, about 40%, about 30%, about 20%, about 10%, about 5%, about 4%, about 3%, about 2%, or about 1% of the Factor VIII portion of said polypeptide is processed Factor VIII.
55. The composition of any one of claims 39-54, wherein said chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, wherein said processed Factor VIII is fused to one Fc of the two Fc portions, when the Factor VIII is measured in vitro by a chromogenic assay.
56. The composition of any one of claims 39-55, wherein said chimeric polypeptide comprising single chain Factor VIII has a Factor Xa generation rate comparable to processed Factor VIII.
57. The composition of any one of claims 39-56, wherein said chimeric polypeptide comprising single chain Factor VIII has a Factor Xa generation rate comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
58. The composition of any one of claims 39-57, wherein said single chain Factor VIII in the chimeric polypeptide has a Factor IXa interaction rate comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
59. The composition of any one of claims 39-58, wherein said single chain Factor VIII in the chimeric polypeptide is inactivated by activated Protein C at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
60. The composition of any one of claims 39-59, wherein said single chain Factor VIII in the chimeric polypeptide binds to von Willebrand Factor at a level comparable to processed Factor VIII in a chimeric polypeptide consisting of two Fc portions and processed Factor VIII.
61. The composition of any one of claims 39-60, wherein said chimeric polypeptide comprising single chain Factor VIII has Factor VIII activity in vivo comparable to a chimeric polypeptide consisting of two Fc portions and processed Factor VIII, which is fused to one of the two Fc portions.
62. The composition of any one of claims 39-61, wherein about 30%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 70%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
63. The composition of any one of claims 39-61, wherein about 40%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 60%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
64. The composition of any one of claims 39-61, wherein about 50%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 50%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
65. The composition of any one of claims 39-61, wherein about 60%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 40%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
66. The composition of any one of claims 39-61 , wherein about 70% of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 30% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
67. The composition of any one of claims 39-61 , wherein about 80%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 20%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
68. The composition of any one of claims 39-61 , wherein about 90%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 10%> of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
69. The composition of any one of claims 39-61 , wherein about 95%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 5% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
70. The composition of any one of claims 39-61 , wherein about 99%> of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII and about 5% of the Factor VIII portion of the chimeric polypeptide is processed Factor VIII.
71. The composition of any one of claims 39-61 , wherein about 100%) of the Factor VIII portion of the chimeric polypeptide is single chain Factor VIII.
72. A composition of any one of claims 39-71 , which is a pharmaceutical composition further comprising an excipient.
73. A method of treating a bleeding condition comprising administering a therapeutically effective amount of the composition of any one of claims 39-72.
74. The method of claim 73, wherein said treatment is prophylactic treatment.
75. The method of claim 73, wherein said treatment is on-demand treatment.
76. The method of any one of claims 73-75, wherein said bleeding condition is selected from the group consisting of a bleeding coagulation disorder, hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, and bleeding in the illiopsoas sheath.
77. The method of claim 76, wherein said bleeding coagulation disorder is hemophilia.
78. The method of claim 77, wherein said bleeding coagulation disorder is hemophilia A.
79. The method of any one of claims 1 to 38 and 73 to 78, wherein said administration maintains a Factor VIII trough level in at least 90%> of said patient population at a level above 1%>.
80. The method of any one of claims 1 to 38 and 73 to 78, wherein said administration maintains a Factor VIII trough level in at least 60%> of said population at a level above 3%>.
81. A method of maintaining Factor VIII in a patient population at a level above 1% for at least one day, two days, or three days comprising administering a therapeutically effective amount of rFVIIIFc to said population, wherein the therapeutically effective amount is at least 25 IU/kg of rFVIIIFc and wherein at least 90% of said population maintains the Factor VIII trough level at least 1% or more.
82. A method of maintaining Factor VIII in a patient population at a level above 3% for at least one day, two days, three days comprising administering a therapeutically effective amount of rFVIIIFc to said population, wherein the therapeutically effective amount is at least 65 IU/kg of rFVIIIFc and wherein at least 90% of said population maintains the Factor VIII trough level at least 3% or more.
83. The method of any one of claims 4 to 38 and 73 to 82, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645.
84. The method of any one of claims 4 to 38 and 73 to 83, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1648.
85. The method of any one of claims 4 to 38 and 73 to 84, wherein the single chain Factor VIII comprises a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
86. The method of claim 85, wherein the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid from the amino acid substituted at the amino acid position corresponding to Arginine 1648.
87. The method of any one of claims 83-86, wherein the substitution or mutation is an amino acid other than arginine.
88. The composition of any one of claims 39 to 72, wherein the single chain FVIII comprises an intact intracellular processing site.
89. The composition of any one of claims 39 to 72, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645 in full- length mature Factor VIII.
90. The composition of any one of claims 39 to 72, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1648 in full- length mature Factor VIII.
91. The composition of any one of claims 39 to 72, wherein the single chain Factor VIII comprises a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
92. The composition of claim 91, wherein the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid than the amino acid substituted at the amino acid position corresponding to Arginine 1648.
93. The composition of any one of claims 89 to 92, wherein the substitution or mutation is an amino acid other than arginine.
94. The method of any one of claims 73 to 82, wherein the single chain Factor VIII comprises an intact intracellular processing site.
95. The method of any one of claims 73 to 82, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1645 in full-length mature Factor VIII.
96. The method of any one of claims 73 to 82, wherein the single chain Factor VIII comprises a substitution or mutation at an amino acid position corresponding to Arginine 1648 in full-length mature Factor VIII.
97. The method of any one of claims 73 to 82, wherein the single chain Factor VIII comprises a substitution or mutation at amino acid positions corresponding to Arginine 1645 and Arginine 1648 in full-length Factor VIII.
98. The method of claim 97, wherein the amino acid substituted at the amino acid position corresponding to Arginine 1645 is a different amino acid than the amino acid substituted at the amino acid position corresponding to Arginine 1648 in full-length Factor VIII.
99. The method of any one of claims 95 to 98, wherein the substitution or mutation is an amino acid other than arginine.
100. The method of claim 87 or 99, wherein the amino acid residue other than arginine is selected from the group consisting of isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, histidine, ornithine, pyrrolysine, or taurine.
101. The composition of claim 93, wherein the amino acid residue other than arginine is selected from the group consisting of isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, valine, alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, selenocysteine, serine, tyrosine, histidine, ornithine, pyrrolysine, or taurine.
102. The method of any one of claims 1 to 38, 73 to 87, or 94 to 100, wherein said subject is a pediatric subject.
103. A method of preventing, decreasing, or treating a bleeding episode in a subject comprising administering to the subject an effective amount of a long-acting Factor VIII (FVIII) protein, wherein the subject expresses a high level of von Willebrand Factor (VWF) in plasma.
104. A method of claim 103, wherein the subject has been identified as expressing a high level of VWF in plasma.
105. A method of preventing, decreasing, or treating a bleeding episode in a subject comprising:
(a) identifying a subject having high levels of VWF by measuring the level of VWF in the plasma of said subject, wherein a VWF level of at least about 100 IU/dL identifies the subject as having a high level of VWF; and (b) administering to the subject an effective amount of a long-acting FVIII protein.
106. The method of any of claims 103-105, wherein the subject is a human.
107. The method of any of claims 103-106, wherein the subject is a pediatric subject.
108. The method of any of claims 103-107, wherein the subject has hemophilia A.
109. The method of any of claims 103-108, wherein the high level of VWF is at least about lOO IU/dL.
110. The method of any of claims 103-109, wherein the high level of VWF is between about 100 IU/dL and about 200 IU/dL.
111. The method of claim 110, wherein the high level of VWF is about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
112. The method of any of claims 103-111, wherein the subject has the blood serotype A, B, or
AB.
113. The method of any of claims 103-112, wherein said long-acting FVIII protein has a half- life in said subject of between about 20 and about 40 hours.
114. The method of claim 113, wherein said long-acting FVIII protein has a half-life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours.
115. The method of claim 114, wherein said long-acting FVIII protein has a half-life of between about 20 and 27 hours.
116. The method of any of claims 103-115, wherein said long-acting FVIII protein has a half- life that is at least about 1.2 times greater than the half- life of said said long-acting FVIII protein when administered to an individual having average levels of VWF.
117. The method of claim 116, wherein said long-acting FVIII protein has a half- life that is at least about about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold times greater than the half-life of said said long-acting FVIII protein when administered to an individual having average levels ofVWF.
118. The method of any one of claims 103-117, wherein the effective amount is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg.
119. The method of any of claims 103-118, wherein the effective amount is at least about 65IU/kg to at least about 90IU/kg.
120. The method of claim 119, wherein the effective amount is 80 IU/kg.
121. The method of any of claims 103-120, wherein the long-acting FVIII protein is administered every 72 hours or longer.
122. The method of claim 121, wherein the long-acting FVIII protein is administered about once a week or longer.
123. The method of claim 121, wherein the effective amount of the long-acting FVIII protein is administered about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
124. The method of claim 121, wherein the long-acting FVIII is administered at a dosage of 80 IU/kg once every 72 hours.
125. The method of claim 124, wherein the subject is a pediatric subject.
126. The method of any of claims 103-125, wherein the administration resolves greater than 5- 20%, greater than 5-15%, greater than 5-10%, greater than 10-20%), or greater than 10-15%) of bleeding episodes.
127. The method of any of claims 103-126, wherein a trough level of plasma Factor VIILC in the subjects is maintained above 1-3 or 3-5 IU/dl.
128. The method of any of claims 103-127, wherein the administration prevents a bleeding episode in the subject.
129. The method of claim 128, wherein the bleeding episode is spontaneous.
130. The method of claim 129, wherein the administration resolves greater than 80-100%o, greater than 80-90%>, greater than 85-90%>, greater than 90-100%o, greater than 90-95%>, or greater than 95- 100%) of bleeding episodes.
131. The method of any of claims 103-130, wherein the administration maintains homeostatis in the population of the subjects in need of a surgery.
132. The method of any of claims 103-131, wherein the long-acting FVIII protein is administered prior to, during, or after the surgery.
133. The method of claim 132, wherein the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
134. The method of claims 131-133, wherein the surgery is an emergency surgery.
135. The method of any of claims 103-134, wherein the long-acting FVIII protein has a half- life longer than a polypeptide consisting of FVIII.
136. The method of any of claims 103-135, wherein the long-acting FVIII protein is pegylated, hesylated, or polysialylated.
- I l l -
137. The method of any of claims 103-136, wherein the long-acting FVIII protein is a chimeric protein comprising a FVIII portion and a second portion.
138. The method of claim 137, wherein the second portion is an Fc region, albumin, a PAS sequence, transferrin or CTP (28 amino acid C-terminal peptide (CTP) of hCG with its 4 O-glycans), polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin binding polypeptide, albumin-binding small molecules, or any combinations thereof.
139. The method of claims 137 or 138, wherein the second portion is fused to the amino- terminus or the carboxy-terminus of the FVIII portion.
140. The method of claims 137 or 138, wherein the second portion is inserted between two amino acids in the FVIII portion.
141. The method of any of claims 137-140, wherein the chimeric protein is a FVIIIFc monomer dimer hybrid.
142. The method of claim 141, wherein the FVIII portion is a single chain.
143. The method of claim 142, wherein the FVIII portion comprises a heavy chain and a light chain.
144. The method of any of claims 137-143, wherein the FVIII portion comprises full-length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain.
145. The method of claim 141, wherein the FVIII portion comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6.
146. The method of claim 145, wherein the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6.
147. The method of any of claims 137-146, wherein the second portion of the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
148. The method of claim 147, wherein the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
149. The method of any one of claims 103-148, wherein the long-acting FVIII polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
150. A method of treating a subject diagnosed with bleeding disorder, comprising measuring the half- life of FVffl-Fc in said subject, wherein a half- life that is at least about 1.2 times greater than the half-life of FVIII-Fc in a normal subject indicates the subject is a candidate for long interval dosing, and administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days.
151. A method of treating a subject diagnosed with bleeding disorder, comprising administering a FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days to a subject, wherein the half-life of FVIII-Fc in said subject is at least about 1.2 times greater than the half- life of FVIII-Fc when administered to a subject having average levels of VWF.
152. The method of claims 150 or 151, wherein the plasma half-life of FVIII-Fc in said subject is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold times greater than the plasma half-life of FVIII-Fc when administered to a subject having average levels of VWF.
153. The method of any of claims 150-152, wherein said FVIII-Fc plasma half-life is between 20-40 hours.
154. The method of claim 153, wherein said long-acting FVIII protein has a half-life of about 21 hours, 22 hours, 23 hours, 24 hours, 25 hours, 26 hours, 27 hours, 28 hours, 29 hours, 30 hours, 31 hours, 31 hours, 32 hours, 33 hours, 34 hours, 35 hours, 36 hours, 37 hours, 38 hours, 39 hours, or 40 hours.
155. The method of claim 154, wherein said long-acting FVIII protein has a half-life of between about 20 and 27 hours.
156. A method of treating a subject diagnosed with bleeding disorder, comprising measuring the half- life of a short-acting FVIII administered to said subject, wherein a half- life that is at least about 1.2 times greater than the half-life of said short-acting FVIII in a subject having average VWF levels indicates that the subject is a candidate for long interval dosing, and administering a long-acting FVIII-Fc polypeptide in an effective amount and at a dosing interval of at least 3 days.
157. The method of claim 156, wherein the half-life of the short-acting FVIII in said subject is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold greater than the half-life of a short-acting FVIII when administered to a subject having average levels of VWF.
158. The method of any of claims 150-157, wherein the subject is a human.
159. The method of any of claims 150-158, wherein the subject is a pediatric subject.
160. The method of any of claims 150-159, wherein the subject has hemophilia A.
161. The method of any of claims 150-160, wherein the subject has the blood serotype A, B, or
AB.
162. The method of any of claims 150-161, wherein the long-acting FVIII-Fc is administered in an effective amount that is at least about 20IU/kg, at least about 25 IU/kg, at least about 30IU/kg, at least about 35 IU/kg, at least about 40IU/kg, at least about 45 IU/kg, at least about 50IU/kg, at least about 55 IU/kg, at least about 60IU/kg, at least about 65IU/kg, at least about 70IU/kg, at least about 75IU/kg, at least about 80IU/kg, at least about 85IU/kg, or at least about 90IU/kg.
163. The method of any of claims 150-161, wherein the effective amount is at least about 65IU/kg to at least about 90IU/kg.
164. The method of any of claims 150-163, wherein the effective amount of the FVIII-Fc protein is administered about once every week, about once every 10 days, about once every two weeks, about once every 15 days, about once every 20 days, about once every three weeks, about once every 25 days, about once every four weeks, or about once every one month.
165. The method of any of claims 150-164, wherein the administration resolves greater than 5- 20%, greater than 5-15%, greater than 5-10%, greater than 10-20%), or greater than 10-15%) of bleeding episodes.
166. The method of any of claims 150-165, wherein a trough level of plasma Factor VIILC in the subjects is maintained above 1-3 or 3-5 IU/dl.
167. The method of any of claims 150-166, wherein the administration prevents a bleeding episode in the subject.
168. The method of claim 167, wherein the bleeding episode is spontaneous.
169. The method of claim 167, wherein the administration resolves greater than 80-100%), greater than 80-90%), greater than 85-90%), greater than 90-100%), greater than 90-95%), or greater than 95- 100%) of bleeding episodes.
170. The method of any of claims 150-169, wherein the administration maintains homeostatis in the population of the subjects in need of a surgery.
171. The method of any of claims 150-170, wherein the FVIII-Fc protein is administered prior to, during, or after the surgery.
172. The method of claims 170 or 171, wherein the surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
173. The method of any of claims 170-172, wherein the surgery is an emergency surgery.
174. The method of any of claims 150-173, wherein the FVIII-Fc protein has a half-life longer than a polypeptide consisting of FVIII.
175. The method of any of claims 150-174, wherein the FVIII-Fc protein is pegylated, hesylated, or polysialylated.
176. The method of claims 174 or 175, wherein the FVIII-Fc protein is a FVIIIFc monomer dimer hybrid.
177. The method of any of claims 172-176, wherein the FVIII portion is a single chain.
178. The method of claim 177, wherein the FVIII portion comprises a heavy chain and a light chain.
179. The method of any 174-178, wherein the FVIII portion comprises full-length factor VIII, mature factor VIII, or factor VIII with a full or partial deletion of the B domain.
180. The method of claim 179, wherein the FVIII portion comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6.
181. The method of claim 180, wherein the FVIII portion comprises amino acids 1 to 1438 of SEQ ID NO: 2 or amino acids 1 to 2332 of SEQ ID NO: 6.
182. The method of 174-181, wherein the second portion of the chimeric polypeptide comprises an Fc region which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
183. The method of claim 182, wherein the second portion comprises amino acids 1439 to 1665 of SEQ ID NO: 2 or amino acids 2333 to 2559 of SEQ ID NO: 6.
184. The method of any of claims 150-183, wherein the FVIII-Fc polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
185. A method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing with a long-acting FVIII polypeptide, comprising measuring the expression levels of plasma VWF, wherein an VWF expression level of at least 100 IU/dL indicates that the subject is a candidate for long interval dosing using a long-acting FVIII polypeptide.
186. The method of claim 185, wherein said VWF expression level is at least about 110 IU/dL, about 120 IU/dL, about 130 IU/dL, about 140 IU/dL, about 150 IU/dL, about 160 IU/dL, about 170 IU/dL, about 180 IU/dL, about 190 IU/dL, or about 200 IU/dL.
187. A method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half-life of FVIII-Fc in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of FVIII-Fc when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing.
188. The method of claim 187, wherein the half-life of FVIII-Fc is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold greater than the half-life of FVIII-Fc when administered to a subject having average levels of VWF.
189. A method for determining whether a subject diagnosed with bleeding disorder is a candidate for long interval dosing of a long-acting FVIII polypeptide, comprising measuring the half-life of short-acting FVIII in said subject, wherein a half-life that is at least about 1.2-fold greater than the half-life of short-acting FVIII when administered to a subject having average VWF levels indicates the subject is a candidate for long interval dosing.
190. The method of claim 189, wherein the half-life is at least about 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, or 2.5-fold greater than the half-life of FVIII-Fc when administered to a subject having average levels of VWF.
191. The method of any of claims 1 -38, 73-87, 94-100, and 102-190, wherein said chimeric polypeptide comprises a Fc portion.
192. The method of claim 191, where said dosing interval is at least about one and one-half to six times longer, one and one -half to five times longer, one and one-half to four times longer, one and one- half to three times longer, or one and one -half to two times longer, than the dosing interval required for an equivalent dose of a polypeptide consisting of said Factor VIII portion.
193. The method of claim 192, where said dosing interval is at least about one and one-half, two, two and one-half, three, three and one-half, four, four and one -half, five, five and one-half or six times longer than the dosing interval required for an equivalent dose of a polypeptide consisting of said Factor VIII portion.
194. The method of any of claims 1-38, 73-87, 94-100, and 102-193, wherein said dosing interval of said chimeric polypeptide is about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
195. The method of any of claims 1-38, 73-87, 94-100, and 102-194, wherein said subject is in need of prophylactic treatment.
196. The method of any of claims 1-38, 73-87, 94-100, and 102-193, wherein said subject is in need of on-demand treatment.
197. The method of claim 196, wherein said subject is in need of treatment for a bleeding episode.
198. The method of claim 197, wherein said subject is in need of treatment for hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath.
199. The method of claim 196, wherein said subject is in need of surgical prophylaxis, perioperative management, or treatment for surgery.
200. The method of claim 199, wherein said surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
201. The method of any of claims 196-200, wherein said dosing interval of said chimeric polypeptide is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
202. The method of any of claims 1-38, 73-87, 94-100, and 102-201, wherein said therapeutic dose is 10-100 IU/kg.
203. The method of claim 201, wherein said therapeutic dose is 10-20, 20-30, 30-40, 40-50, 50- 60, 60-70, 70-80, 80-90, or 90-100 IU/kg.
204. The method of claim 202, wherein said therapeutic dose is 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
205. The method of any of claims 1 -38, 73-87, 94-100, and 102-204, wherein said Factor VIII is human Factor VIII.
206. The method of any of claims 1 -38, 73-87, 94-100, and 102-205, wherein said Factor VIII has a full or partial deletion of the B domain.
207. The method of claim 205, wherein the Factor VIII portion of said chimeric polypeptide is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2; amino acids 20 to 2351 of SEQ ID NO: 6).
208. The method of claim 207, wherein the Factor VIII portion of said chimeric polypeptide is identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
209. The method of claim 205, wherein the Factor VIII portion of said chimeric polypeptide is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
210. The method of claim 209, wherein the Factor VIII portion of said chimeric polypeptide is identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
211. The method of claim 205 or 206, wherein said second portion of said chimeric polypeptide is at least 90% or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2; or amino acids 2352 to 2578 of SEQ ID NO:6).
212. The method of claim 211, wherein said second portion of said chimeric polypeptide is identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
213. The method of any of claims 1-38, 73-87, 94-100, and 102-212, wherein said chimeric polypeptide is in the form of a hybrid comprising a second polypeptide in association with said chimeric polypeptide, wherein said second polypeptide consists essentially of an Fc.
214. The method of claim 213, wherein said chimeric polypeptide comprises a sequence at least 90%) or 95%) identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95%) identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids lto 1684 of SEQ ID NO:2).
215. The method of claim 214, wherein said chimeric polypeptide comprises a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
216. The method of any of claims 213-215, wherein said second polypeptide consists essentially of a sequence at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90% or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
217. The method of claim 216, wherein said second polypeptide consists essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
218. The method of any of claims 1 -38, 73-87, 94-100, and 102-217, wherein said chimeric polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
219. A method of administering Factor VIII to a human subject in need thereof, comprising administering to the subject a therapeutic dose of a chimeric polypeptide comprising a Factor VIII portion and a second portion to obtain an area under the plasma concentration versus time curve (AUC) at least about one and one-quarter times greater than the AUC obtained by an equivalent dose of a polypeptide consisting of said Factor VIII portion.
220. The method of claim 219, wherein said chimeric polypeptide is administered at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
221. The method of claim 219 or 220, wherein said subject is in need of prophylactic treatment.
222. The method of claim 219 or 220, wherein said subject is in need of on-demand treatment.
223. The method of any of claims 219-222, wherein said subject is in need of treatment for a bleeding episode.
224. The method of any of claim 219-222, wherein said subject is in need of treatment for hemarthrosis, muscle bleed, oral bleed, hemorrhage, hemorrhage into muscles, oral hemorrhage, trauma, trauma capitis, gastrointestinal bleeding, intracranial hemorrhage, intra-abdominal hemorrhage, intrathoracic hemorrhage, bone fracture, central nervous system bleeding, bleeding in the retropharyngeal space, bleeding in the retroperitoneal space, or bleeding in the illiopsoas sheath.
225. The method of claim 222, wherein said subject is in need of surgical prophylaxis, perioperative management, or treatment for surgery.
226. The method of claim 225, wherein said surgery is minor surgery, major surgery, tooth extraction, tonsillectomy, inguinal herniotomy, synovectomy, total knee replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery, or joint replacement surgery.
227. The method of any of claims 219-226, wherein said dosing interval of said chimeric polypeptide is about once every 24-36, 24-48, 24-72, 24-96, 24-120, 24-144, 24-168, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, or 72 hours or longer.
228. The method of any of claims 219-227, wherein said therapeutic dose is 10-100 IU/kg.
229. The method of claim 228, wherein said therapeutic dose is 10-20, 20-30, 30-40, 40-50, 50- 60, 60-70, 70-80, 80-90, or 90-100 IU/kg.
230. The method of claim 229, wherein said therapeutic dose is 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
231. The method of any of claims 219-230, wherein said Factor VIII is human Factor VIII.
232. The method of any of claims 219-231, wherein said Factor VIII has a full or partial deletion of the B domain.
233. The method of claim 231, wherein the Factor VIII portion of said chimeric polypeptide is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO: 6).
234. The method of claim 233, wherein the Factor VIII portion of said chimeric polypeptide is identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
235. The method of claim 231, wherein the Factor VIII portion of said chimeric polypeptide is at least 90%) or 95% identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
236. The method of claim 235, wherein the Factor VIII portion of said chimeric polypeptide is identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
237. The method of claim 231 or 232, wherein said second portion of said chimeric polypeptide is at least 90% or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
238. The method of claim 237, wherein said second portion of said chimeric polypeptide is identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO: or; amino acids 2352 to 2578 of SEQ ID NO:6).
239. The method of any of claims 219-238, wherein said chimeric polypeptide is in the form of a hybrid comprising a second polypeptide in association with said chimeric polypeptide, wherein said second polypeptide consists essentially of an Fc.
240. The method of claim 239, wherein said chimeric polypeptide comprises a sequence at least 90%) or 95%) identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95%) identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
241. The method of claim 240, wherein said chimeric polypeptide comprises a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
242. The method of any of claims 239-241, wherein said second polypeptide consists essentially of a sequence at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90% or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
243. The method of claim 242, wherein said second polypeptide consists essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90% or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
244. The method of any of claims 219-243, wherein said chimeric polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
245. A method of administering Factor VIII to a human subject in need thereof, comprising administering to the subject a therapeutic dose of a polypeptide comprising a Factor VIII and an
Fc at a dosing interval of about every three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, or fourteen days or longer.
246. The method of any of claim 245, wherein said subject is in need of prophylactic treatment.
247. The method of claim 245 or 246, wherein said therapeutic dose is 10-100 IU/kg.
248. The method of claim 247, wherein said therapeutic dose is 10-20, 20-30, 30-40, 40-50, 50- 60, 60-70, 70-80, 80-90, or 90-100 IU/kg.
249. The method of claim 248, wherein said therapeutic dose is 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 IU/kg.
250. The method of any of claims 245-249, wherein said Factor VIII is human Factor VIII.
251. The method of any of claims 245-250, wherein said Factor VIII has a full or partial deletion of the B domain.
252. The method of claim 250, wherein said Factor VIII is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO:6).
253. The method of claim 252, wherein said Factor VIII is identical to a Factor VIII amino acid sequence shown in Table 2 without a signal sequence (amino acids 20 to 1457 of SEQ ID NO:2 or amino acids 20 to 2351 of SEQ ID NO : 6) .
254. The method of claim 250, wherein said Factor VIII is at least 90% or 95% identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
255. The method of claim 254, wherein said Factor VIII is identical to a Factor VIII amino acid sequence shown in Table 2 with a signal sequence (amino acids 1 to 1457 of SEQ ID NO:2 or amino acids 1 to 2351 of SEQ ID NO:6).
256. The method of claim 250 or 251, wherein said second portion is at least 90% or 95% identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO:6).
257. The method of claim 256, wherein said second portion is identical to the Fc amino acid sequence shown in Table 2 (amino acids 1458 to 1684 of SEQ ID NO:2 or amino acids 2352 to 2578 of SEQ ID NO: 6).
258. The method of any of claims 245-257, wherein said chimeric polypeptide is a Factor VIII-Fc chimeric polypeptide in the form of a hybrid comprising a second polypeptide in association with said chimeric polypeptide, wherein said second polypeptide consists essentially of an Fc.
259. The method of claim 258, wherein said chimeric polypeptide comprises a sequence at least 90% or 95%) identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90%> or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
260. The method of claim 259, wherein said chimeric polypeptide comprises a sequence identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) without a signal sequence (amino acids 20 to 1684 of SEQ ID NO:2) or at least 90% or 95% identical to the Factor VIII and Fc amino acid sequence shown in Table 2A(i) with a signal sequence (amino acids 1 to 1684 of SEQ ID NO:2).
261. The method of any of claims 258-260, wherein said second polypeptide consists essentially of a sequence at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
262. The method of claim 261, wherein said second polypeptide consists essentially of a sequence identical to the amino acid sequence shown in Table 2A(ii) without a signal sequence (amino acids 21 to 247 of SEQ ID NO:4) or at least 90%> or 95% identical to the amino acid sequence shown in Table 2A(ii) with a signal sequence (amino acids 1 to 247 of SEQ ID NO:4).
263. The method of any of claims 1-38, 73-87, 94-100, and 102-262, wherein said polypeptide is administered as part of a pharmaceutical composition comprising at least one excipient.
264. The method of any one of claims 1-38, 73-87, 94-100, and 102-263, wherein said chimeric polypeptide has Factor VIII activity.
PCT/US2012/045784 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof WO2013009627A2 (en)

Priority Applications (30)

Application Number Priority Date Filing Date Title
EP12811808.0A EP2729161B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
JP2014519074A JP2014522838A (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides and methods of use thereof
EA201490039A EA029045B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
PL12811808T PL2729161T3 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
CN201280043194.2A CN103796670A (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
SI201231567T SI2729161T1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
KR1020187003966A KR102110736B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
AU2012282875A AU2012282875B2 (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
RS20190355A RS58578B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
KR1020147003130A KR101829603B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
US14/131,600 US10010622B2 (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
BR112014000466A BR112014000466A2 (en) 2011-07-08 2012-07-06 chimeric and hybrid factor viii polypeptides, methods of using them
ES12811808T ES2722209T3 (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
EP18211156.7A EP3513804B1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
CA2841066A CA2841066C (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
PL18211156T PL3513804T3 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
NZ619438A NZ619438B2 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
DK12811808.0T DK2729161T3 (en) 2011-07-08 2012-07-06 FACTOR VIII CHEMICAL AND HYBRID POLYPEPTIDES AND PROCEDURES FOR USE THEREOF
MYPI2014000038A MY180714A (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
MX2014000202A MX350581B (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof.
EP22163636.8A EP4169525A1 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
LTEP12811808.0T LT2729161T (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
IL230333A IL230333B (en) 2011-07-08 2014-01-06 Factor viii chimeric and hybrid polypeptides, and methods of uses thereof
AU2016204986A AU2016204986B2 (en) 2011-07-08 2016-07-15 Factor VIII Chimeric And Hybrid Polypeptides, And Methods Of Use Thereof
AU2018202936A AU2018202936B2 (en) 2011-07-08 2018-04-27 Factor VIII Chimeric and Hybrid Polypeptides, and Methods of Use Thereof
US15/991,629 US10881742B2 (en) 2011-07-08 2018-05-29 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
HRP20190535TT HRP20190535T1 (en) 2011-07-08 2019-03-19 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
CY20191100325T CY1122900T1 (en) 2011-07-08 2019-03-19 CHIMERIC AND HYBRID FACTOR VIII POLYPEPTIDES AND METHODS OF USING THEREOF
IL271870A IL271870B (en) 2011-07-08 2020-01-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
US17/112,280 US20210220476A1 (en) 2011-07-08 2020-12-04 Factor viii chimeric and hybrid polypeptides, and methods of use thereof

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
US201161506015P 2011-07-08 2011-07-08
US61/506,015 2011-07-08
US201161522647P 2011-08-11 2011-08-11
US61/522,647 2011-08-11
US201161541561P 2011-09-30 2011-09-30
US61/541,561 2011-09-30
US201161569158P 2011-12-09 2011-12-09
US61/569,158 2011-12-09
US201261586443P 2012-01-13 2012-01-13
US61/586,443 2012-01-13
US201261622789P 2012-04-11 2012-04-11
US61/622,789 2012-04-11
US201261657641P 2012-06-08 2012-06-08
US61/657,641 2012-06-08

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14/131,600 A-371-Of-International US10010622B2 (en) 2011-07-08 2012-07-06 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof
US15/991,629 Division US10881742B2 (en) 2011-07-08 2018-05-29 Factor VIII chimeric and hybrid polypeptides, and methods of use thereof

Publications (2)

Publication Number Publication Date
WO2013009627A2 true WO2013009627A2 (en) 2013-01-17
WO2013009627A3 WO2013009627A3 (en) 2013-03-07

Family

ID=47506823

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/045784 WO2013009627A2 (en) 2011-07-08 2012-07-06 Factor viii chimeric and hybrid polypeptides, and methods of use thereof

Country Status (27)

Country Link
US (3) US10010622B2 (en)
EP (3) EP4169525A1 (en)
JP (5) JP2014522838A (en)
KR (2) KR101829603B1 (en)
CN (2) CN103796670A (en)
AR (1) AR087091A1 (en)
AU (3) AU2012282875B2 (en)
BR (1) BR112014000466A2 (en)
CA (1) CA2841066C (en)
CL (1) CL2014000024A1 (en)
CO (1) CO6940381A2 (en)
CY (1) CY1122900T1 (en)
DK (2) DK3513804T3 (en)
EA (1) EA029045B1 (en)
ES (2) ES2722209T3 (en)
HR (2) HRP20220639T1 (en)
HU (1) HUE043763T2 (en)
IL (2) IL230333B (en)
LT (2) LT2729161T (en)
MX (1) MX350581B (en)
MY (1) MY180714A (en)
PL (2) PL3513804T3 (en)
PT (2) PT2729161T (en)
RS (2) RS63241B1 (en)
SI (2) SI2729161T1 (en)
TW (3) TWI687226B (en)
WO (1) WO2013009627A2 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637640B2 (en) 2009-07-27 2014-01-28 Baxter International Inc. Blood coagulation protein conjugates
US8637007B2 (en) 2006-12-15 2014-01-28 Baxter International Inc. Factor VIIa-polysialic acid conjugate having prolonged in vivo half-life
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
WO2014144795A1 (en) * 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Factor viii polypeptide formulations
US8945897B2 (en) 2010-07-26 2015-02-03 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
EP2914293A4 (en) * 2012-10-30 2016-04-20 Biogen Ma Inc Methods of using fviii polypeptide
EP2956477A4 (en) * 2013-02-15 2016-12-28 Biogen Ma Inc Optimized factor viii gene
US9795683B2 (en) 2009-07-27 2017-10-24 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
EP2870250B1 (en) 2012-07-06 2018-04-18 Bioverativ Therapeutics Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
US10202595B2 (en) 2012-06-08 2019-02-12 Bioverativ Therapeutics Inc. Chimeric clotting factors
US10287564B2 (en) 2012-06-08 2019-05-14 Bioverativ Therapeutics Inc. Procoagulant compounds
US10317376B2 (en) 2012-04-11 2019-06-11 Bioverativ Therapeutics Inc. Methods of detecting glycosaminoglycans
US10350301B2 (en) 2009-07-27 2019-07-16 Baxalta Incorporated Blood coagulation protein conjugates
US11168124B2 (en) 2015-10-30 2021-11-09 Spark Therapeutics, Inc. CpG reduced factor VIII variants, compositions and methods and uses for treatment of hemostasis disorders
US11266720B2 (en) 2009-12-06 2022-03-08 Bioverativ Therapeutics Inc. Factor VIII-FC chimeric and hybrid polypeptides, and methods of use thereof
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes

Families Citing this family (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE497783T1 (en) * 2003-05-06 2011-02-15 Syntonix Pharmaceuticals Inc CLOTTING FACTOR VII-FC CHIMERIC PROTEINS FOR THE TREATMENT OF HEMOSTATIC DISEASES
TWI687226B (en) * 2011-07-08 2020-03-11 美商百歐維拉提夫治療公司 Factor viii chimeric and hybrid polypeptides, and methods of use thereof
US10078811B2 (en) 2013-11-29 2018-09-18 Fedex Corporate Services, Inc. Determining node location based on context data in a wireless node network
US10453023B2 (en) 2014-05-28 2019-10-22 Fedex Corporate Services, Inc. Methods and node apparatus for adaptive node communication within a wireless node network
US11238397B2 (en) 2015-02-09 2022-02-01 Fedex Corporate Services, Inc. Methods, apparatus, and systems for generating a corrective pickup notification for a shipped item using a mobile master node
US9985839B2 (en) 2015-07-08 2018-05-29 Fedex Corporate Services, Inc. Systems, apparatus, and methods of event monitoring for an event candidate within a wireless node network based upon sighting events, sporadic events, and benchmark checkpoint events
EP3433809A4 (en) 2016-03-23 2019-10-02 Fedex Corporate Services, Inc. Systems, apparatus, and methods for self-adjusting a broadcast setting of a node in a wireless node network
CN106279437B (en) 2016-08-19 2017-10-31 安源医药科技(上海)有限公司 Hyperglycosylated human coagulation factor VIII fusion proteins and preparation method thereof and purposes
CN107759697B (en) 2016-08-19 2023-03-24 安源医药科技(上海)有限公司 Method for producing fusion protein
EP3502143A4 (en) 2016-08-19 2020-07-15 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
JP7370974B2 (en) 2017-11-07 2023-10-30 ラニ セラピューティクス, エルエルシー Clotting factor preparations for delivery to intestinal tissues using a swallowable drug delivery device
WO2019175384A2 (en) 2018-03-16 2019-09-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from urocortin 3 and uses thereof for the diagnosis and treatment of type 1 diabetes
EP3765065A2 (en) 2018-03-16 2021-01-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from secretogranin v and uses thereof for the diagnosis and treatment of type 1 diabetes
EP3765064A1 (en) 2018-03-16 2021-01-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antigenic peptides deriving from pcsk2 and uses thereof for the diagnosis and treatment of type 1 diabetes
JP2021519089A (en) * 2018-03-28 2021-08-10 オリオニス バイオサイエンシーズ,インコーポレイテッド Bifunctional protein and its preparation
EP3793588A1 (en) * 2018-05-18 2021-03-24 Bioverativ Therapeutics Inc. Methods of treating hemophilia a
AU2020270420A1 (en) * 2019-05-03 2021-11-18 Rani Therapeutics, Llc Clotting factor preparations for delivery into tissue of the intestinal tract using a swallowable drug delivery device
MX2021015897A (en) 2019-06-19 2022-04-18 Bioverativ Therapeutics Inc Recombinant factor viii-fc for treating hemophilia and low bone mineral density.

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7348004B2 (en) 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
US7404956B2 (en) 2003-05-06 2008-07-29 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4757006A (en) 1983-10-28 1988-07-12 Genetics Institute, Inc. Human factor VIII:C gene and recombinant methods for production
DE3572982D1 (en) 1984-03-06 1989-10-19 Takeda Chemical Industries Ltd Chemically modified lymphokine and production thereof
US4965199A (en) 1984-04-20 1990-10-23 Genentech, Inc. Preparation of functional human factor VIII in mammalian cells using methotrexate based selection
EP0218712B1 (en) 1985-04-12 1992-02-26 Genetics Institute, Inc. Novel procoagulant proteins
KR910006424B1 (en) 1985-08-21 1991-08-24 인코텍스 비.브이 Method of manufacturing knitted briefs
JP2525022B2 (en) 1986-01-03 1996-08-14 ジェネティックス・インスチチュ−ト・インコ−ポレ−テッド VIII: Improved production method for c-factor protein
US5595886A (en) 1986-01-27 1997-01-21 Chiron Corporation Protein complexes having Factor VIII:C activity and production thereof
US5610278A (en) 1986-06-24 1997-03-11 Novo Nordisk A/S Process for producing a coagulation active complex of factor VIII fragments
US4912040A (en) 1986-11-14 1990-03-27 Genetics Institute, Inc. Eucaryotic expression system
CA1331157C (en) 1987-04-06 1994-08-02 Randal J. Kaufman Method for producing factor viii:c-type proteins
US6060447A (en) 1987-05-19 2000-05-09 Chiron Corporation Protein complexes having Factor VIII:C activity and production thereof
US6346513B1 (en) 1987-06-12 2002-02-12 Baxter Trading Gmbh Proteins with factor VIII activity: process for their preparation using genetically-engineered cells and pharmaceutical compositions containing them
IL86693A (en) 1987-06-12 1994-06-24 Stichting Centraal Lab Proteins with factor VIII activity, process for their preparation using genetically engineered cells and pharmaceutical compositions containing them
DE3720246A1 (en) 1987-06-19 1988-12-29 Behringwerke Ag FACTOR VIII: C-LIKE MOLECULE WITH COAGULATION ACTIVITY
FR2619314B1 (en) 1987-08-11 1990-06-15 Transgene Sa FACTOR VIII ANALOG, PREPARATION METHOD AND PHARMACEUTICAL COMPOSITION CONTAINING THE SAME
US4994371A (en) 1987-08-28 1991-02-19 Davie Earl W DNA preparation of Christmas factor and use of DNA sequences
US5004803A (en) 1988-11-14 1991-04-02 Genetics Institute, Inc. Production of procoagulant proteins
JP2989002B2 (en) 1988-12-22 1999-12-13 キリン―アムジエン・インコーポレーテツド Chemically modified granulocyte colony stimulating factor
ATE148171T1 (en) 1989-02-21 1997-02-15 Univ Washington MODIFIED FORMS OF REPRODUCTIVE HORMONES
SE465222C5 (en) 1989-12-15 1998-02-10 Pharmacia & Upjohn Ab A recombinant human factor VIII derivative and process for its preparation
US6552170B1 (en) 1990-04-06 2003-04-22 Amgen Inc. PEGylation reagents and compounds formed therewith
EP0575545B1 (en) 1991-03-15 2003-05-21 Amgen Inc. Pegylation of polypeptides
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
US6376463B1 (en) 1992-04-07 2002-04-23 Emory University Modified factor VIII
US5859204A (en) 1992-04-07 1999-01-12 Emory University Modified factor VIII
US5364771A (en) 1992-04-07 1994-11-15 Emory University Hybrid human/porcine factor VIII
US5563045A (en) * 1992-11-13 1996-10-08 Genetics Institute, Inc. Chimeric procoagulant proteins
SE504074C2 (en) 1993-07-05 1996-11-04 Pharmacia Ab Protein preparation for subcutaneous, intramuscular or intradermal administration
SE9503380D0 (en) 1995-09-29 1995-09-29 Pharmacia Ab Protein derivatives
US6458563B1 (en) 1996-06-26 2002-10-01 Emory University Modified factor VIII
CA2225189C (en) 1997-03-06 2010-05-25 Queen's University At Kingston Canine factor viii gene, protein and methods of use
US6200560B1 (en) 1998-10-20 2001-03-13 Avigen, Inc. Adeno-associated virus vectors for expression of factor VIII by target cells
DK1252192T3 (en) 2000-02-11 2006-11-20 Merck Patent Gmbh Enhancement of the serum half-life of antibody-based fusion proteins
DE60230456D1 (en) * 2001-10-05 2009-01-29 Expression Therapeutics Llc FACTOR VIII POLYPEPTIDES AT HIGH EXPRESSION LEVEL AND METHOD OF USE
ES2545090T3 (en) 2001-12-21 2015-09-08 Human Genome Sciences, Inc. Albumin and GCSF fusion proteins
US7041635B2 (en) 2003-01-28 2006-05-09 In2Gen Co., Ltd. Factor VIII polypeptide
ATE497783T1 (en) 2003-05-06 2011-02-15 Syntonix Pharmaceuticals Inc CLOTTING FACTOR VII-FC CHIMERIC PROTEINS FOR THE TREATMENT OF HEMOSTATIC DISEASES
US7211559B2 (en) 2003-10-31 2007-05-01 University Of Maryland, Baltimore Factor VIII compositions and methods
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
NZ555032A (en) 2004-11-12 2010-02-26 Bayer Healthcare Llc Site-directed modification of FVIII
SG123635A1 (en) 2004-12-28 2006-07-26 Nanoscience Innovation Pte Ltd Nanostructured zinc oxide and a method of producing the same
US7855279B2 (en) 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
US8048848B2 (en) 2006-02-03 2011-11-01 Prolor Biotech Ltd. Long-acting interferons and derivatives thereof and methods thereof
AU2007223888A1 (en) 2006-03-06 2007-09-13 Amunix, Inc. Genetic packages and uses thereof
EP2097096B1 (en) * 2006-12-22 2017-05-31 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
EP2185701A4 (en) 2007-08-15 2011-03-02 Amunix Operating Inc Compositions and methods for modifying properties of biologically active polypeptides
US8239182B2 (en) 2007-12-04 2012-08-07 Spansion Llc Data transmission system-on-chip memory model based validation
CN102348715B (en) 2009-02-03 2017-12-08 阿穆尼克斯运营公司 Extension recombinant polypeptide and the composition for including the extension recombinant polypeptide
EA025416B1 (en) 2009-12-06 2016-12-30 Байоджен Хемофилия Инк. Method for preventing or treating bleeding episodes using a long-acting factor viii chimeric polypeptide
WO2012006623A1 (en) * 2010-07-09 2012-01-12 Biogen Idec Hemophilia Inc. Systems for factor viii processing and methods thereof
EA201291482A1 (en) * 2010-07-09 2013-10-30 Байоджен Айдек Хемофилия Инк. CHEMERIC COAGULATION FACTORS
TWI687226B (en) * 2011-07-08 2020-03-11 美商百歐維拉提夫治療公司 Factor viii chimeric and hybrid polypeptides, and methods of use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7348004B2 (en) 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
US7404956B2 (en) 2003-05-06 2008-07-29 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
EP2173890A1 (en) 2007-06-21 2010-04-14 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
US20100292130A1 (en) 2007-06-21 2010-11-18 Technische Universitat Munchen Biological active proteins having increased in vivo and/or in vitro stability

Non-Patent Citations (10)

* Cited by examiner, † Cited by third party
Title
"Genes II", 1985, JOHN WILEY & SONS
BAI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 7292 - 7296
BRANDSMA ET AL., BIOTECHNOL. ADV., vol. 29, 2011, pages 230 - 238
FRANCIS, FOCUS ON GROWTH FACTORS
KIM ET AL., J. PHARMACOL. EXP. THER., vol. 334, 2010, pages 682 - 692
LI ET AL., TRENDS PHARMACOL. SCI., vol. 23, 2002, pages 206 - 209
MALIK F ET AL., EXP. HEMATOL., vol. 20, 1992, pages 1028 - 35
SOMMERMEYER ET AL., KRANKENHAUSPHARMAZIE, vol. 8, 1987, pages 271 - 278
WANG ET AL., J. CONTROLLED RELEASE, vol. 155, 2011, pages 386 - 392
WEIDLER ET AL., ARZNEIM.-FORSCHUNG/DRUG RES., vol. 41, 1991, pages 494 - 498

Cited By (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637007B2 (en) 2006-12-15 2014-01-28 Baxter International Inc. Factor VIIa-polysialic acid conjugate having prolonged in vivo half-life
US10414793B2 (en) 2009-07-27 2019-09-17 Baxalta Incorporated Nucleophilic catalysts for oxime linkage
US8637640B2 (en) 2009-07-27 2014-01-28 Baxter International Inc. Blood coagulation protein conjugates
US8809501B2 (en) 2009-07-27 2014-08-19 Baxter International Inc. Nucleophilic catalysts for oxime linkage
US11564992B2 (en) 2009-07-27 2023-01-31 Takeda Pharmaceutical Company Limited Nucleophilic catalysts for oxime linkage
US10350301B2 (en) 2009-07-27 2019-07-16 Baxalta Incorporated Blood coagulation protein conjugates
US10576160B2 (en) 2009-07-27 2020-03-03 Baxalta Incorporated Nucleophilic catalysts for oxime linkage
US9492555B2 (en) 2009-07-27 2016-11-15 Baxalta Incorporated Nucleophilic catalysts for oxime linkage
US10772968B2 (en) 2009-07-27 2020-09-15 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
US9731024B2 (en) 2009-07-27 2017-08-15 Baxalta Incorporated Nucleophilic catalysts for oxime linkage
US9795683B2 (en) 2009-07-27 2017-10-24 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
US11040109B2 (en) 2009-07-27 2021-06-22 Takeda Pharmaceutical Company Limited Blood coagulation protein conjugates
US11266720B2 (en) 2009-12-06 2022-03-08 Bioverativ Therapeutics Inc. Factor VIII-FC chimeric and hybrid polypeptides, and methods of use thereof
US8945897B2 (en) 2010-07-26 2015-02-03 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
US8642737B2 (en) 2010-07-26 2014-02-04 Baxter International Inc. Nucleophilic catalysts for oxime linkage
US10317376B2 (en) 2012-04-11 2019-06-11 Bioverativ Therapeutics Inc. Methods of detecting glycosaminoglycans
EP3521321A1 (en) 2012-04-11 2019-08-07 Bioverativ Therapeutics Inc. Methods of detecting glycosaminoglycans
US11261437B2 (en) 2012-06-08 2022-03-01 Bioverativ Therapeutics Inc. Procoagulant compounds
US10202595B2 (en) 2012-06-08 2019-02-12 Bioverativ Therapeutics Inc. Chimeric clotting factors
US11168316B2 (en) 2012-06-08 2021-11-09 Bioverativ Therapeutics, Inc. Chimeric clotting factors
US10287564B2 (en) 2012-06-08 2019-05-14 Bioverativ Therapeutics Inc. Procoagulant compounds
EP2870250B1 (en) 2012-07-06 2018-04-18 Bioverativ Therapeutics Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
US10023628B2 (en) 2012-07-06 2018-07-17 Bioverativ Therapeutics Inc. Cell line expressing single chain factor VIII polypeptides and uses thereof
EP2870250B2 (en) 2012-07-06 2022-06-29 Bioverativ Therapeutics Inc. Cell line expressing single chain factor viii polypeptides and uses thereof
EP3446700A1 (en) * 2012-10-30 2019-02-27 Bioverativ Therapeutics Inc. Methods of using fviii polypeptide
EP3943102A1 (en) * 2012-10-30 2022-01-26 Bioverativ Therapeutics Inc. Methods of using fviii polypeptide
EP2914293A4 (en) * 2012-10-30 2016-04-20 Biogen Ma Inc Methods of using fviii polypeptide
US10370431B2 (en) 2013-02-15 2019-08-06 Bioverativ Therapeutics Inc. Optimized factor VIII gene
EP3889173A1 (en) * 2013-02-15 2021-10-06 Bioverativ Therapeutics Inc. Optimized factor viii gene
EP2956477A4 (en) * 2013-02-15 2016-12-28 Biogen Ma Inc Optimized factor viii gene
US11787851B2 (en) 2013-02-15 2023-10-17 Bioverativ Therapeutics Inc. Optimized factor VIII gene
EP4223772A3 (en) * 2013-02-15 2023-10-18 Bioverativ Therapeutics Inc. Optimized factor viii gene
US10786554B2 (en) 2013-03-15 2020-09-29 Bioverativ Therapeutics Inc. Factor VIII polypeptide formulations
EP3666283A1 (en) * 2013-03-15 2020-06-17 Bioverativ Therapeutics Inc. Factor viii polypeptide formulations
EA030357B1 (en) * 2013-03-15 2018-07-31 Биовератив Терапьютикс Инк. Factor viii polypeptide formulations
US9623088B2 (en) 2013-03-15 2017-04-18 Bioverativ Therapeutics Inc. Factor VIII polypeptide formulations
EP4122487A1 (en) * 2013-03-15 2023-01-25 Bioverativ Therapeutics Inc. Factor viii polypeptide formulations
WO2014144795A1 (en) * 2013-03-15 2014-09-18 Biogen Idec Ma Inc. Factor viii polypeptide formulations
US11168124B2 (en) 2015-10-30 2021-11-09 Spark Therapeutics, Inc. CpG reduced factor VIII variants, compositions and methods and uses for treatment of hemostasis disorders
US11753461B2 (en) 2016-02-01 2023-09-12 Bioverativ Therapeutics Inc. Optimized factor VIII genes

Also Published As

Publication number Publication date
TWI687226B (en) 2020-03-11
RS58578B1 (en) 2019-05-31
IL271870B (en) 2022-04-01
TWI764092B (en) 2022-05-11
AR087091A1 (en) 2014-02-12
EA201490039A1 (en) 2014-06-30
TW201822788A (en) 2018-07-01
AU2016204986B2 (en) 2018-05-10
EP3513804B1 (en) 2022-03-23
HRP20220639T1 (en) 2022-07-08
IL271870A (en) 2020-02-27
DK2729161T3 (en) 2019-04-08
SI3513804T1 (en) 2022-07-29
EP2729161A2 (en) 2014-05-14
KR20180020305A (en) 2018-02-27
EP2729161B1 (en) 2018-12-19
DK3513804T3 (en) 2022-06-20
MX2014000202A (en) 2014-10-06
LT3513804T (en) 2022-05-25
PT3513804T (en) 2022-06-02
KR101829603B1 (en) 2018-02-19
AU2018202936B2 (en) 2020-07-02
EP2729161A4 (en) 2015-06-24
ES2722209T3 (en) 2019-08-08
US10881742B2 (en) 2021-01-05
EA029045B1 (en) 2018-02-28
BR112014000466A2 (en) 2017-02-21
KR102110736B1 (en) 2020-05-14
MX350581B (en) 2017-09-11
JP2019048877A (en) 2019-03-28
JP2022003096A (en) 2022-01-11
PT2729161T (en) 2019-04-01
CA2841066C (en) 2023-09-26
KR20140066157A (en) 2014-05-30
PL2729161T3 (en) 2019-08-30
AU2018202936A1 (en) 2018-05-17
LT2729161T (en) 2019-04-10
SI2729161T1 (en) 2019-05-31
WO2013009627A3 (en) 2013-03-07
TWI626947B (en) 2018-06-21
US10010622B2 (en) 2018-07-03
HRP20190535T1 (en) 2019-05-17
JP7273487B2 (en) 2023-05-15
JP2014522838A (en) 2014-09-08
AU2016204986A1 (en) 2016-08-04
CL2014000024A1 (en) 2014-11-21
NZ619438A (en) 2016-06-24
ES2913994T3 (en) 2022-06-07
EP3513804A1 (en) 2019-07-24
US20180360982A1 (en) 2018-12-20
AU2012282875A1 (en) 2013-05-02
CY1122900T1 (en) 2021-05-05
JP2017088623A (en) 2017-05-25
CO6940381A2 (en) 2014-05-09
TW201315480A (en) 2013-04-16
CN107261122A (en) 2017-10-20
CA2841066A1 (en) 2013-01-17
US20210220476A1 (en) 2021-07-22
US20140294821A1 (en) 2014-10-02
AU2012282875B2 (en) 2016-04-21
CN103796670A (en) 2014-05-14
IL230333B (en) 2020-05-31
TW202042838A (en) 2020-12-01
JP2023134851A (en) 2023-09-27
MY180714A (en) 2020-12-07
PL3513804T3 (en) 2022-07-11
HUE043763T2 (en) 2019-09-30
RS63241B1 (en) 2022-06-30
EP4169525A1 (en) 2023-04-26

Similar Documents

Publication Publication Date Title
US20210220476A1 (en) Factor viii chimeric and hybrid polypeptides, and methods of use thereof
AU2018267631B2 (en) Methods of Reducing Immunogenicity Against Factor VIII in Individuals Undergoing Factor VIII Therapy
US20210069300A1 (en) Factor viii polypeptide formulations
MX2012006347A (en) Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof.
US20200030420A1 (en) Methods of using a fixed dose of a clotting factor
US20150266944A1 (en) Methods of Using FVIII Polypeptide
AU2013204237B8 (en) Methods of reducing immunogenicity against Factor VIII in individuals undergoing Factor Vlll therapy
NZ619438B2 (en) Factor viii chimeric and hybrid polypeptides, and methods of use thereof
EA045299B1 (en) FACTOR VIII POLYPEPTIDE PREPARATIONS

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: 12811808

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2012282875

Country of ref document: AU

Date of ref document: 20120706

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2841066

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014519074

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/000202

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: P20/2014

Country of ref document: AE

WWE Wipo information: entry into national phase

Ref document number: 201490039

Country of ref document: EA

WWE Wipo information: entry into national phase

Ref document number: 14011134

Country of ref document: CO

ENP Entry into the national phase

Ref document number: 20147003130

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012811808

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014000466

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 14131600

Country of ref document: US

ENP Entry into the national phase

Ref document number: 112014000466

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140108