METHODS OF USING A FIXED DOSE OF A CLOTTING FACTOR

The present invention provides methods of administering a clotting factor by a fixed dosing regimen; methods of reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder; and a kit comprising a dotting factor useful for a fixed dosing regimen. While plasma-derived and recombinant clotting factor products allow hemophilia patients to live longer and healthier, hemophilia still remains one of the most costly and complex conditions to manage.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
BACKGROUND OF THE INVENTION Field of the Invention

The present invention relates generally to the field of therapeutics for hemostatic disorders.

Background Art

While plasma-derived and recombinant clotting factor products allow hemophilia patients to live longer and healthier, hemophilia still remains one of the most costly and complex conditions to manage. The cost of clotting factor products exceeds $50,000 a year per patient. See Blankenship C. S., Biotechnol. Healthc. 2008, 5(4): 37-40. According to the National Heart, Lung, and Blood Institute, National institute of Health (NIH), approximately 18,000 people in the U.S. have hemophilia, and 400 babies are born with the disease each year. Morbidity & Mortality: 2012 Chart Book on Cardiovascular, Lung and Blood Disease, page 5, National Heart, Lung, and Blood Institute, NIH. Due to its complexity, this chronic disease requires a special therapeutic management process for doctors, pharmacies, and patients. Clinicians often assess lifestyle, psychosocial requirements, and the home environment when evaluating a patient's or guardian's ability to provide adequate care.

In hemophilia, blood clotting is disturbed by a lack of certain plasma blood clotting factors. Hemophilia A, the most common form of hemophilia, is caused by Factor VIII deficiency. Hemophilia B is caused by decreased synthesis of Factor IX protein or synthesis of defective Factor IX having reduced activity. Treating hemophilia involves replacing missing or defective clotting factor with recombinant or plasma-derived FVIII or FIX. For patients who have developed antibodies against recombinant or plasma-derived FVIII or FIX, Factor VII can be used as a bypass therapy. Commercially available clotting factors are usually administered by peripheral intravenous injection. However, for patients with small veins or children who require frequent injections, clotting factors can be administered by a central venous access device. See Blankenship C. S., Biotechnol. Healthc. 2008, 5(4): 37-40.

Many biologics including clotting factors are administered based on patient body size. Body sized-based dosing is assumed to minimize inter-patient variability in pharmacokinetics (PK). Currently, three FIX products are approved by the Food and Drug Administration (FDA). The first, BENEFIX®, is a recombinant FIX product marketed by Pfizer. The second and third products are plasma-derived FIX products, ALPHANINE® marketed by Grifols and MONONINE® marketed by CSL Behring. According to their labels, these three products are dosed based on individual body weight. In particular, BENEFIX® is supplied as a lyophilized powder in five different dosages: 250 IU, 500 IU, 1000 IU, 2000 IU, and 3000 IU. MONONINE® is supplied as a single dose vial with Sterile Water for Injection at 500 IU and 1000 IU. ALPHANINE is supplied in lyophilized form as single doses at 500 IU, 1000 IU, and 1500 IU. The FIX dose required for each patient is calculated based on the formula:


Number of factor IX IU required (IU)=Body Weight (kg)×Desired Factor IX Increase (% or IU/dL)×Reciprocal of Observed Recovery (IU/kg per IU/dL)   (A)

Several Factor VIII products are also commercially available, which include recombinant FVIII products (ADVATE® and RECOMBINATE® marketed by Baxter, KOGENATE® FS marketed by Bayer, HELIXATE® FS marketed by CSL-Behring, and XYNTHA® and REFACTO® marketed by PFIZER) and Plasma-derived FVIII products (HEMOFIL-M® marketed by Baxter, MONARC-M® by American Red Cross, and MONOCLATE-P® marketed by CSL Behring). The required VIII dose for each patient is calculated using the following formula:


Number of factor FVIII IU required (IU)=Body Weight (kg)×Desired Factor FVIII Increase (IU/dL or % of normal)×0.5(IU/kg, per IU/dL)   (B)

A Factor VII product, NOVOSEVEN® marketed by Novo Nordisk, is also commercially available. The dosages of NOVOSEVEN® are also calculated based on body weight: 90 μg/kg bolus injection, every two hours for Hemophilia A or B with inhibitors, 15-30 μg/kg every 4-6 hours for congenital FVII deficiency, or 70-90 μg/kg every 2-3 hours for acquired hemophilia. See NOVOSEVEN® label, page 1, Jan. 2010, version 3, Novo Nordisk A/S.

However, administering clotting factors via body weight-based dosing can be inconvenient and costly for patients. The invention as described herein provides improved clotting factor-dosing strategies.

BRIEF SUMMARY OF THE INVENTION

In certain embodiments, the present invention provides a method of providing a clotting factor comprising administering a fixed dose of a clotting factor to a subject in need thereof In certain embodiments, a method of reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject comprising administering a fixed dose of a clotting factor to a subject in need thereof is provided. In some, aspects, the clotting factor is a modified clotting factor. In some embodiments, the modified clotting factor comprises a clotting factor and a heterologous moiety, e.g., heterologous moiety which increases in vivo half-life of the clotting factor. In some aspects the heterologous moiety is a non-polypeptide moiety or a polypeptide moiety. In certain aspects, the heterologous moiety comprises albumin, albumin binding polypeptide, an FcRn binding partner, PAS, the C-terminal peptide (CTP) of the β subunit of human chorionic gonadotropin, polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin-binding small molecules, or combinations thereof In certain, aspects, the modified clotting factor is a long-acting clotting factor.

In some embodiments, the fixed dose of a clotting factor is administered at regular intervals of every day, every two days, every three days, twice a week, every four days, every five days, every six days, every week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, every two weeks, every three weeks, or every four weeks. In certain embodiments, the fixed dose is administered as needed to control bleeding.

In some aspects, the clotting factor has a wide therapeutic window. For example, the therapeutic window for the clotting factor can be a maximum serum concentration (Cmax) of about 150% of normal and a minimum serum concentration (Cmin) of about 1% of normal.

In other aspects, the clotting factor has a narrow therapeutic window.

In certain embodiments provided herein, the body weight effect on, clearance (θBW_CL) of the clotting factor is equal to or less than about 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, about 0.65, about 0.60, about 0.59, about 0.58, about 0.57, about 0.56, about 0.55, about 0.54, about 0.53, about 0.52, about 0.51, about 0.50, about 0.49, about 0.48, about 0.47, about 0.46, about 0.45, about 0.44, about 0.43, about 0.42, about 0.41, about 0.40, about 0.35, about 0.30, about 0.25, about 0.20, about 0.15, about 0.10, about 0.05, or about 0. Alternatively, or in addition, the body weight effect on the central volume of distribution (θBW_V1) of the clotting factor is equal to or less than about 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, about 0.65, about 0.60, about 0.59, about 0.58, about 0.57, about 0.56, about 0.55, about 0.54, about 0.53, about 0.52, about 0.51, about 0.50, about 0.49, about 0.48, about 0.47, about 0.46, about 0.45, about 0.44, about 0.43, about 0.42, about 0.41, about 0.40, about 0.35, about 0.30, about 0.25, about 0.20. about 0.15, about 0.10, about 0.05, or about 0.

In specific embodiments, the θBW_CL of the clotting factor is equal to or less than about 0.500 and/or the νBW_V1 of the clotting factor is equal to or less than about 0.467. For example, in some embodiments the θBW_CL of the clotting factor is about 0.500 and/or the θBW_V1 of the clotting factor is about 0.467.

In some embodiments of the method provided herein the body weight of the subject does not produce pharmacodynamic variability within subjects. In other aspects, administration of a fixed dose of the clotting factor results in reduced variability of pharmacokinetic parameters across all body weights, as compared to administration of a body weight-based dose of the clotting factor. For example, in certain embodiments the pharmacokinetic parameter is area under the curve (AUC) and variability in AUC for a fixed dose of the clotting factor is less than ±50%, less than ±45%, less than ±40%, less than ±35%, less than ±30%, or less than ±25% across all body weights.

In certain aspects of the method provided herein the clotting factor is a long-acting FIX polypeptide. The long-acting FIX polypeptide can include a FIX polypeptide and an FcRn binding partner, and the FcRn binding partner can include an Fc region. The long-acting FIX polypeptide can further include a second FcRn binding partner, which can include a second Fc region. In certain aspects the FcRn binding partner and the second FcRn binding partner are associated, e.g., by a covalent bond, e.g., by a disulfide bond. In other aspects the second FcRn binding partner is not linked to an amino acid sequence by a peptide bond. In certain embodiments, the long-acting FIX polypeptide is FIX monomer dimer hybrid.

According to the present disclosure, the fixed dose of a long acting FIX polypeptide can be standard across all body weights, e.g., about 4000 IU per dose which is, e.g., administered weekly, or about 8000 IU which is, e.g., administered weekly. In other embodiments, the fixed dose is administered every 10 days.

In certain aspects a fixed dose of a long acting FIX polypeptide is stratified into multiple (e.g., two or more) fixed dose amounts based on specified weight categories, such as low body weight, normal body weight, and high body weight. For example, the fixed dose can be stratified into three fixed dose amounts suitable for subjects with low, normal, or high body weight. In one embodiment, the normal, low, or high body weight is determined based on age, height, gender, frame size, general health, or any combination thereof. In another embodiment, the normal, low, or high body weight is determined independently of age, height, gender, frame size, general health, or any combination thereof In other embodiments, the normal body weight for a human subject is between about 50+10 kg, and about 100±10 kg. In some embodiments, the low body weight for a human subject is less than about 50±10 kg. In still other embodiments, the high body weight for a human subject is greater than, about 100±10 kg.

In some aspects, the fixed dose is administered weekly (L e., once a week). In other aspects, the fixed dose is administered every 10 days. In one embodiment, the subject has a low body weight and the fixed dose is about 5000 IU per dose administered every 10 days or about 6000 IU per dose administered every 10 days. In another embodiment, the subject has a normal body weight and the fixed dose is about 7500 IU per dose administered every 10 days or about 8000 IU per dose administered every 10 days. In other embodiments, the subject has a high body weight and the fixed dose is about 10000 IU per dose administered every 10 days or about 12000 IU per dose administered every 10 days.

In further aspects, the clotting factor is a long-acting FVIII polypeptide. For example, the long-acting FVIII polypeptide comprises a FVIII polypeptide and an FcRn binding partner, e.g., an Fc region. In some embodiments, the long-acting FVIII polypeptide further comprises a second FcRn binding partner, e.g., a second Fc region. In one example, the FcRn binding partner and the second FcRn binding partner are associated, e.g., by a covalent bond, e.g., a disulfide bond. In another example, the long-acting FVIII polypeptide is FVIII monomer dimer hybrid. In other examples, the FVIII polypeptide in the long-acting polypeptide is a full-length FVIII or a B-domain deleted FVIII.

In one aspect, the fixed dose is standard across all body weights. In one embodiment, the fixed dose is administered twice weekly. In another embodiment, the fixed dose is administered weekly. In other embodiments, the fixed dose is stratified into multiple (e.g., two or more) dose amounts based on specified weight categories, e.g., low body weight, normal body weight, and high body weight. In other embodiments, the fixed dose is stratified into three dose sizes suitable for subjects with low, normal, or high body weight. In some embodiments, the normal, low, or high body weight is determined based on age, height, gender, frame size, general health, or any combination thereof In other embodiments, the low, normal, or high body weight is determined independently of age, height, gender, frame size, general health, or any combination thereof In one aspect, the normal body weight for a human subject is between about 50±10 kg and about 100±10 kg. In another aspect, the low body weight for a human subject is less than about 50±10 kg. In other aspects, the high body weight for a human subject is greater than about 100±10 kg. In one example, the fixed dose for the long-acting FVIII polypeptide is administered twice weekly at about 2000 IU, about 2,500 IU, about 3,000 IU, about 3,500 IU, or about 4,000 IU. In another example, the fixed dose is administered weekly.

In some aspects, the fixed dose of the clotting factor is to prevent one or more bleeding episodes. In one embodiment, the fixed dose of the clotting factor is for individualized interval prophylaxis of a bleeding episode. In another embodiment, the fixed dose of the clotting factor is for on-demand or episodic treatment of a bleeding episode. In other embodiments, the fixed dose of the clotting factor is for perioperative management of a bleeding episode. In certain embodiments, the subject is in need of controlling or preventing bleeding or bleeding episodes, for example, in need of peri-operative management or in need of management of bleeding associated with surgery or dental extraction. In some embodiments, the subject will undergo, is undergoing, or has undergone major surgery. In certain embodiments, the subject is in need of prophylactic treatment or in need of on-demand treatment.

In other aspects, the fixed dose is provided in a single container (e.g., vial) or in two or more containers (e.g., vials), the total contents of which provide the fixed dosage amount.

The invention also includes use of a fixed dosage of a clotting factor for the manufacture of a medicament for reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject in need thereof The medicament can be administered according to the method described herein.

Further included is a fixed dosage of a modified clotting factor for use in reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject in need thereof The fixed dosage of the invention is suitable for administration according to the method described herein.

The present invention also includes a pharmaceutical composition comprising a fixed dose of a modified clotting factor and a pharmaceutically acceptable carrier for use to reduce, ameliorate, or prevent one or more symptoms of a bleeding disease or disorder in a subject in need thereof. The pharmaceutical composition is suitable for administration according to the method described herein.

The present invention further includes a kit comprising the pharmaceutical composition described herein and instructions to administer the fixed dose of the clotting factor to the subject. In one embodiment, the entire fixed dose is administered. In another embodiment, the fixed dose is provided in a single container (e.g., vial). In other embodiments, the fixed dose is provided in two or more containers (e.g., vials), the total contents of which provide the fixed dosage.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows a diagram of the three-compartment model for predicting population PK for rFIXFc. CL, clearance; V, volume of distribution; Q, inter-compartmental clearance. V1 shows volume of distribution in central compartment; and V2 and V3 show volume of distribution in, peripheral compartments. Q2 is inter-compartmental clearance between V1 and V2. Q3 is inter-compartmental clearance between V1 and V3.

FIG. 2A shows clearance (CL) estimates of baseline (week 1) and repeat PK (week 26) profiles. FIG. 2B shows Volume of Distribution of central compartment (V1) estimates of baseline (week 1) and repeat PK (week 26) profiles. The thick line in the middle of FIG. 2A and 2B indicates mean, which did not change much between two occasions.

FIGS. 3A to 3E show individual PK parameters versus body weight (BW). FIG. 3A shows clearance in dL/h. FIG. 3B shows Volume of Distribution of central compartment (V1) in dL. FIG. 3C shows inter-compartmental clearance (Q2) in dL/h. FIG. 3D shows Volume of Distribution in a peripheral compartment (V2) in dL/h. FIG. 3E shows Volume of Distribution of a peripheral compartment (V3).

FIG. 4A shows goodness-of-fit plots of FIX activity predicted by the population PK model compared to observed FIX activity. FIG. 4B shows goodness-of-fit plots of FIX activity predicted by the individual PK model compared to observed FIX activity.

FIG. 5A shows Visual Predictive Check (VPC) plots of the population PK model for 50 IU/kg dose. FIG. 5B shows VPC plots of the population PK model for 100 IU/kg dose. Gray and black lines represent 10th, 50th, and 90th percentile of the simulated (gray) and observed (black) data, respectively.

FIG. 6 shows validation of the population PK model with the trough/peak records. R2=0.9857, P<0.001.

FIG. 7A shows the 97.5th, median, and 2.5th percentiles of the simulated FIX activity-time profiles at steady state in 1000 subjects following fixed dosing (4000 IU once weekly; dotted line) compared with the 97.5th, median, and 2.5th percentiles of the simulated FIX activity-time profiles at steady state in 1000 subjects following BW-based dosing (50 IU/kg once weekly; solid line). FIG. 7B shows the 97.5th, median, and 2.5th percentiles of the simulated FIX activity-time profiles at steady state in 1000 subjects following fixed dosing (8000 IU every 10 days) compared with the 97.5th, median, and 2.5th percentiles of the simulated FIX activity-time profiles at steady state in 1000 subjects following BW-based dosing (100 IU/kg every 10 days; solid lines).

FIG. 8 shows the percentiles of population within the target therapeutic range following the fixed dosing and BW-based dosing approaches in the BW-stratified populations.

DETAILED DESCRIPTION OF THE INVENTION

The present invention is derived from the recognition that a fixed dosing regimen can be suitable for a clotting factor. The present invention thus provides a method of administering a fixed dose of a clotting factor to a subject in need thereof or a population of two or more subjects in need thereof. Administration of the fixed dose of the clotting factor can reduce, ameliorate, or prevent one or more symptoms of a bleeding disease or disorder. For example, administration of the fixed dose of the clotting factor can control or prevent a bleeding episode. The invention also includes a kit comprising one or more pharmaceutical compositions sand an instruction manual, wherein the one or more pharmaceutical composition comprises a fixed dose of a clotting factor.

I. Definitions

It must be noted that, as used in this specification and the appended claims, the singular forms “a”, “an” and “the” include plural referents unless the context clearly dictates otherwise. The terms “a” (or “an”), as well as the terms “one or more,” and “at least one” can be used interchangeably herein.

Furthermore, “and/or” where used herein is to be taken as specific disclosure of each of the two specified features or components with or without the other. Thus, the term “and/or” as used in a phrase such as “A and/or B” herein is intended to include “A and B,” “A or B,” “A” (alone), and “B” (alone). Likewise, the term “and/or” as used in, a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).

The term “about” is used herein to mean approximately, roughly, around, or in the regions of. When the term “about” is used in conjunction with a numerical range, it modifies that range by extending the boundaries above and below the numerical values set forth. In general, the term “about” is used herein to modify a numerical value above and below the stated value by a variance of 10 percent, up or down (higher or lower).

It is understood that wherever embodiments are described herein with the language “comprising,” otherwise analogous embodiments described in terms of “consisting of” and/or “consisting essentially of” are also provided.

The term “polypeptide,” “peptide” and “protein” >are used interchangeably and refer, to a polymeric compound comprised of covalently linked amino acid residues.

The term “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, but are not limited to, those in Tables 4 and 6, which encode the polypeptides of Table 5 and 7. Polynucleotides also include fragments, variants, analogues, or derivatives of the polynucleotides of Tables 4 and 6, e.g., those that encode fragments of the polypeptides of Table 5, 7, or 8.

Unless, defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure is related. For example, the Concise Dictionary of Biomedicine and Molecular Biology, Juo, Pei-Show, 2nd ed., 2002, CRC Press; The Dictionary of Cell and Molecular Biology, 3rd ed., 1999, Academic Press; and the Oxford Dictionary Of Biochemistry And Molecular Biology, Revised, 2000, Oxford University Press, provide one of skill with a general dictionary of many of the terms used in this disclosure.

Units, prefixes, and symbols are denoted in their Système International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, amino acid sequences are written left to right in amino to carboxy orientation. The headings provided herein are not limitations of the various aspects or embodiments of the disclosure, which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification in its entirety. Amino acids are referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, are referred to by their commonly accepted single-letter codes.

An “isolated” polypeptide, antibody, polynucleotide, vector, cell, or composition refers to a polypeptide, antibody, polynucleotide, vector, cell, or composition that is in a form not found in nature. Isolated polypeptides, antibodies, polynucleotides, vectors, cells or compositions include those which have been purified to a degree that they are no longer in a form in which they are found in nature. In some aspects, an antibody, polynucleotide, vector, cell, or composition that is isolated is substantially pure. In some aspects an antibody, polynucleotide, vector, cell, or composition that is isolated is “recombinant.”

The term “administering,” as used herein, means to prescribe or to give a pharmaceutically acceptable clotting factor to a subject via a pharmaceutically acceptable route. Examples of routes of administration include, but are not limited to, intravenous, e.g., intravenous injection and intravenous infusion, e.g., via central venous access. Additional routes of administration include subcutaneous, intramuscular, oral, nasal, and pulmonary administration. A clotting factor (e.g., a FIX or FVIII or modified clotting factor protein) may be administered as part of a pharmaceutical composition comprising at least one excipient.

The term “modified clotting factor” as used herein means a clotting factor sequence that is modified in the polypeptide or polynucleotide sequence by deletion, substitution, insertion, conjugation, linkage, fusion, glycosylation, or any types of modifications that are not present in the polypeptide sequences in the wild-type clotting factor (e.g., FIX or FVIII) or the commercially available clotting factor (e.g., REFACTO® Or XYNTHA® for SQ BDD FVIII; RECOMBINATE®, ADVATE®, OR HELIXATE® for full-length FVIII; or BENEFIX®, ALPHANINE®, or MONONINE® for FIX).

The terms “long-acting” and “long-lasting” are used interchangeably herein. In one embodiment, the term “long-acting” or “long-lasting” indicates that the clotting activity as a result of administration of a “long-acting” clotting factor is longer than the clotting activity of a wild-type clotting factor (also referred to as “short acting” or “shorter acting” clotting factor) (e.g., BENEFIX® or plasma-derived FIX (“pdFIX”) for FIX, or SQ B domain deleted FVIII (e.g., REFACTO®) or mature full-length FVIII, e.g., RECOMBINATE®, for FVIII). The “longer” clotting activity can be measured by any known methods in the art, e.g., aPTT assay, chromogenic assay, ROTEM, TGA, and etc. In one embodiment, the “longer” clotting activity can be shown by the T1/2beta (activity). In another embodiment, the “longer” clotting activity can be shown the level of the clotting factor present in plasma, e.g., by the T1/2beta (antigen). In other embodiments, the long-acting or long-lasting clotting factor works longer in a coagulation cascade, e.g., is active for a longer period, compared to a wild-type clotting factor (e.g., BENEFIX® or plasma-derived FIX (“pdFIX”) for FIX or SQ B domain deleted FVIII REFACTO®) or mature full-length FVIII, e.g., RECOMBINATE®, for FVIII). The long-acting or long-lasting clotting factor can comprise one or more heterologous moieties that extend in vivo half-life of the clotting factor. Examples of the heterologous moieties are described below.

The term “chimeric clotting factor” as used herein, means a polypeptide that includes within it at least two polypeptides (or portions thereof such as subsequences or peptides) from different sources. Chimeric clotting factor can include two, three, four, five, six, seven, or more, polypeptides or portions thereof from different sources, such as different genes, different cDNAs, or different animal or other species. Chimeric clotting factors can include one or more linkers joining the different polypeptides or portions thereof. Thus, the polypeptides or portions thereof can be joined directly or they may be joined indirectly, via linkers, or both, within a single chimeric polypeptide. In certain embodiments, chimeric clotting factors can include additional peptides such as signal sequences and sequences such as 6His and FLAG that aid in protein purification or detection. In addition, chimeric clotting factors can have amino acid or peptide additions to the N- and/or C-termini. Exemplary chimeric clotting factors of the invention are Factor IX-Fx chimeric polypeptides or FVIII-Fc chimeric polypeptides.

“Dosing interval,” as used herein, means the amount of time that elapses between multiple doses being administered to a subject. Dosing interval can thus be indicated as a range. The dosing interval in the methods of the invention using a clotting factor can depend on the specific clotting factor. For example, a dosing interval of a long-acting clotting factor can be at least about one and a quarter, at least one and one-half to ten times longer than the dosing interval required for an equivalent amount (in IU/kg) of the wild-type clotting factor (i.e., a short-acting clotting factor). The dosing interval when administering, e.g., a Factor IX-Fc chimeric polypeptide (or a hybrid) of the invention can be at least about three times longer than the dosing interval required for an equivalent amount of said Factor IX without the FxRn BP (defined below), e.g., Fc, portion (i.e., a polypeptide consisting of said Factor IX). The dosing interval when administering, e.g., a Factor VIII-Fc chimeric polypeptide (or a hybrid) of the invention can be at least about one and a quarter, at least one and one-half times longer than the dosing interval required for an equivalent amount of the FVIII without the FcRn BP, e.g., Fc, portion (i.e., a polypeptide consisting of the FVIII). The dosing interval may be at least about one and one-half to fifteen times longer than the dosing interval required for an equivalent amount of the FIX or FVIII without, e.g., the Fc portion (or a polypeptide consisting of the FIX or FVIII portion).

The term “dosing frequency” as used herein refers to the frequency of administering doses of a clotting factor in a given time. Dosing frequency can be indicated as the number of doses per a given time, e.g., once a week or once in two weeks.

“Therapeutic dose,” “dose,” “effective dose,” or “dosing amount” as used herein, means a dose that achieves a plasma trough level of a clotting activity at least about 1 IU/dl or above in the subject administered with the clotting factor. For the purpose of this invention, a “dose” can refer to the amount of the clotting factor required to maintain a plasma trough level of a clotting activity of at least about 1 IU/dl or above 1 IU/dl, at least about 2 IU/dl or above 2 IU/dl, at least about 3 IU/dl or above 3 IU/dl, at least about 4 IU/dl or above 4 IU/dl, at least about 5 IU/dl or above 5 IU/dl, at least about 6 IU/dl or above 6 IU/dl, at least about 7 IU/dl or above 7 IU/dl, at least about 8 IU/dl or above 8 IU/dl, at least about 9 IU/dl or above 9 IU/dl, at least about 10 IU/dl or above 10 IU/dl, at least about 11 IU/dl or above 11 IU/dl, at least about 12 IU/dl or above 12 IU/dl, at least about 13 IU/dl or above 13 IU/dl, at least about 14 IU/dl or above 14 IU/dl, at least about 15 IU/dl or above 15 IU/dl, or at least about 20 IU/dl or above 20 IU/dl, throughout the administration of the clotting factor. In another embodiment, the “dose” reduces or decreases the frequency of bleeding or symptoms of a bleeding disorder. In other embodiments, the “dose” stops on-going, uncontrollable bleeding or bleeding episodes. In still other embodiments, the “dose” prevents spontaneous bleeding or bleeding episodes in a subject susceptible to such spontaneous bleeding or bleeding episodes. The “dose” or “therapeutic dose” need not cure hemophilia.

The term “fixed dosing” or “fixed dose” as used herein means a dosing amount given to a subject regardless of the body weight, or who have a body weight within a given range. In one example, a fixed dose can be given to any subjects in need thereof whether they have a low body weight (e.g., lower than 10th percentile of a body distribution), a normal body weight (e.g., between 10th percentile and 90th percentile of a body weight distribution), or a high body weight (e.g., higher than 90th percentile of a body weight distribution). In another example, fixed dosing can be stratified over two or more patient populations. For example, a first fixed dose can be given to a subject having a low extreme body weight (e.g., lower than 10th percentile of a body weight distribution); a second fixed dose can be given to a subject having a normal or high extreme body weight (e.g., equal to or higher than 10th percentile of a body weight distribution). In another example, fixed dosing can be stratified over three or more groups, for example a first fixed dose can be given to subjects having a low body weight (e.g., lower than 10th percentile of a body weight distribution); a second fixed dose can be given to subjects having a normal or high body weight (e.g., equal to or higher than 10th percentile of a body weight distribution), and a third fixed dose can be given to subjects having a high body weight (e.g., higher than 90th percentile of a body weight distribution).

The fixed dosing regimen can be stratified into two or more fixed dose amounts based on specified weight categories. In one embodiment, the weight, categories are low body weight, normal body weight, and high body weight. For example, the fixed dose can be stratified into multiple fixed dose amounts (e.g., three) suitable for subjects who fall within the weight categories, e.g., those with low, normal, or high body weight. The ranges of each body weight can be determined based on the patient's age, gender, frame size, height, general health, or any combinations thereof or independently of age, height, gender, frame size, general health, or any combination there. A person of ordinary skill in the art can assess the factors related to body weight and can determine the specific body weight category for a subject.

The phrase “normal body weight” as used herein means a body weight of a typical individual. Therefore, the phrase “normal body weight” is used interchangeably with the phase “typical body weight.” In one example, a subject having a normal body body weight has a body weight between about 50 kg±10 kg and about 110 kg±10 kg. In a particular example, a subject having a normal body weight has a body weight between 57 kg and 104 kg. The normal body weight is above a low body weight and below a high body weight.

The phrase “low body weight” as used herein means a body weight that is lower than the body weight of a typical individual. In one example, a subject having a low body weight is underweight. In another example, a subject having a low body weight has a body weight lower than about 50 kg±10 kg. In other embodiments, a low body weight is a low extreme body weight. In a particular example, a subject having a low body weight has a body weight lower than about 57 kg.

The phrase “high body weight” as used herein means a body weight that is higher than the body weight of typical individual. In one example, a subject having a high body weight is obese. In another example, a subject having a high body weight has a body weight higher than about 110 kg±10 kg. In other embodiments, a high body weight is a high extreme body weight. In a particular example, a subject having a high body weight has a body weight higher than about 104 kg.

The term “prophylaxis of one or more bleeding episodes,” “prevent one or more bleeding episodes” or “prophylactic treatment” as used herein means administering a clotting factor in fixed doses to a subject over a course of time to increase the level of clotting activity in a subject's plasma. In one embodiment, “prophylaxis of one or more bleeding episodes” or “prevent one or more bleeding episodes” indicates use of a clotting factor to prevent or inhibit occurrence of one or more spontaneous or uncontrollable bleeding or bleeding episodes or to reduce the frequency of one or more spontaneous or uncontrollable bleeding or Heeding episodes. “Routine prophylaxis” is used to prevent or reduce the frequency of bleeding episodes in subjects with hemophilia A or B. In another embodiment, the increased clotting activity level is sufficient to decrease the incidence of spontaneous bleeding or to prevent bleeding in the event of an unforeseen injury. Prophylactic treatment decreases or prevents bleeding episodes, for example, those described under on-demand treatment.

The term “about once a week” as used herein means approximate number, and “about once a week” can include every seven days±two days, i.e., every five days to every nine days. The dosing frequency of “once a week” thus can be every five days, every six days, every seven days, every eight days, or every nine days.

The term “individualized interval prophylaxis” as used herein means use of a long-acting clotting factor for an individualized dosing interval or frequency for a subject to prevent or inhibit occurrence of one or more spontaneous and/or uncontrollable bleeding or bleeding episodes or to reduce the frequency of one or more spontaneous and/or uncontrollable bleeding or bleeding episodes to the subject. In one embodiment, the “individualized interval” includes every 10 days±3 days, i.e. every seven days to every 13 days. The dosing frequency of the “individualized interval prophylaxis” thus can be every seven days, every eight days, every nine days, every ten days, every 11 days, every 12 days, or every 13 days.

The term “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 short term need such as planned surgery. The “on-demand treatment” is used interchangeably with “episodic” treatment. Conditions that may require on-demand treatment include 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. Bleeding episodes other than these known in the art are also included. The subject can be in need of surgical prophylaxis, peri-operative management, or treatment for surgery. Such surgeries include, but are not limited to, minor surgery, major surgery, tooth extraction, tonsillectomy, other dental/thoraco-facial surgeries, inguinal herniotomy, synovectomy, total knee replacement, other joint replacement, craniotomy, osteosynthesis, trauma surgery, intracranial surgery, intra-abdominal surgery, intrathoracic surgery. Surgeries other than these are also included.

Additional non-limiting conditions that can require on-demand treatment include minor hemorrhage hemarthroses, superficial muscle hemorrhage, soft tissue hemorrhage, moderate hemorrhage, intramuscle or soft tissue hemorrhage with dissection, mucous membrane hemorrhage, hematuria, major hemorrhage, hemorrhage of the pharynx, hemorrhage of the retropharynx, hemorrhage of the retroperitonium, hemorrhage, of the central bruises, cuts, scrapes, joint hemorrhage, nose bleed, intracranial bleeding, intraperitoneal bleeding, minor spontaneous hemorrhage, bleeding after major trauma, moderate skin bruising, or spontaneous hemorrhage into joints, muscles, internal organs or the brain. Additional reasons for on-demand treatment include the need for peri-operative management for surgery or dental extraction, major surgery, extensive oral surgery, urologic surgery, hernia surgery, orthopedic surgery such as replacement of knee, hip, or other major joint.

The term “treatment” or “treating” as used herein means amelioration or reduction of one or more symptoms of bleeding diseases or disorders including, but not limited to, hemophilia A or hemophilia B. In one embodiment, “treatment of” or “treating” a bleeding disease or disorder includes prevention of one or more symptoms of a bleeding disease or disorder. In a bleeding disease or disorder caused by a clotting factor deficiency (e.g., a low baseline clotting activity), the term “treatment” or “treating” means a clotting factor replacement therapy. By administering a clotting factor to a subject, the subject can achieve and/or maintain: a plasma trough level of a clotting activity at about 1 IU/dl or above 1 IU/dl. In other embodiments, “treatment” or “treating” means reduction of the frequency of one or more symptoms of bleeding diseases or disorders, e.g., spontaneous or uncontrollable bleeding episodes. “Treatment,” however, need not be a cure.

The term “perioperative management” as used herein means use of a clotting factor before, concurrently with, or after an operative procedure, e.g., a surgical operation. The use for “perioperative management” of one or more bleeding episode includes surgical prophylaxis before: (i.e., preoperative), during (L e., intraoperative), or after (i.e., postoperative) a surgery to prevent One or more bleeding or bleeding episode or reducing or inhibiting spontaneous and/or uncontrollable bleeding episodes before, during, and after a surgery.

Pharmacokinetic (PK) parameters include the terms above and the following terms, which have their ordinary meaning in the art, unless otherwise indicated. Some of the terms are explained in more detail in the Examples. PK parameters can be based on clotting factor antigen level (often denoted parenthetically herein as “antigen”) or clotting activity level (often denoted parenthetically herein as “activity”). In the literature, PK parameters are often based on clotting activity level due to the presence in the plasma of some patients of endogenous, inactive clotting factor, which interferes with the ability to measure administered (i.e., exogenous) clotting factor using antibody against clotting factor. However, when a clotting factor is administered, clotting factor antigen can be accurately measured using antibody to the heterologous polypeptide. In addition, certain PK parameters can be based on model predicted data (often denoted parenthetically herein as “model predicted”) or on observed data (often denoted parenthetically herein as “observed”).

“Baseline,” as used herein, is the lowest measured plasma clotting factor level in a subject prior to administering a dose. The clotting factor plasma levels can be measured at two time points prior to dosing: at a screening visit and immediately prior to dosing. Alternatively, (a) the baseline in patients whose pretreatment clotting activity is <1% of normal, who have no detectable clotting factor antigen, and have nonsense genotypes can be defined as 0%, (b) the baseline for patients with pretreatment clotting activity <1% of normal and who have detectable clotting factor antigen can be set at 0.5%, (c) the baseline for patients whose pretreatment clotting activity is between 1-2% is Cmin (the lowest activity throughout the PK study), and (d) the baseline for patients whose pretreatment clotting activity is ≥2% can be set at 2% of normal. Activity above the baseline pre-dosing can be considered residue drug from prior treatment, and can be decayed to baseline and subtracted from the PK data following clotting factor dosing.

“Trough,” as used herein, is the lowest plasma clotting activity level reached after administering a dose of a clotting factor molecule (e.g., chimeric clotting factor) and before the next dose is administered, if any. Trough is used interchangeably herein with “threshold.” Baseline clotting factor levels are subtracted from measured clotting factor levels to calculate the trough level.

“Subject,” as used herein means a mammal. The subject can be a human, e.g., a human patient. Subject as used herein includes an individual who is known to have at least one incidence of uncontrolled bleeding episodes, who has been diagnosed with a disease or disorder associated with uncontrolled bleeding episodes, e.g., a bleeding disease or disorder, e.g., hemophilia A or hemophilia B, who are susceptible to uncontrolled bleeding episodes, e.g., hemophilia, or any combinations thereof. Subjects can also include an individual who is in danger of one or more uncontrollable, bleeding episodes prior to a certain activity, e.g., a surgery, a sport activity, or any strenuous activities. The subject can have a baseline clotting activity less than 1%, less than 0.5%, less than 2%, less than 2.5%, less than 3%, or less than 4%.

“Variant,” as used herein, refers to a polynucleotide or polypeptide differing from the original polynucleotide or polypeptide, but retaining essential properties thereof, e.g., clotting 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 polypeptide and polynucleotide fragments, deletions, insertions, and modified versions of original polypeptides.

II. Clotting Factors

The present invention is directed to a clotting factor suitable for a fixed dosing regimen. A suitable dosing strategy can be identified for a particular drug based on its pharmacokinetic (PK) and/or pharmacodynamic (PD) properties. For example, a good drug candidate for a fixed dosing strategy provides more consistent exposure of the drug across subjects when administered by a fixed dosing regimen rather than by a body weight based dosing regimen. Advantages of the present invention include: improved regimen compliance; reduced break through bleeds; increased protection of joints from bleeds; prevention of joint damage; reduced morbidity; reduced mortality; prolonged protection from bleeding; decreased thrombotic events; and improved quality of life. In one embodiment, a clotting factor suitable for a fixed dosing regimen exhibits a wide therapeutic window. In another embodiment, a clotting factor suitable for a fixed dosing regimen has less inter-individual variability of pharmacokinetic parameters (e.g., AUC or Cmax) when administered by a fixed dosing regimen compared to the inter-individual variability of pharmacokinetic parameters when administered by a body-weight based dosing regimen. In another embodiment, a clotting factor suitable for a fixed dosing regimen has inter-individual variability of pharmacokinetic parameters (e.g., AUC or Cmax) that is similar when administered either by a fixed dosing regimen or by a body-weight based dosing regimen.

In one aspect, the pharmaceutical properties of a clotting factor suitable for a fixed dosing regimen can be represented by the following formulas:


AUC=Dose/CL   (C)


CL=Typical CL×(BW/Typical BW)exponent   (D)


Cmax=Dose/V   (E)


V=Typical V×(BW/TypicalBW)exponent   (F)

The exponent for formula (D) indicates a body weight effect on clearance (θBW_CL). The exponent for formula (F) indicates a body weight effect on the central volume of distribution (θBW_V1). In another aspect of the invention, the body weight effect on clearance (θBW_CL) of the clotting factor is equal to or less than 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, 0.67, 0.66, 0.65, 0.64, 0.63, 0.62, 0.61, 0.6, 0.59, 0.58, 0.57, 0.56, 0.55, 0.54, 0.53, 0.52, 0.51, 0.50, 0.49, 0.48, 0.47, 0.46, 0.45, 0.44, 0.43, 0.42, 0.41, 0.40, 0.39, 0.38, 0.37, 0.36, 0.35, 0.34, 0.32, 0.31, 0.3, 0.25, 0.2, 0.15, 0.1, 0.5, or 0. In other aspects, the body weight effect on the central volume of distribution (θBW_V1) of the clotting factor is equal to or less than 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, 0.67, 0.66, 0.65, 0.64, 0.63, 0.62, 0.61, 0.6, 0.59, 0.58, 0.57, 0.56, 0,55, 0.54, 0.53, 0.52, 0.51, 0.50, 0.49. 0.48, 0.47, 0.46, 0.45, 0.44, 0.43, 0.42, 0.41, 0.40, 0.39, 0.38, 0.37, 0.36, 0.35, 0,34, 0.32, 0.31, 0.3, 0.25, 0.2, 0.15, 0.1, 0.5, or 0. In still other aspects, θBW_CL is equal to or less than 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, 0.67, 0.66, 0.65, 0.64, 0.63, 0.62, 0.61, 0.6, 0.59, 0.58, 0.57, 0.56, 0.55, 0.54, 0.53, 0.52, 0.51, 0.50, 0.49, 0.48, 0.47, 0.46, 0.45, 0.44, 0.43, 0.42, 0.41, 0.40, 0.39, 0.38, 0.37, 0.36, 0.35, 0.34, 0.32, 0.31, or 0.3, 0.25, 0.2, 0.15, 0.1, 0.5, or 0 and θBW_V1 is equal to or less than 0.75, 0.74, 0.73, 0.72, 0.71, 0.70, 0.69, 0.68, 0.67, 0.66, 0.65, 0.64, 0.63, 0.62, 0.61, 0.6, 0.59, 0.58, 0.57, 0.56, 0.55, 0.54, 0.53, 0.52, 0.51, 0.50, 0.49, 0.48, 0.47, 0.46, 0.45, 0.44, 0.43, 0.42, 0.41, 0.40 0.39, 0.38, 0.37, 0.36, 0.35, 0.34, 0.32, 0.31, or 0.3, 0.25, 0.2, 0.15, 0.1, 0.5, or 0. In some aspects, the clotting factor has θBW_CL equal to or less than about 0.500 and θBW_V1 equal to or less than 0.467. In other embodiments, the clotting factor has θBW_CL equal to about 0 and θBW_V1 equal to or less than 0.492. In other embodiments, the clotting factor has θBW_CL equal to or less than about 0.436 and θBW_V1 equal to or less than about 0.396.

In certain aspects, a clotting factor is administered to a population of two or more subjects. In some aspects, the area under curve (AUC) or Cmax between a high extreme body weight subject and a low extreme body weight subject after administration of the fixed dosing of the clotting factor is similar to or less variable than AUC or Cmax between a high body weight subject and a low body weight, after administration of a body weight-based dosing amount of the clotting factor. In one embodiment, the variability in AUC is less than ±50%, less than ±45%, less than ±40%, less than +35%, less than ±30%, or less than ±25%. In another embodiment, the variability in Cmax is less than ±50%, less than ±45%, less than ±40%, less than ±35%, less than ±30%, or less than ±25%.

In other aspects, the clotting factor has a wide therapeutic window. In one embodiment, the therapeutic window for the clotting factor comprises a maximum serum concentration (Cmax) of about 150% of normal and a minimum serum concentration (Cmin) of about 1% of normal. In still other aspects, the body weight of the subject does not drive pharmacodynamic variability when administered by a fixed dosing regimen based dosing regimen. In some aspects, a clotting factor of the invention has low or no off-target toxicity. In certain aspects, a clotting factor is cleared primarily through cellular uptake in liver.

In some aspects, a clotting factor has less pharmacokinetic inter-subject variability than a clotting factor suitable for a body-weight based dosing. In one embodiment, the inter-subject variability is about 20% to about 50%, about 21% to about 49%, about 22% to about 48%, about 23% to about 47%, about 24% to about 46%, about 25% to about 46%, about 26% to about 46% for total clearance (CL) and Volume of Distribution at Steady State (Vss).

A clotting factor suitable for a fixed dosing strategy can be a wild-type clotting factor, a commercially available clotting factor, or a modified clotting factor. Examples of the wild-type clotting factors include, but are not limited to, full-length recombinant FIX (e.g., BENEFIX®), plasma-derived FIX (e.g., ALPHANINE®, or MONONINE®), or full-length recombinant FVIII (e.g., RECOMBINATE®, ADVATE®, or HELIXATE®), or B-domain deleted recombinant FVIII (e.g., REFACTO® or XYNTHA®).

Clotting factors for the invention can be modified. Modified clotting factors includes any clotting factors that have improvements in one or more aspects, pharmacokinetics (PK), pharmacodynamics (PD), stability, expression, or any combinations thereof In one embodiment, a modified clotting factor comprises a clotting factor and a heterologous moiety. In another embodiment, a clotting factor for the invention is a long-acting clotting factor. Long-acting clotting factors can comprise a heterologous moiety that increases in vivo half-life of the clotting factor. In other embodiments, the heterologous moiety for the modified clotting factor (e.g., long-acting clotting factor) is a polypeptide moiety or a non-polypeptide moiety. In yet other embodiments, a heterologous moiety comprises albumin, albumin binding polypeptide, an FcRn binding partner, PAS, the C-terminal peptide (CTP) of the β subunit of human chorionic gonadotropin, polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin-binding small molecules, or combinations thereof. Examples of the heterologous moieties are described below. In some embodiments, a clotting factor of the invention is a chimeric clotting factor. In some embodiments, the heterologous moiety is linked to the N-terminus or the C-terminus of the FVIII polypeptide or inserted between two amino acids of the FVIII polypeptide.

A. Factor IX

In certain embodiments, a clotting factor of the invention is a modified FIX polypeptide. In one example, a modified clotting factor useful for the invention comprises a long-acting FIX polypeptide, which is a chimeric polypeptide comprising a FIX polypeptide and an FcRn binding partner. The FIX polypeptide of the invention comprises a functional Factor IX polypeptide in its normal role in coagulation, unless otherwise specified. Thus, the FIX polypeptide includes variant polypeptides that are functional and the polynucleotides that encode such functional variant polypeptides. In one embodiment, the FIX polypeptides are the human, bovine, porcine, canine, feline, and, murine FIX polypeptides. The full-length polypeptide and polynucleotide sequences of FIX are known, as are many functional variants, e.g., fragments, mutants and modified versions. FIX polypeptides include full-length FIX, full-length FIX minus Met at the N-terminus, full-length FIX minus the signal sequence, mature FIX (minus the signal sequence and propeptide), and mature FIX with an additional Met at the N-terminus. FIX can be made by recombinant means (“recombinant Factor IX” or “rFIX”), i.e., it is not naturally occurring or derived from plasma.

A great many functional FIX variants are known. International publication number WO 02/040544 A3, which is herein incorporated by reference in its entirety, discloses mutants that exhibit increased resistance to inhibition by heparin at page 4, lines 9-30 and page 15, lines 6-31. International publication number WO 03/020764 A2, which is herein incorporated by reference in its entirety, discloses FIX mutants with reduced T cell immunogenicity in Tables 2 and 3 (on pages 14-24), and at page 12, lines 1-27. International publication number WO 2007/149406 A2, which is herein incorporated by reference in its entirety, discloses functional mutant FIX molecules that exhibit increased protein stability, increased in vivo and in vitro half-life, and increased resistance to proteases at page 4, line 1 to page 19, line 11. WO 2007/149406 A2 also discloses chimeric and other variant FIX molecules at page 19, line 12 to page 20, line 9. International publication number WO 08/118507 A2, which is herein incorporated by reference in its entirety, discloses FIX mutants that exhibit increased clotting activity at page 5, line 14 to page 6, line 5. International publication number WO 09/051717 A2, which is herein incorporated by reference in its entirety, discloses FIX mutants having an increased number of N-linked and/or 0-linked glycosylation sites, which results in an increased half-life and/or recovery at page 9, line 11 to page 20,=line 2. International publication number WO 09/137254 A2, which is herein incorporated by reference in its entirety, also discloses Factor IX mutants with increased numbers of glycosylation sites at page 2, paragraph [006] to page 5, paragraph [011] and page 16, paragraph [044] to page 24, paragraph [057]. International publication number WO 09/130198 A2, which is herein incorporated by reference in its entirety, discloses functional mutant FIX molecules that have an increased number of glycosylation sites, which result in an increased half-life, at page 4, line 26 to page 12, line 6. International publication number WO 09/140015 A2, which is herein incorporated by reference in its entirety, discloses functional FIX mutants that an increased number of Cys residues, which may be used for polymer (e.g., PEG) conjugation, at page 11, paragraph [0043] to page 13, paragraph [0053]. The FIX polypeptides described in International Application No. PCT/US2011/043569 filed Jul. 11, 2011 and published as WO 2012/006624 on Jan. 12, 2012 are also incorporated herein by reference in its entirety.

In addition, hundreds of non-functional mutations in FIX have been identified in hemophilia patients, many of which are disclosed in Table 1, at pages 11-14 of International publication number WO 09/137254 A2, which is herein incorporated by reference in its entirety. Such non-functional: mutations are not included in the invention, but provide additional guidance for which mutations are more or less likely to result in a functional FIX polypeptide.

In one embodiment, the Factor IX (or Factor IX portion of a chimeric polypeptide) may be at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a FIX amino acid sequence shown in Table 5A without a signal sequence and propeptide sequence (amino acids 1 to 415 of SEQ ID NO:2), or alternatively, with a propeptide sequence, or with a propeptide and signal sequence (full-length FIX).

Factor IX coagulant activity is expressed as International Unit(s) (IU). One IU of FIX activity corresponds approximately to the quantity of FIX in one milliliter of normal human plasma. Several assays are available for measuring Factor IX activity, including the one stage clotting assay (activated partial thromboplastin time; aPTT), thrombin generation time (TGA) and rotational thromboelastometry (ROTEM®).

A chimeric polypeptide comprising a FIX polypeptide and an FcRn binding partner can comprise an amino acid sequence at least 60%, at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the Factor IX and FcRn BP, e.g., the Fc amino acid sequence shown in Table 5A without a signal sequence and propeptide sequence (amino acids 1 to 642 of SEQ ID NO:2), or alternatively, with a propeptide sequence, or alternatively with a signal sequence and a propeptide sequence.

A long-acting or long-lasting FIX polypeptide can be a hybrid FIX polypeptide. Hybrid FIX polypeptide means a combination of a FIX chimeric polypeptide with a second polypeptide. The chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via non-covalent 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 covalent bond(s) such as disulfide bonds. The chimeric peptide and the second peptide may be associated with each other via more than one type of bond, such as non-covalent and disulfide bonds. Hybrids are described in WO 2004/101740, WO2005/001025, U.S. Pat. No. 7,404,956, U.S. Pat. No. 7,348,004, and WO 2006/074199, 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. In other embodiments, the second polypeptide is a polypeptide comprising an FcRn BP, e.g., Fc. In some embodiments, the chimeric polypeptide is a Factor IX-FcRn BP, e.g., Factor IX-Fc chimeric polypeptide, and the second polypeptide consists essentially of Fc. See, e.g., Table 5A and B (SEQ ID NOs:2 and 4). See, e.g., U.S. Pat. No. 7,404,956, which is incorporated herein by reference in its entirety.

The second polypeptide in a hybrid may comprise or consist essentially of a sequence at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the amino acid sequence shown in Table 5B without a signal sequence (SEQ ID NO:4), or alternatively, with a signal sequence.

In some embodiments, a long-acting FIX polypeptide is a FIX monomer dimer hybrid. Monomer-dimer hybrid can comprise two polypeptide chains, one chain comprising a FIX polypeptide and a first Fc region, and another chain comprising, consisting essentially of, or consisting of a second Fc region. In certain aspects, a FIX monomer dimer hybrid consists essentially of or consists of two polypeptide chains, a first chain consisting essentially of or consisting of a FIX polypeptide and a second chain consisting essentially of or consisting of a second Fc region.

A long-acting FIX polypeptide can be encoded by 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:1 or 3 (the Factor IX portion, the Fc portion, individually or together) or the complementary strand thereto, the nucleotide coding sequence of known mutant and recombinant Factor IX 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 or 4 (the Factor IX 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.

B. Factor VIII

In some embodiments, a clotting factor for the invention is a modified FVIII polypeptide. In one aspect, a modified FVIII polypeptide is a long-acting FVIII polypeptide. In another aspect, a long-acting FVIII polypeptide comprises a FVIII polypeptide and an FcRn binding partner. The FVIII polypeptide 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. In addition, 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 NO: 6 or 8 (Table 7A and 7B). Factor VIII polypeptides include, e.g., full-length factor VIII, fall-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.

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. 19:274-8 (2002)), incorporated herein by reference in its entirety. In addition, 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); U.S. Pat. No. 6,251,632), incorporated herein by reference in its entirety.

The human factor VIII gene was isolated and expressed in mammalian cells (Toole, J. J., et al., Nature 312:342-347 (1984); Gitschier, J., et al., Nature 312:326-330 (1984); Wood, W. I., et al., Nature 312:330-337 (1984); Vehar, G. A., et al., Nature 312:337-342 (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 NO: 3B and 4B, respectively, of US Application Publ. No. 2005/0100990, incorporated herein by reference in its entirety.

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. U.S. 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.

A number of functional factor VIII molecules, including B-domain deletions, are disclosed in the following patents U.S. Pat. No. 6,316,226 and U.S. Pat. No. 6,346,513, both assigned to Baxter; U.S. Pat. No. 7,041,635 assigned to In2Gen; U.S. Pat. No. 5,789,203, U.S. Pat. No. 6,060,447, U.S. Pat. No. 5,595,886, and U.S. Pat. No. 6,228,620 assigned to Chiron; U.S. Pat. No. 5,972,885 and U.S. Pat. No. 6,048,720 assigned to Biovitrum, U.S. Pat. No. 5,543,502 and U.S. Pat. No. 5,610,278 assigned to Novo Nordisk; U.S. Pat. No. 5,171,844 Immuno Ag; U.S. Pat. No. 5,112,950 assigned to Transgene S.A.; U.S. Pat. No. 4,868,112 assigned to Genetics Institute, each of which is incorporated herein by reference its entirety.

The porcine factor VIII sequence is published, (Toole, J. J., et al., Proc. Natl. Acad. Sci. USA 83:5939-5942 (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 88:4209-4214 (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 A1 and A2 domains of porcine factor VIII and a chimeric factor VIII with porcine A1 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. U.S. Pat. No. 6,458,563, incorporated herein by reference in its entirety assigned to Emory discloses a B-domain deleted porcine Factor VIII.

The factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% identical to a Factor VIII amino acid sequence shown in Tables 7A and 7B without a signal sequence (amino acids 20 to 1457 of SEQ ID NO: 6; and amino acids 20 to 2351 of SEQ ID NO: 8), wherein said Factor VIII portion has Factor VIII activity. The Factor VIII (or Factor VII portion of a chimeric polypeptide) may be identical to a Factor VIII amino acid sequence show in Tables 7A and 7B without a signal sequence (amino acids 20 to 1457 of SEQ ID NO: 6; and amino acids 20 to 2351 of SEQ ID NO: 8).

The Factor VIII (or Factor VIII portion of a chimeric polypeptide) may be at least 60%, at least 70%, at least 80%, at >least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or at least 100% identical to a Factor VIII amino acid sequence shown in Tables 7A and 7B with a signal sequence (amino acids 1 to 1457 of of SEQ ID NO: 6 and amino acids 1 to 2351 of SEQ ID NO: 8), wherein the Factor VIII 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 Tables 7A and 7B with a signal sequence (amino acids 1 to 1457 of SEQ ID NO: 6 and amino acids 1 to 2351 of SEQ ID NO: 8).

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-Arg1648 of full-length human factor VIII. The other human factor VIII domains are defined by the following amino acid residues: A1, residues Ala1-Arg372; A2, residues Ser373-Arg740; A3, residues Ser1690-Ile2032; C1, residues Arg2033-Asn2172; C2, residues Ser2173-Tyr2332. The A3-C1-C2 sequence includes residues Ser1690-Tyr2332. The remaining sequence, residues Glu1649-Arg1689, 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 SQ BDD FVIII), which has the same sequence as the Factor VIII portion of the sequence in Table 7A (amino acids 1 to 1457 or 20 to 1457 of SEQ ID NO:6). 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: 6, or residue 1648 of full-length Factor VIII, which corresponds to Arginine residue 1667 of SEQ ID NO: 8. 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 5762/Q1657 of SEQ ID NO: 8 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.

A “B domain deleted factor VIII” may have the full or partial deletions disclosed in U.S. Pat. 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. Pat. No. 6,316,226 (also in U.S. Pat. No. 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. Pat. No. 5,789,2.03 (also U.S. Pat. No. 6,060,447, U.S. Pat. No. 5,595,886, and U.S. Pat. No. 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 U.S. Pat. No. 5,972,885; col. 6, lines 1-22 and example 1 of U.S. Pat. No. 6,048,720; col. 2, lines 17-46 of U.S. Pat. No. 5,543,502; col. 4, line 22 to col. 5, line 36 of U.S. Pat. No. 5,171,844; col. 2, lines 55-68, FIG. 2, and example 1 of U.S. Pat. No. 5,112,950; col. 2, line 2 to col. 19, line 21 and table 2 of U.S. Pat. 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. Pat. No. 7,041,635; or col. 4, lines 25-53, of U.S. Pat. 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. 83:5939-5942 (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.

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 (A1-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 or unprocessed FVIII. The single chain Factor VIII can comprise an intracellular processing site, which corresponds to Arginine at residue 754 of B domain deleted Factor VIII (i.e., residue 773 of SEQ ID NO: 6) or at residue 1648 of full-length Factor VIII (i.e., residue 1657 of SEQ ID NO: 8).

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+, Mn2+, or Cu2+. Fatouros et al., Intern. J Pharm. 155(1): 121-131 (1997); Wakabayashi et al., JBC. 279(13): 12677-12684 (2004).

In other embodiments, a FVIII polypeptide of the invention is processed FVIII comprising a light chain and a heavy chain of FVIII. In yet other embodiments, a FVIII polypeptide is single chain FVIII. In other embodiments, the single chain 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.

In one embodiment, a chimeric polypeptide comprising a FVIII polypeptide and an FcRn binding partner can comprise an amino acid sequence at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the Factor VIII and FcRn BP, e.g., the Fc amino acid sequence shown in Table 5B without a signal sequence (SEQ ID NO:4).

In another embodiment, a chimeric polypeptide comprising a FVIII polypeptide and an FcRn binding partner can comprise an amino acid sequence at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the Factor VIII and FcRn BP, e.g., the Fc amino acid sequence shown in Table 5B without a signal sequence (SEQ ID NO:4).

A long-acting or long-lasting FVIII polypeptide can be a hybrid FVIII polypeptide. Hybrid FVIII polypeptide means a combination of a FVIII chimeric polypeptide with a second polypeptide. The chimeric polypeptide and the second polypeptide in a hybrid may be associated with each other via non-covalent 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 covalent bond(s) such as disulfide bonds. The chimeric peptide and the second peptide may be associated with each other via more than one type of bond, such as non-covalent and disulfide bonds. The second polypeptide may be a second copy of the same chimeric polypeptide or it may be a non-identical chimeric polypeptide. In other embodiments, the second polypeptide is a polypeptide comprising an FcRn BP, e.g., Fc. In some embodiments, the chimeric polypeptide is a Factor VIII-FcRn BP, e.g., Factor VIII-Fc chimeric polypeptide, and the second polypeptide consists essentially of Fc.

The second polypeptide in a hybrid may comprise or consist essentially of a sequence at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, <or 100% identical to the amino acid sequence shown in Table 5B without a signal sequence (SEQ ID NO:4), or alternatively, with a signal sequence.

In some embodiments, a long-acting FVIII polypeptide is a FVIII monomer dimer hybrid. Monomer-dimer hybrids can comprise two polypeptide chains, one chain comprising a FVIII polypeptide and a first Fc region, and another chain comprising, consisting essentially of, or consisting of a second Fc region. In certain aspects, a FVIII monomer dimer hybrid consists essentially of or consists of two polypeptide chains, a first chain consisting essentially of or consisting of a polypeptide and a second chain consisting essentially of or consisting of a second Fc region.

In some embodiments, a long-acting FVIII polypeptide comprises a FVIII polypeptide and at least one heterologous moiety, which increases in vivo half-life of the FVIII polypeptide, wherein the at least one heterologous moiety is linked to the C-terminus or N-terminus of the FVIII polypeptide or inserted between two amino acids of the FVIII polypeptide.

A long-acting FVIII polypeptide can be encoded by 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:5 or 7 (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:6 or 8 (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.

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:1 or 3, the ORF (open reading frame), or any fragment specified as described herein.

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, 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. (1990) 6:237-245), 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 certain embodiments, the parameters used in a FASTDB alignment of DNA sequences to calculate percent identity are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=1, Joining Penalty=30, Randomization Group Length=0, Cutoff Score=1, Gap Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the length of the subject nucleotide sequence, whichever is shorter.

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.

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.

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, 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.

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 NOs:2, 4, 6, or 8 (the FIX portion, the FVIII portion, the Fc portion, individually or together), or a known FIX, FVIII, or Fc polypeptide sequence, can be determined conventionally using known computer programs. In one embodiment, 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 certain embodiments, the parameters used in a FASTDB amino acid alignment are: Matrix PAM=0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=1, 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.

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.

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.

The polynucleotide variants may contain alterations in the coding regions, non-coding regions, or both. Certain embodiments include polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants can be produced by silent substitutions due to the degeneracy of the genetic code. Moreover, variants in which 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are included. 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 those preferred by 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.

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. The authors of 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, of 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)

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. 268:22105-22111 (1993), incorporated herein by reference in its entirety) conducted extensive mutational analysis of human cytokine IL-1a. They used random mutagenesis to generate over 3,500 individual IL-1a 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.

As stated above, polypeptide variants include modified polypeptides. Modifications include 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, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.

C. Half-Life Extension

In certain aspects, a modified clotting factor of the invention comprises at least one heterologous moiety which increases the in vivo half-life of the protein. In vivo half-life of a modified clotting factor can be determined by any method known to those of skill in the art, e.g., clotting activity assays (chromogenic assay or one stage clotting aPTT assay) to detect plasma clotting activity levels or ELISA to detect plasma clotting factor antigen level.

In certain aspects, a heterologous moiety which increases in vivo half-life of the modified clotting factor of the invention can comprise, without limitation, heterologous polypeptide such as albumin, an immunoglobulin Fc region, the β subunit of the C-terminal peptide (CTP) of human chorionic gonadotropin, a PAS sequence, a HAP sequence, a transferrin, albumin-binding moieties, or any fragments, derivatives, variants, or combinations of these polypeptides. In certain aspects the modified clotting factor of the invention comprises a heterologous polypeptide which increases in vivo half-life. In other related aspects a heterologous moiety can include an attachment site for a non-polypeptide moiety such as polyethylene glycol (PEG), hydroxyethyl starch (HES), polysialic acid, or any derivatives, variants, or combinations of these elements.

In other embodiments, a modified clotting factor of the invention is conjugated to one or more polymers. The polymer can be water-soluble or non-water-soluble. The polymer can be covalently or non-covalently attached to a clotting factor or to other moieties conjugated to a clotting factor. Non-limiting examples of the polymer can be poly(alkylene oxide), poly(vinyl pyrrolidone), poly(vinyl alcohol), polyoxazoline, or poly(acryloylmorpholine). Additional types of polymer-conjugated clotting factor are disclosed in U.S. Pat. No. 7,199,223, which is disclosed by reference in its entirety.

In certain aspects, a modified clotting factor of the invention can comprise one, two, three or more heterologous moieties, which can each be the same or different molecules.

D. FcRn Binding Partner

FcRn binding partner (“FcRn BP”) comprises 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 FcRn BP includes any variants of IgG Fc that are functional. For example, 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. 1994, Nature 372:379, 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. FcRn BPs include 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. 1994, J. Exp. Med. 180: 2377), incorporated herein by reference in its entirety.) An FcRn BP may comprise the CH2 and CH3 domains of an immunoglobulin with or without the hinge region of the immunoglobulin. In a particular embodiment, an FcRn BP is an Fc region. Exemplary FcRn BP variants are provided in WO 2004/101740 and WO 2006/074199, incorporated herein by reference in its entirety.

FcRn BP (or FcRn BP portion of a chimeric polypeptide) may contain one or more mutations, and combinations of mutations.

FcRn BP (or FcRn BP portion of a chimeric polypeptide) may contain mutations conferring, increased half-life such as M252Y, S2541, 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 U.S. 2009/0264627 A1, which is incorporated herein by reference in its entirety; and the mutants disclosed at page 2, paragraphs [0014] to [0021] of U.S. 20090163699 A1, which is incorporated herein by reference in its entirety.

FcRn BP (or FcRn BP portion of a chimeric polypeptide) may also include 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 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 IgG1 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, Q311A, D312A, N315A, K317A, E318A, K320A, K322A, S324A, K326A, A327Q, P329A, A330Q, A330S, P331A, P331S, 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 wild type proline substituted by alanine at position number 238. In addition to alanine other amino acids may be substituted for the wild type 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 IgG1 “ELLG” to the corresponding sequence from IgG2 “PVA” (with one amino acid deletion). It has been bind to IgG1 when such mutations have been introduced (Ward and Ghetie 1995, Therapeutic Immunology 2:77, which is incorporated herein by reference in its entirety; and Armour et al. 1999, Eur. J. Immunol. 29:2613, 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 T256A, T307A, E380A, and N434A (Shields et al. 2001, J. Biol. Chem. 276:6591, which is incorporated herein by reference in its entirety).

The FcRn BP (or FcRn BP portion of a chimeric polypeptide) may be at least 70%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the Fc amino acid sequence shown in Table 5B without a signal sequence (SEQ ID NO:4), or alternatively, with a signal sequence.

Myriad mutants, fragments, variants, and derivatives are described, e.g., in PCT Publication Nos. WO 2011/069164 A2, WO 2012/006623 A2, WO 2012/006635 A2 , or WO 2012/006633 A2, all of which are incorporated herein by reference in their entireties.

E. Albumins

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one albumin polypeptide or fragment, variant, or derivative thereof, wherein the modified clotting factor has procoagulant activity and can be expressed in vivo or in, vitro in a host cell. Human serum albumin (HSA, or HA), a protein of 609 amino acids in its full-length form, is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. The term “albumin” as used herein includes full-length albumin or a functional fragment, variant, derivative, or analog thereof. Examples of albumin or the fragments or variants thereof are disclosed in US Pat. Publ. Nos. 2008/0194481A1, 2008/0004206 A1, 2008/0161243 A1, 2008/0261877 A1, or 2008/0153751 A1 or PCT Appl. Publ. Nos. 2008/033413 A2, 2009/058322 A1, or 2007/021494 A2, which, are incorporated herein by reference in their entireties.

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:18). 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 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.

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one attachment site for a non-polypeptide small molecule, variant, or derivative that can bind to albumin thereof For example, a modified clotting factor of the invention can include one or more organic albumin binding moieties attached to the clotting factor. An example of such albumin binding moieties is 2-(3-maleimidopropanamido)-6-(4-(4-iodophenyl)butanamido)hexanoate (“Albu” tag) as disclosed by Trusse et al., Bioconjugate Chem. 20:2286-2292 (2009).

F. CTP

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one C-terminal peptide (CTP) of the β subunit, of human chorionic gonadotropin or fragment, variant, or derivative thereof. One or more CTP peptides fused to or inserted into a clotting factor is known to increase the in vivo half-life of that protein. See, e.g., U.S. Pat. No. 5,712,122, incorporated by reference herein in its entirety. Exemplary CTP peptides include DPRFQDSSSSKAPPPSLPSPSRLPGPSDTPIL (SEQ ID NO:9) or SSSSKAPPPSLPSLPSRLPGPSDTPILPQ. (SEQ ID NO:10). See, e.g., U.S. Patent Application Publication No. U.S. 2009/0087411 A1, incorporated by reference.

G. PAS

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one PAS peptide or fragment, variant, or derivative thereof. 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, 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% 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, Glu, Gly, His, Ile, 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.

Non-limiting examples of the PAS peptides include ASPAPAPASPAAPAPSAPA (SEQ ID NO: 11), AAPASPAPAAPSAPAPAAPS (SEQ ID NO:12), APSSPSPSAPSSPSPASPSS (SEQ ID NO:13), APSSPSPSAPSSPSPASPS (SEQ ID NO:14), SSPSAPSPSSPASPSPSSPA (SEQ ID NO:15), AASPAAPSAPPAAASPAAPSAPPA (SEQ 16), ASAAAPAAASAAASAPSAAA (SEQ ID NO:17) or any variants, derivatives, fragments, or combinations thereof. Additional examples of PAS sequences are known from, e.g., U.S. Pat. Publ. No. 2010/0292130 A1 and PCT Appl. Publ. No. WO 2008/155134 A1 European issued patent EP2173890.

H. HAP

In certain aspects, a modified clotting factor :of the invention comprises a clotting factor and at least one homo-amino acid polymer (HAP) peptide or fragment, variant, or derivative thereof. A HAP peptide can comprise a repetitive sequence of glycine, which has at least 50 amino acids, at least 100 amino acids, 120 amino acids; 140 amino acids, 160 amino acids, 180 amino acids, 200 amino acids, 250 amino acids; 300 amino acids, 350 amino acids, 400 amino acids, 450 amine acids, or 500 amino acids in length. A HAP sequence capable of extending half-life of a moiety fused to or linked to the HAP sequence. Non-limiting examples of the HAP sequence includes, but are not limited to (Gly)n, (Gly4Ser)n or S(Gly4Ser)n, wherein n is 1, 2, 3, 4, 5, 6, 7; 8, 9, 10, 11, 12 13, 14, 15, 16, 17, 18, 19, or 20. In one embodiment, n is 20, 21, 22, 23, 24, 25, 26, 26, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, or 40. In another embodiment, n is 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200. See, e.g., Schlapschy M et al., Protein Eng. Design Selection, 20: 273-284 (2007).

I. Transferrin

In certain aspects, a modified clotting factor of the invention comprises at least one transferrin peptide or fragment, variant, or derivative thereof linked to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity. Any transferrin can be linked to or inserted into a modified clotting factor 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 595936 (www.ncbi.nlm.nih.gov), all of which are herein incorporated by reference in, their entirety.

Transferrin transports iron through transferrin receptor (TfR)-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)).

J. PEG

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one attachment site for a non-polypeptide heterologous moiety or fragment, variant, or derivative thereof linked to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity. For example, a modified clotting factor of the invention can include one or more polyethylene glycol (PEG) moieties attached to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity.

PEGylated clotting, factor can refer to a conjugate formed between clotting factor 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 modified clotting factor 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.

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. WO92/16221 and WO95/34326. As a non-limiting example, clotting factor variants can contain cysteine substitutions in one or more permissive loops as described herein, and the cysteines can be further conjugated to PEG polymer. See Mei, et al., Blood 116:270-279 (2010) and U.S. Pat. No. 7,632,921, which are incorporated herein by reference in their entireties.

K. HES

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one hydroxyethyl starch (HES) polymer conjugated to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity. 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).

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 200 kD, from 3 to 100 kD, or from 4 to 70 kD. 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.

L. PSA

In certain aspects, a modified clotting factor of the invention comprises a clotting factor and at least one polysialic acid (PSA) polymer conjugated to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity. PSAs are naturally occurring unbranched polymers of sialic acid produced by certain bacterial strains and in mammals in certain cells. See, e.g., Roth J. et al. (1993) in Polysialic Acid: From Microbes to Man, eds. Roth J., Rutishauser U., Troy F. A. (BirkhauserVerlag, Basel, Switzerland), pp. 335-348. PSAs can be produced in various degrees of polymerization from n=about 80 or more sialic acid residues down to n=2 by limited acid hydrolysis or by digestion with neuraminidases, or by fractionation of the natural, bacterially derived forms of the polymer. There are a number of PSA attachment methods available to those skilled in the art, e.g., the same PEG attachment methods described above. In certain aspects, an activated PSA can also be attached to a cysteine amino acid residue on the clotting factor. See, e.g., U.S. Pat. No. 5,846,951.

M. Clearance Receptors

In certain aspects, the in Vivo half-life of a modified clotting factor of the invention can be, extended where the modified clotting factor comprises at least one fragment of a clotting factor clearance receptor or fragment, variant, or derivative thereof linked to or inserted into the clotting factor, wherein the modified clotting factor has procoagulant activity. For example, insertion of soluble forms of clearance receptors, such as the low density lipoprotein-related protein receptor LRP1, or fragments thereof, can block binding of FVIII to clearance receptors and thereby extend its in vivo half-life, LRP1 is a 600 kDa integral membrane protein that is implicated in the receptor-mediate clearance of a variety of proteins, including FVIII. See, e.g., Lenting et al., Haemophilia 16 -16 (2010). Other suitable FVIII clearance receptors are, e.g., LDLR (low-density lipo protein receptor), VLDLR (very low-density lipoprotein receptor), and megalin (LRP-2), or fragments thereof. See, e.g., Bovenschen et al., Blood 106:906-912 (2005); Bovenschen, Blood 116:5439-5440 (2010); Martinelli et al., Blood 116:5688-5697 (2010).

III. Dosing Strategies for Clotting Factors

The present invention provides a dosing strategy for a clotting factor. A good dosing strategy provides reduced interpatient variability in pharmacokinetics and pharmacodynamics. While clotting factors have routinely been dosed based on the body weight of the patient, the present invention shows that a fixed dosing regimen is suitable for clotting factors that have a wide therapeutic window.

In one aspect, the invention provides methods of administering a clotting factor to a subject in need thereof, comprising administering to the subject a fixed dose of a clotting factor. Administration of the clotting factor is a replacement therapy by providing a recombinant clotting factor to a subject with clotting factor deficiency. Administration of the clotting factor can reduce the number of bleeding episodes or prevent the symptoms of a bleeding disorder in the subject.

In another aspect, the invention provides a method of reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject comprising administering a fixed dose of a clotting factor to the subject in need thereof. The invention also provides use of a fixed dose of a clotting factor for the manufacture of a medicament for reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject in need thereof. The one or more symptoms of a bleeding disease or disorder can be one or more bleeding episodes. The bleeding episodes can be spontaneous or caused by trauma or surgery. The invention can control bleeding, or prevent one or more bleeding episodes. The subject can be bleeding at the time of administration or be expected to be bleeding, or can be susceptible to bleeding as the result of minor hemorrhage, hemarthroses, superficial muscle hemorrhage, soft tissue hemorrhage, moderate hemorrhage, intramuscle or soft tissue hemorrhage with dissection, mucous membrane hemorrhage, hematuria, major hemorrhage, hemorrhage of the pharynx, hemorrhage of the retropharynx, hemorrhage of the retroperitonium, hemorrhage of the central nervous system, bruises, cuts, scrapes, joint hemorrhage, nose bleed, mouth bleed, gum bleed, intracranial bleeding, intraperitoneal bleeding, minor spontaneous hemorrhage, bleeding after major trauma, moderate skin bruising, or spontaneous hemorrhage into joints, muscles, internal organs or the brain. Such subjects also include those in need of peri-operative management, such as management of bleeding associated with surgery or dental extraction. in one aspect, the subject is in need of prophylaxis of one or more bleeding episodes. In another aspect, the subject is in need of individualized interval prophylaxis. In other aspects, the subject is in need of on-demand treatment or episodic treatment of one or more bleeding episodes. In still other aspects, the subject is in need of perioperative management of one or more bleeding episodes.

In other aspects, the invention includes a method of manufacturing a pharmaceutical composition, or compositions comprising formulating a fixed dose of a clotting factor. The fixed dose manufactured by the present method can be administered to a subject in need thereof. The pharmaceutical composition(s) can comprise, consist essentially or, or consist of a fixed dose of a clotting factor and one or more pharmaceutically acceptable carrier or excipient, but does not comprise any additional amount of the clotting factor. In some embodiments, the entire fixed dose is administered to the subject, i.e., no portion of the composition is left unused.

In some aspects, the invention provides a pharmaceutical composition comprising a fixed dose of a clotting factor and a pharmaceutically acceptable carrier for use to reduce, ameliorate, or, prevent one or more symptoms of a bleeding disease or disorder to a subject in need thereof. The pharmaceutical composition can comprise, consist essentially or, or consist of a fixed dose of a clotting factor and one or more pharmaceutically acceptable carrier or excipient, but does not comprise any additional amount of the clotting factor. In some embodiments, the entire fixed dose is administered to the subject, i.e., no portion of the composition is left unused. In certain embodiments, the pharmaceutical composition comprises a fixed dose of a cloning factor, wherein the fixed dose is provided in two or more (e.g., two, three, four, or five) vials. The total contents of which, provide the fixed dosage of the clotting factor.

A clotting factor can be formulated as a pharmaceutical composition. The pharmaceutical composition can be formulated for administration to humans. The pharmaceutical compositions used in the methods of this invention comprise pharmaceutically acceptable carriers, including, e.g., ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat. Various methods of formulating the invention are well known in the art.

In still other aspects, the invention is directed to a kit for administration of a fixed dose amount of a clotting factor comprising one or more containers (e.g., vials) of a pharmaceutical composition and an instructional material. In one embodiment, a kit comprises a single vial of a pharmaceutical composition comprising a fixed dose of a clotting factor and an instructional material, wherein the composition in the single vial is to be administered in its entirety to a subject in need thereof The instruction material that can be inserted in the kit can comprise instructions to administer the pharmaceutical composition of the clotting factor to the subject. In another embodiment, a kit comprises an x number of the pharmaceutical compositions, wherein x is any integer, and an instructional material, wherein each of the pharmaceutical compositions, e.g., each vial, comprises a portion of a clotting factor, wherein the total amount of the clotting factor, when combined, is a fixed dose for a subject in need thereof. For example, x can be 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. When the kit comprises two or more (i.e., x=2 or more) pharmaceutical compositions (e.g., vials), each comprising a portion of a clotting factor, the two or more compositions can be combined together into one vial or syringe. Techniques for combining vials, e.g., by using a large syringe, are known in the art. The combination of the portions of the two or more compositions provides the fixed dose of the cloning factor that is to be administered to the subject in need thereof In some embodiments, the entire fixed dose is administered to the subject, i.e., no portion of the composition is left unused.

In one example, a kit for administration of a fixed dose amount of a clotting factor (e.g., a fixed dose of 4000 IU) comprises a first container of a pharmaceutical composition and a second container of a pharmaceutical composition, wherein the first container comprises a first portion of a fixed dose of a clotting factor (e.g., a vial containing 2,000 IU of the clotting factor) and the second container comprises a second portion of the fixed dose of the clotting factor (e.g., a second vial containing 2,000 IU of the clotting factor) and wherein the total amount of the first container and the second container, when combined, is the fixed dose (e.g., 4,000 IU). In another example, a kit for administration of a fixed dose amount of a clotting factor (e.g., a fixed dose of 6000 IU) comprises a first container of a pharmaceutical composition, a second container of a pharmaceutical composition, and a third container of a pharmaceutical composition, wherein the first container comprises a first portion of a fixed dose of a modified clotting factor (e.g., a vial containing 2,000 IU of the clotting factor), the second container comprises a second portion of the fixed dose of the clotting factor (e.g., a second vial containing 2,000 IU of the clotting factor), and the third container comprises a third portion of the fixed dose of the clotting factor (e.g., a third vial containing 2,000 IU of the clotting factor) and wherein the total amount of the first container, the second container, and the third container is the fixed dose (e.g., 6,000 IU). In other examples, the first portion of the first pharmaceutical composition, the second portion of the pharmaceutical composition, and the third portion of the pharmaceutical composition are the same or different. The combination of the first and second composition (i.e., vials) and the third, if present, is the fixed dose. The entire fixed dose is then administered to the subject in need thereof.

In some embodiments, the two or more pharmaceutical compositions (e.g., vials) in a kit can be administered separately. For example, <a first pharmaceutical composition comprising a first portion, of a fixed dose of a clotting factor is first administered to a subject in need thereof, and a second pharmaceutical composition comprising a second portion of a fixed dose of a clotting factor is then administered to the subject.

The present invention also identifies the fixed dose that can treat or prevent one or more bleeding episodes in a subject, regardless of the body weight. Administration of the appropriate dosing amount for the dosing interval can achieve a plasma trough, level of a clotting, activity at least about 1 IU/dl or above 1 IU/dl during the interval in a subject administered with a clotting factor. In one embodiment, the invention includes a dosing amount (or ranges of the dosing amount) and a dosing interval (or ranges of the dosing interval) that can maintain a plasma trough level of a clotting activity at least about 1 IU/dl (1%) or above 1 IU/dl (1%), at least about 2 IU/dl (2%) or above 2 IU/dl (2%), at least about 3 IU/dl (3%) or above 3 IU/dl (3%), at least about 4 IU/dl (4%) or above 4 IU/dl (4%), at least about 5 IU/dl (5%) or above 5 IU/dl (5%), at least about 6 IU/dl (6%) or above 6 IU/dl (6%), at least about 7 IU/dl (7%) or above 7 IU/dl (7%), at least about 8 IU/dl (8%) or above 8 IU/dl (8%), at least about 9 IU/dl (9%) or above 9 IU/dl (9%), at least about 10 IU/dl (10%) or above 10 IU/dl (10%), at least about 11 IU/dl (11%) or above 11 IU/dl (11%), at least about 12 IU/dl (12%) or above 12 IU/dl (12%), at least about 13 IU/dl (13%) or above 13 IU/dl (13%), at least about 14 IU/dl (14%) or above 14 IU/dl (14%), at least about 15 IU/dl (15%) or above 15 IU/dl (15%), at least about 16 IU/dl (16%) or above 6 IU/dl (16%), at least about 17 IU/dl (17%) or above 17 IU/dl (17%), at least about 18 IU/dl (8) or above 18 IU/dl (18 IU/dl (18%), at least about 19 IU/dl (19%) or above 19 IU/dl (19%) least about 20 IU/dl (20%) or above 20 IU/dl (20%) throughout the interval.

In another embodiments, a plasma trough level of a clotting factor is maintained between about 1% and about 5%, between about 1% and about 6%, between about 1% and about 7%, between about 1% and about 8%, between about 1% and about 9%. between about 1% and about 10%, between about 1% and about 12%, between about 1% and about 14%, between about 1% and about 15%, between about 1% and about 17%, between about 1% and about 19%, between about 1% and about 20%, between about 1% and about 22%, between about and about 24%, between about 1% and about 25%, between about 1% and about 30%, between about 1% and about 35%.

In other embodiments, the trough is 1-5 or 1-3 IU/dl after about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13 or about 14 days after administration of a clotting factor. In some embodiments, the plasma level of the clotting factor reaches an average trough of at least about 1 IU/di after at least about 6 days or reaches a trough of at least about 1, 2, 3, 4, or 5 IU/dl after at least about 6 days in a subject. In some embodiments, the plasma level of the clotting factor reaches an average trough of about 1-5 or 1-3 IU/dl. Such trough or average trough may be reached after about 6, about 7, about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, about 18, about 19, about 20, about 21, about 22, about 23, about 24, about 25, about 26, about 27, about 28, about 29, about 30, about 31, about 32, about 33, about 34, about 35, about 36, about 37, about 38, about 39, or about 40 days.

In some embodiments, a dosing amount (or ranges of the dosing amount) and a dosing interval (or ranges of the dosing interval) are selected to reduce or decrease the frequency of bleeding or bleeding disorder. In other embodiments, the dosing amount (or ranges of the dosing amount) and the dosing interval (or ranges of the dosing interval) of a clotting factor stops on-going, uncontrollable bleeding or bleeding episodes in a subject administered with the dosing amount during the dosing interval. In still other embodiments, the dosing amount (or ranges of the dosing amount) and the dosing interval (or ranges of the dosing interval) of a clotting factor prevents spontaneous bleeding or bleeding episodes in a subject susceptible to such spontaneous bleeding or bleeding episodes.

In one aspect, a fixed dose of a FIX polypeptide is about 4,000 IU per dose, 6000 IU or about 8,000 IU per dose. In one embodiment, a dosing interval is at least about every five days, about every six days, at least about every seven days, at least about every eight days, at least about every nine days, at least about every ten days, at least about every 11 days, at least about every 12 days, at least about every 13 days, at least about every 14 days, at least about every 15 days, at least about every 16 days, at least about every 17 days, at least about every 18 days, at least about every 19 days, at least about every 20 days, or at least about every 21 days. In another embodiment, a fixed dose of a FIX polypeptide is about 4,000 IU per dose and is administered weekly, i.e., once per week. In other embodiments, a fixed dose of a FIX polypeptide is about 8,000 IU per dose and is administered every 10 days or once every two weeks. In yet other embodiments, the fixed dose of a long-acting FIX polypeptide is not calculated by the formula:


Number of factor IX IU required (IU)=Body Weight (kg)×Desired Factor IX Increase (% or IU/dL)×1 IU/kg per IU/dL)   (A)

In certain aspects of the invention, the method, the use, the pharmaceutical composition, or the kit further comprises administering an additional dosing amount of a clotting factor.

In certain embodiments, the fixed dosing strategy is a stratified dosing regimen. For example, the fixed dose can be stratified into two or more dose sizes based on specified weight categories. The weight categories can be low body weight, normal body weight, and high body weight. In one embodiment, the fixed dose is stratified into three dose, sizes suitable for, subjects with low, normal, or high body weight. The normal, low, or high body weight can be determined based on age, height, gender, frame size, general health, or any combination thereof or independently of age, height, gender, frame size, general health, or any combination thereof In another embodiment, a subject has a low body weight, and the fixed dose of a long- acting FIX polypeptide is about 5,000 IU per dose or about 6,000 IU per dose, which is administered at an interval longer than 7 days, e.g., every 10 days. In other embodiments, a subject has a normal body weight and the fixed dose is about 7500 IU per dose or about 8000 IU per dose, which is administered at an interval longer than 7 days, e.g., every 10 days. hi some embodiments, a subject has a high body weight and the fixed dose is about 10000 IU, per dose administered every 10 days or about 12000 IU per dose administered every 10 days.

The dosing interval can, alternatively, be an individualized interval that is determined for each subject based on pharmacokinetic data or other information about that subject. The individualized dose/dosing interval combination may be the same as those for fixed interval regimens in the preceding paragraphs, or may differ. The regimen can initially be at a fixed dosing interval, and then it can change to an individualized dosing interval.

In certain embodiments of the invention, the method of the invention further comprises measuring a baseline FIX activity of a subject prior to the initial administration of a FIX polypeptide. Measuring of a baseline FIX activity can employ any known clotting assays in the art, e.g., one step aPTT assay, two step chromogenic assay, ROTEM, TGA, or etc.

In one aspect, a fixed dose of a FVIII polypeptide is, about 2000 IU, about 2,500 IU, about 3,000 IU, about 3,500 IU, or about 4,000 IU per dose. In one embodiment, the fixed dose is administered twice a week (i.e., two times per week). In another embodiment, the fixed dose is administered weekly (i.e., once a week). In other embodiments, the entire fixed dose its administered to the subject, i.e., no portion of the composition is left unused. In yet other embodiments, the fixed dose of a long-acting FVIII polypeptide is not calculated by the formula:


Number of factor FVIII IU required (IU)=Body Weight (kg)×Desired Factor F VIII Increase (IU/dL or % of normal)×0.5(IU/kg per IU/dL)   (B)

IV. Methods of Making

A clotting factor can be manufactured in a host cell comprising a vector encoding the clotting factor. As used herein, an expression vector refers to any nucleic acid construct which contains the necessary elements for the transcription, and translation of an, inserted coding sequence, or in the case of an RNA viral vector, the necessary elements for replication and translation, when introduced into an appropriate host cell. Expression vectors can include plasmids, phagemids, viruses, and derivatives thereof.

A gene expression control sequence as used herein is any regulatory nucleotide sequence, such as a promoter sequence or promoter-enhancer combination, which facilitates the efficient transcription and translation of the coding nucleic acid to which it is operably linked. The gene expression control sequence may, for example, be a mammalian or viral promoter, such as a constitutive or inducible promoter. Constitutive mammalian promoters include, but are not limited to, the promoters for the following genes: hypoxanthine phosphoribosyl transferase (HPRT), adenosine deaminase, pyruvate kinase, beta-actin promoter, and other constitutive promoters. Exemplary viral promoters which function constitutively in eukaryotic cells include, for example, promoters from the cytomegalovirus (CMV), simian virus (e.g., SV40), papilloma virus, adenovirus, human immunodeficiency virus (HIV), Rous sarcoma virus, cytomegalovirus, the long terminal repeats (LTR) of Moloney leukemia virus, and other retroviruses, and the thymidine kinase promoter of herpes simplex virus. Other constitutive promoters are known to those of ordinary skill in the art. The promoters useful as gene expression sequences of the invention also include inducible promoters. Inducible promoters are expressed in the presence of an inducing agent. For example, the metallothionein promoter is induced to promote transcription and translation in the presence of certain metal ions. Other inducible promoters are known to those of ordinary skill in the art.

Examples of vectors include, but are not limited to viral vectors or plasmid vectors. Plasmid vectors have been extensively described in the art and are well-known to those, of skill in the art. See, e.g., Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, 1989. In the last few years, plasmid vectors have been found to be particularly advantageous for delivering genes to cells in vivo because of their inability to replicate within and integrate into a host genome. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operably encoded within the plasmid. Some commonly used plasmids available from commercial suppliers include pBR322, PUC18, pUC19, various pcDNA plasmids, pRC/CMV, various pCMV plasmids, pSV40, and pBlueScript. Additional examples of specific plasmids include pcDNA3.1, catalog number V79020; pcDNA3.1/hygro, catalog number V87020; pcDNA4/myc-His, catalog number V86320; and pBudCE4.1, catalog number V53220, all from Invitrogen (Carlsbad, Calif.). Other plasmids are well-known to those of ordinary skill in the art. Additionally, plasmids may be custom designed using standard molecular biology techniques to remove and/or add specific fragments of DNA.

The expression vector or vectors are then transfected or co-transfected into a suitable target cell, which will express the polypeptides. Transfection techniques known in the art include, but are not limited to, calcium phosphate precipitation (Wigler et al. (1978) Cell 14:725), electroporation (Neumann et al. (1982) EMBO J 1:841), and liposome-based reagents. A variety of host-expression vector systems may be utilized to express the proteins described herein including both prokaryotic and eukaryotic cells.

These include, but limited to, microorganisms such as bacteria (e.g., E. coli) transformed with recombinant bacteriophage DNA or plasmid DNA expression vectors containing an appropriate coding sequence; yeast or filamentous fungi transformed with recombinant yeast or fungi expression vectors containing an appropriate coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing an appropriate coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus or tobacco mosaic virus) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing an appropriate coding sequence; or animal cell systems, including mammalian cells (e.g., HEK 293, CHO, Cos, HeLa, HKB11, and BHK cells).

In one embodiment, the host cell is a eukaryotic cell. As used herein, a eukaryotic cell refers to any animal or plant cell having a definitive nucleus. Eukaryotic cells of animals include cells of vertebrates, e.g., mammals, and cells of invertebrates, e.g., insects. Eukaryotic cells of plants specifically can include, without limitation, yeast cells. A eukaryotic cell is distinct from a prokaryotic cell, e.g., bacteria.

In certain embodiments, the eukaryotic cell is a mammalian cell. A mammalian cell is any cell derived from a mammal. Mammalian cells specifically include, but are not limited to, mammalian cell lines. In one embodiment, the mammalian cell is a human cell. In another embodiment, the mammalian cell is a HEK 293 cell, which is a human embryonic kidney cell line. HEK 293 cells are available as CRL-1533 from American Type Culture Collection, Manassas, VA, and as 293-H cells, Catalog No. 11631-017 or 293-F cells, Catalog No. 11625-019 from Invitrogen (Carlsbad, Calif.). In some embodiments, the mammalian cell is a PER.C6® cell, which is a human cell line derived from retina. PER.C6® cells are available from Crucell (Leiden, The Netherlands). In other embodiments, the mammalian cell is a Chinese hamster ovary (CHO) cell. CHO cells are available from American Type Culture Collection, Manassas, Va. (e.g., CHO-K1; CCL-61). In still other embodiments, the mammalian cell is a baby hamster kidney (BHK) cell. BHK cells are available from American Type Culture Collection, Manassas, Va. (e.g., CRL-1632). In some embodiments, the mammalian cell is a HKB11 cell, which is a hybrid cell line of a HEK293 cell and a human B cell line. Mei et al., Mol. Biotechnol. 34(2): 165-78 (2006).

The method can further comprise purification steps. Various known purifications steps are well known in the art.

Having now described the present invention in detail, the same will be more 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 Population Pharmacokinetic Analysis of a Long-Acting Recombinant Factor IX-Fc Fusion Protein (rFIXFc) in Patients with Severe Hemophilia B

BACKGROUND: Population pharmacokinetic (popPK) models are developed to understand the sources of variability in dose requirements (covariates) and to help individualize dosing regimens if necessary. Dosing histories and patient-specific data are used to gain an understanding of drug disposition in order to discern specific demographic and/or clinical factors that may be predictors of PK parameters. By characterizing the population PK of long-acting FIX-Fc (rFIXFc) in patients with severe hemophilia B (≤2 IU/dL plasma FIX activity), a model of estimated population PK parameters of rFIXFc can be established. This model may assist physicians who wish to tailor dosing for individual patients with sparse PK samples. This model may also help determine the suitability of rFIXFc for a fixed dosing regimen.

METHODS: Male subjects with severe hemophilia B were included from a phase 1/2a clinical study (n=12) and a phase 3 clinical study (B-LONG, n≤123) of rFIXFc. Male subjects with severe hemophilia B were treated with long-lasting recombinant FIX-Fc (rFIXFc) in an amount of 50 IU/kg or 100 IU/kg. The subjects ranged in age from 12 to 76 years and in body weight from 45 to 186 kg. The modeling dataset included 135 baseline PK profiles at Week 1, as well as 21 repeat PK profiles at Week 26, with a total of 1400 measured FIX activity records. The final population PK model was validated using 1027 trough/peak FIX activity records from 119 patients.

In the popPK analysis, plasma FIX activity was measured by the one-stage (activated partial thromboplastin time) clotting assay. Corrected FIX activity was calculated using the formula:


Corrected FIX activity=Measured FIX activity−Baseline−Residual decay.

Baseline FIX activity was defined as the lowest level of activity (LLACT) recorded at screening, predose, postdose, or from historical clinical records. The baseline is defined as 0 when the LLACT is less than 1% (lower limit of quantification). The baseline FIX activity is equal to LLACT when LLACT is from 1% to 2% (i.e., 1≤LLACT≤2).

Prestudy residual decay was performed using terminal half-life obtained from a noncompartmental analysis of the individual data by the following formula:


Residual decay=(predose−baseline)×e−decay rate×time

Clearance was presented by the following formula:


CL=Typical CL×(BW/Typical BW)exponent,

where typical BW is 73 kg.

Volume of distribution was presented by the following formula:


V=Typical V×(BW/Typical BW)exponent,

where typical BW is 73 kg.

For the popPK model development, NONMEM VII version 1.0 (ICON Development Solutions, Ellicott City, Md.) was used. The modeling and qualification steps are presented below in Table 1.

TABLE 1 Modeling and Qualification Steps Steps Model selection Base model and Inter-individual variability (IIV) Base Model, IIV on CL/V1/Q2/V2/Q3 evaluation Inter-occasion variability (IOV) evaluation Base Model with IOV on CL and V1 Covariate Modelling Final model, body weight as covariate on Internal qualification (bootstrap and VPC) CL and V1 External qualification using trough/peak records CL, clearance; V, volume of distribution; Q, inter-compartmental clearance; VPC, visual predictive check

A first order conditional estimation with interaction method (FOCEI) was used to estimate the popPK parameters. Residual errors were modeled as combined proportional and additive errors. Stepwise forward addition (p<0.005) and backward elimination (p<0.001) covariate modeling was performed. Potential covariates assessed in this analysis included: body weight (BW), Age, Race, Blood type, Human Immunodeficiency Virus status, Hepatitis C Virus status, haematocrit, IgGi and albumin concentration, and FIX genotype.

Model qualifications included bootstrap, visual predictive check (VPC) and validation with trough/peak records. The mean relative prediction error (an indicator of accuracy) was calculated as:

1 N i = 1 i = N [ DV - 1 PRED ] DV

RESULTS: The rFIXFc disposition was best described by a three-compartment base model (FIG. 1). The model was further improved by including intra-subject random changes at different occasions (i.e., inter-occasion variability, IOV) for CL and V1 (FIG. 2). IOV was smaller than inter-individual variability (IIV), indicating that individual PK was more accurate than the mean popPK for individual PK prediction.

BW was the only statistically significant covariate on CL and V1 (volume of the central compartment). However, the impact of body weight on the PK of rFIXFc was limited. Body weight was found to be a significant covariate for rFIXFc disposition (FIG. 3), although the impact of BW was limited. For example, the BW exponent on CL and V1 was 0.436 and 0.396, respectively, and inclusion of BW reduced inter-individual variability (IIV) for both CL and V1 only by 3.4% and 2.5%, respectively. None of the other covariates assessed, including age, race, blood type or genotype, were significant covariates in this model.

The final popPK model is summarized below in Table 2.

TABLE 2 Summary of the final rFIXFc population pharmacokinetic model. 95% non- Population parametric CI IIVb IOV Parameter Estimate from bootstrapa (%) (%) CL = Typical CL × ( BW 72 ) 0.436 Typical CL for a 73 kg 2.39 2.29, 2.49 17.7 15.1 subject (dL/h) BW exponent on CL 0.436 0.272, 0.584 V 1 = Typical V 1 × ( BW 72 ) 0.396 Typical V1 for a 73 kg 71.4 58.5, 76.0 21.7 17.4 subject (dL) BW exponent on V1 0.396 0.169, 0.580 Q2 (dL/h) 1.67 1.35, 1.89 35.8 V2 (dL) 87.0 79.0, 95.5 46.2 Q3 (dL/h) 39.3 16.6, 141 V3 (dL) 39.9 36.6, 52.4 37.7 Residual Error: Proportional 10.6% Additive 0.24 IU/dL CI, confidence interval; IIV, inter-individual variability; IOV, inter occasion variability; CL, clearance; BW, body weight; V, volume of distribution; Q, inter-compartmental clearance

For a typical 73 kg subject, the predicted popPK values for clearance, volume of central compartment, and volume of distribution at steady state are 2.39 dL/h, 71.4 dL, and 198 dL, respectively. Goodness-of-fit plots show that the predicted popPK data generated by the model closely mimic the, observed FIX activity data (FIG. 4).

The results of the popPK model were validated using the observed FIX activity data. The median and 80% interval for observed and predicted FIX activity time profiles nearly overlapped, indicating that the final model was able to reproduce both the central tendency and variability of the observed FIX activity data on the time scale (FIG. 5). The strong correlation between observed and predicted FIX activities in the trough/peak dataset suggested that the final popPK model is predictive (FIG. 6).

Finally, the overall relative prediction error was −3.23% with a 95% confidence interval of −5.27% to −1.23%. Post hoc estimates from this popPK analysis were very similar to the results from the conventional PK analysis shown below in Table 3.

TABLE 3 Post hoc empirical Bayesian estimates of key PK parameters. Parameter Phase 3 Mean (SD) Phase 1/2a Mean (SD) Clearance (CL), mL/h/kg 3.42 (0.89) 2.82 (0.58) Volume of central compartment (V1), mL 102 (29.6) 96.2 (24.7) Incremental in vivo recovery, IU/dL per 1.02 (0.45) 1.04 (0.19) IU/kg Volume of distribution at steady-state 297 (90.5) 234 (70.8) (Vss), mL/kg Terminal Half-life, h 86.7 (27.9) 70.9 (13.9) Mean residence time (MRT), h 89.4 (25.9) 82.5 (15.5) SD, standard deviation

CONCLUSIONS: The three-compartment popPK model predicted disposition of rFIXFc with modest inter-individual variability (IIV). Individual PK parameters derived from the three-compartment popPK model were similar to those derived from the two-compartment conventional PK analysis, indicating a limited 3rd compartment contribution. For a typical 73 kg subject, the popPK model predicted a clearance of 2.39 dL/h; volume of central compartment of 71.4 dL; and volume of distribution, at steady state of 198 dL. The only significant covariate assessed in the popPK model was BW, although its impact on rFIXFc PK variability was limited.

Drugs with body weight effect on clearance (θBW_CL) and body weight effect on the central volume of distribution (θBW_V1) equal to or less than 0.5 are considered good candidates for fixed dosing regimens due to the limited impact of patient body weight on PK variability. Here, rFIXFc had θBW_CL and θBW-V1 values of 0.436 and 0.396, respectively (Table 2). Furthermore, the inclusion of BW in the population PK model resulted in a modest reduction of approximately 3% in IIV for both CI and V1. These results indicate that body weight had a minimal impact on PK variability and suggest that rFIXFc is suitable tor a fixed dosing regimen.

The final popPK model can be used to simulate dosing regimens and intervals for routine prophylaxis, control and prevention of bleeding episodes, and peri-operative management. This model may assist physicians who wish to tailor dosing for individual patients with sparse PK samples.

Example 2 Fixed Dosing

The body weight of adult patients has a limited impact on PK variability among patients. Therefore, rFIXFc is suitable for fixed dosing regimens that do not use the formula:


Number of factor FIX IU required (IU)=Body Weight (kg)×Desired Factor FIX Increase (IU/dL or % of normal)×0.5(IU/kg per IU/dL).

In this example, fixed dose regimens are established using vials of rFIXFc that contain 2,000 IU per vial. In one alternative, the entire population of adult patients is treated with 2 vials of rFIXFc once weekly. Alternatively, stratified fixed dosing is applied based on the BW range in which the patient belongs to.

METHODS: Patients with hemophilia B are categorized into one of three categories: (i) low body weight; (ii) normal body weight; and (iii) high body weight. Patients weighing, less than 57 kg are categorized as low body weight. Patients weighing between 57 and 104 kg are categorized as normal body weight. Patients weighing more than 104 kg are categorized as high body weight.

Patients in the low body weight category are treated with a single vial of fixed dose long acting FIXFc (i.e., 2,000 IU total) once weekly. Patients in the normal body weight category are treated with two vials of fixed dose long-acting rFIXFc (i.e., 4,000 IU total) once weekly. Patients in the high body weight category are treated with three vials of fixed dose long-acting rFIXFc (i.e., 6,000 IU total) once weekly.

RESULTS: The PK properties of long-acting rFIXFc are minimally affected by the BW. As FIG. 7A and 7B show, the 97.5th, median, and 2.5th percentiles of the simulated FIX activity-time profiles at steady state in 1000 subjects following fixed dosing (4000 IU once weekly and 8000 IU every 10 days; dotted lines) significantly overlap with those of BW-based dosing (50 IU/kg once weekly and 100 IU/kg every 10 days; solid lines). FIG. 8 shows that the percentages of population within the target therapeutic range following the fixed dosing and body weight (BW)-based dosing approaches in the BW-stratified population are similar. These data demonstrate that the clotting factors having a wide therapeutic window can be used for fixed dosing regimen for example, this allows the physician to treat patients with fixed dose regimens of long-acting rFIXFc, eliminating the need to use formulaic dose calculation methods.

Example 3 Population Pharmacokinetic Analysis of a Long-Acting Recombinant Factor VIII-Fc Fusion Protein (rFVIIIFc) in Patients with Severe Hemophilia A

By characterizing the population PK of long-acting FVIII-Fc (rFVIIIFc) in patients with severe hemophilia A, a model of estimated population PK parameters of rFVIIIFc can be established. This model may assist physicians who wish to tailor dosing for individual patients with sparse PK samples. This model may also help determine the suitability of rFVIIIFc for a fixed dosing regimen.

Objectives: To characterize the activity-time profiles of rFVIIIFc in hemophilia A patients by population PK analysis and to identify intrinsic covariates that may affect the variability of rFVIIIFc PK.

The modeling dataset included activity-time profiles of 180 subjects (15 from a Phase 1/2a study and 165 from a Phase 3 study [A-LONG], collected cover over ≤52 weeks of treatment). Subjects were 12 to 65 years old and weighed 41-132 kg. The analysis was done with NONMEM 7 software, and included model building, covariate search, and model qualification steps.

A 2-compartmental model adequately described the activity of rFVIIIFc. The population estimate for clearance (CL)=1.65 dL/h; volume of distribution at steady state (Vss)=44.4 dL. The inter-individual variability (IIV) of CL was moderate (24.3%) and central volume of distribution (V1) was low (13.4%). The inter-occasional variability (IOV) of both CL and V1 was low (20.6 and 12.0% respectively). The additive residual error was very low (0.208 III/dL), as was the proportional residual error (13.6%), approximating the variability of the one-stage clotting assay for FVIII activity. Von Willebrand Factor (VWF) level was identified as the major covariate for CL; higher levels of VWF yielded lower clearance values, reflecting the protective role that VWF has on FVIII activity. Body Weight (BW) and Haematocrit (HCT) were identified as weak covariates on V1.

This is the first population PK analysis that systematically describes and characterizes the prolonged activity profile of long-acting rFVIIIFc. The population PK model of rFVIII activity adequately describes the observed activity-time profiles. The clearance of rFVIIIFc activity is lower than the clearance observed for ADVATE®, resulting in longer duration of activity. The low IIV underlines the consistency and homogeneity of the activity profiles. The low IOV indicates that rFVIIIFc maintains stable and predictable activity with long term administration over time. The set of covariates identified is physiologically relevant. Therefore, the population model developed can be used to simulate various, dosing scenarios in support of dosing regimen selection and other decision making related to rFVIIIFc therapy. This approach represents an advance over the current utilitarian approach, wherein a regimen is not determined to be ineffective until after a patient has a bleeding episode.

The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the skill of the art, readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the skilled artisan in light of the teachings and guidance.

Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

All patents and publications cited herein are incorporated by reference herein in their entirety.

Tables

TABLE 4 Polynucleotide Sequences: FIX-Fc A. FIX-Fc Chain DNA Sequence SEQ ID NO: 1, which encodes SEQ ID NO: 2) pSYN-FIX-030 Nucleotide sequence (nt 1 to 7583): FIX exon 1 (signal peptide, 1st amino acid propeptide): nt 690-777 FIX mini intron: nt 778-1076 FIX propeptide sequence: nt 1077-1126 Mature FIX sequence: nt 1127-2371 Fc: nt 2372-3052 gcgcgcgttgacattgattattgactagttattaatagtaatcaattacggggtcattagttcatagcccatata tggagttccgcgttacataacttacggtaaatggcccgcctggctgaccgcccaacgacccccgcccattgacgt caataatgacgtatgttcccatagtaacgccaatagggactttccattgacgtcaatgggtggagtatttacggt aaactgcccacttggcagtacatcaagtgtatcatatgccaagtacgccccctattgacgtcaatgacggtaaat ggcccgcctggcattatgcccagtacatgaccttatgggactttcctacttggcagtacatctacgtattagtca tcgctattaccatggtgatgcggttttggcagtacatcaatgggcgtggatagcggtttgactcacggggatttc caagtctccaccccattgacgtcaatgggagtttgttttggcaccaaaatcaacgggactttccaaaatgtcgta acaactccgccccattgacgcaaatgggcggtaggcgtgtacggtgggaggtctatataagcagagctctctggc taactagagaacccactgcttactggcttatcgaaattaatacgactcactatagggagacccaagcttcgcgac gtacggccgccaccatgcagcgcgtgaacatgatcatggcagaatcaccaggcctcatcaccatctgccttttag gatatctactcagtgctgaatgtacaggtttgtttccttttttaaaatacattgagtatgcttgccttttagata tagaaatatctgatgctgtcttcttcactaaattttgattacatgatttgacagcaatattgaagagtctaacag ccagcacgcaggttggtaagtactgtgggaacatcacagattttggctccatgccctaaagagaaattggctttc agattatttggattaaaaacaaagactttcttaagagatgtaaaattttcatgatgttttcttttttgctaaaac taaagaattattcttttacatttcagtttttcttgatcatgaaaacgccaacaaaattctgaatcggccaaagag gtataattcaggtaaattggaagagtttgttcaagggaatctagagagagaatgtatggaagaaaagtgtagttt tgaagaagcacgagaagtttttgaaaacactgaaagaacaactgaattttggaagcagtatgttgatggagatca gtgtgagtccaatccatgtttaaatggcggcagttgcaaggatgacattaattcctatgaatgttggtgtccctt tggatttgaaggaaagaactgtgaattagatgtaacatgtaacattaagaatggcagatgcgagcagttttgtaa aaatagtgctgataacaaggtggtttgctcctgtactgagggatatcgacttgcagaaaaccagaagtcctgtga accagcagtgccatttccatgtggaagagtttctgtttcacaaacttctaagctcacccgtgctgagactgtttt tcctgatgtggactatgtaaattctactgaagctgaaaccattttggataacatcactcaaagcacccaatcatt taatgacttcactcgggttgttggtggagaagatgccaaaccaggtcaattcccttggcaggttgttttgaatgg taaagttgatgcattctgtggaggctctatcgttaatgaaaaatggattgtaactgctgcccactgtgttgaaac tggtgttaaaattacagttgtcgcaggtgaacataatattgaggagacagaacatacagagcaaaagcgaaatgt gattcgaattattcctcaccacaactacaatgcagctattaataagtacaaccatgacattgcccttctggaact ggacgaacccttagtgctaaacagctacgttacacctatttgcattgctgacaaggaatacacgaacatcttcct caaatttggatctggctatgtaagtggctggggaagagtcttccacaaagggagatcagctttagttcttcagta ccttagagttccacttgttgaccgagccacatgtcttcgatctacaaagttcaccatctataacaacatgttctg tgctggcttccatgaaggaggtagagattcatgtcaaggagatagtgggggaccccatgttactgaagtggaagg gaccagtttcttaactggaattattagctggggtgaagagtgtgcaatgaaaggcaaatatggaatatataccaa ggtgtcccggtatgtcaactggattaaggaaaaaacaaagctcactgacaaaactcacacatgcccaccgtgccc agctccggaactcctgggcggaccgtcagtcttcctcttccccccaaaacccaaggacaccctcatgatctcccg gacccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtcaagttcaactggtacgtgga cggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtacaacagcacgtaccgtgtggtcagcgt cctcaccgtcctgcaccaggactggctgaatggcaaggagtacaagtgcaaggtctccaacaaagccctcccagc ccccatcgagaaaaccatctccaaagccaaagggcagccccgagaaccacaggtgtacaccctgcccccatcccg ggatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagcgacatcgccgtgga gtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccgtgttggactccgacggctccttctt cctctacagcaagctcaccgtggacaagagcaggtggcagcaggggaacgtcttctcatgctccgtgatgcatga ggctctgcacaaccactacacgcagaagagcctctccctgtctccgggtaaatgagaattcagacatgataagat acattgatgagtttggacaaaccacaactagaatgcagtgaaaaaaatgctttatttgtgaaatttgtgatgcta ttgctttatttgtaaccattataagctgcaataaacaagttggggtgggcgaagaactccagcatgagatccccg cgctggaggatcatccagccggcgtcccggaaaacgattccgaagcccaacctttcatagaaggcggcggtggaa tcgaaatctcgtagcacgtgtcagtcctgctcctcggccacgaagtgcacgcagttgccggccgggtcgcgcagg gcgaactcccgcccccacggctgctcgccgatctcggtcatggccggcccggaggcgtcccggaagttcgtggac acgacctccgaccactcggcgtacagctcgtccaggccgcgcacccacacccaggccagggtgttgtccggcacc acctggtcctggaccgcgctgatgaacagggtcacgtcgtcccggaccacaccggcgaagtcgtcctccacgaag tcccgggagaacccgagccggtcggtccagaactcgaccgctccggcgacgtcgcgcgcggtgagcaccggaacg gcactggtcaacttggccatggtttagttcctcaccttgtcgtattatactatgccgatatactatgccgatgat taattgtcaacacgtgctgatcagatccgaaaatggatatacaagctcccgggagctttttgcaaaagcctaggc ctccaaaaaagcctcctcactacttctggaatagctcagaggcagaggcggcctcggcctctgcataaataaaaa aaattagtcagccatggggcggagaatgggcggaactgggcggagttaggggcgggatgggcggagttaggggcg ggactatggttgctgactaattgagatgcatgctttgcatacttctgcctgctggggagcctggggactttccac acctggttgctgactaattgagatgcatgctttgcatacttctgcctgctggggagcctggggactttccacacc ctcgtcgagctagcttcgtgaggctccggtgcccgtcagtgggcagagcgcacatcgcccacagtccccgagaag ttggggggaggggtcggcaattgaaccggtgcctagagaaggtggcgcggggtaaactgggaaagtgatgtcgtg tactggctccgcctttttcccgagggtgqgggagaaccgtatataagtgcagtagtcgccgtgaacgttcttttt cgcaacgggtttgccgccagaacacaggtaagtgccgtgtgtggttcccgcgggcctggcctctttacgggttat ggcccttgcgtgccttgaattacttccacctggctccagtacgtgattcttgatcccgagctggagccaggggcg ggccttgcgctttaggagccccttcgcctcgtgcttgagttgaggcctggcctgggcgctggggccgccgcgtgc gaatctggtggcaccttcgcgcctgtctcgctgctttcgataagtctctagccatttaaaatttttgatgacctg ctgcgacgctttttttctggcaagatagtcttgtaaatgcgggccaggatctgcacactggtatttcggtttttg gggccgcgggcggcgacggggcccgtgcgtcccagcgcacatgttcggcgaggcggggcctgcgagcgcggccac cgagaatcggacgggggtagtctcaagctggccggcctgctctggtgcctggcctcgcgccgccgtgtatcgccc cgccctgggcggcaaggctggcccggtcggcaccagttgcgtgagcggaaagatggccgcttcccggccctgctc cagggggctcaaaatggaggacgcggcgctcgggagagcgggcgggtgagtcacccacacaaaggaaaggggcct ttccgtcctcagccgtcgcttcatgtgactccacggagtaccgggcgccgtccaggcacctcgattagttctgga gcttttggagtacgtcgtctttaggttggggggaggggttttatgcgatggagtttccccacactgagtgggtgg agactgaagttaggccagcttggcacttgatgtaattctccttggaatttgccctttttgagtttggatcttggt tcattctcaagcctcagacagtggttcaaagtttttttcttccatttcaggtgtcgtgaacacgtggtcgcggcc gcgccgccaccatggagacagacacactcctgctatgggtactgctgctctgggttccaggttccactggtgaca aaactcacacatgcccaccgtgcccagcacctgaactcctgggaggaccgtcagtcttcctcttccccccaaaac ccaaggacaccctcatgatctcccggacccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctg aggtcaagttcaactggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtaca acagcacgtaccgtgtggtcagcgtcctcaccgtcctgcaccaggactggctgaatggcaaggagtacaagtgca aggtctccaacaaagccctcccagcccccatcgagaaaaccatctccaaagccaaagggcagccccgagaaccac aggtgtacaccctgcccccatcccgcgatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaaggct tctatcccagcgacatcgccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctcccg tgttggactccgacggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggcagcaggggaacg tcttctcatgctccgtgatgcatgaggctctgcacaaccactacacgcagaagagcctctccctgtctccgggta aatgactcgagagatctggccggctgggcccgtttcgaaggtaagcctatccctaaccctctcctcggtctcgat tctacgcgtaccggtcatcatcaccatcaccattgagtttaaacccgctgatcagcctcgactgtgccttctagt tgccagccatctgttgtttgcccctcccccgtgccttccttgaccctggaaggtgccactcccactgtcctttcc taataaaatgaggaaattgcatcgcattgtctgagtaggtgtcattctattctggggggtggggtggggcaggac agcaagggggaggattgggaagacaatagcaggcatgctggggatgcggtgggctctatggcttctgaggcggaa agaaccagtggcggtaatacggttatccacagaatcaggggataacgcaggaaagaacatgtgagcaaaaggcca gcaaaaggccaggaaccgtaaaaaggccgcgttgctggcgtttttccataggctccgcccccctgacgagcatca caaaaatcgacgctcaagtcagaggtggcgaaacccgacaggactataaagataccaggcgtttccccctagaag ctccctcgtgcgctctcctgttccgaccctgccgcttaccggatacctgtccgcctttctcccttcgggaagcgt ggcgctttctcatagctcacgctgtaggtatctcagttcggtgtaggtcgttcgctccaagctgggctgtgtgca cgaaccccccgttcagcccgaccgctgcgccttatccggtaactatcgtcttgagtccaacccggtaagacacga cttatcgccactggcagcagccactggtaacaggattagcagagcgaggtatgtaggcggtgctacagagttctt gaagtggtggcctaactacggctacactagaagaacagtatttggtatctgcgctctgctgaagccagttacctt cggaaaaagagttggtagctcttgatccggcaaacaaaccaccgctggtagcggtggtttttttgtttgcaagca gcagattacgcgcagaaaaaaaggatctcaagaagatcctttgatcttttctacggggtctgacgctcagtggaa cgaaaactcacgttaagggattttggtcatgacattaacctataaaaataggcgtatcacgaggccctttcgtct cgcgcgtttcggtgatgacggtgaaaacctctgacacatgcagctcccggagacggtcacagcttgtctgtaagc ggatgccgggagcagacaagcccgtcagggcgcgtcagcgggtgttggcgggtgtcggggctggcttaactatgc ggcatcagagcagattgtactgagagtgcaccatatatgcggtgtgaaataccgcacagatgcgtaaggagaaaa taccgatcaggcgccattcgccattcaggctgcgcaactgttgggaagggcgatcggtgcgggcctcttcgcta ttacgcca B. FC DNA sequence (mouse Igκ signal peptide underlined) (SEQ ID NO: 3, which encodes SEQ ID NO: 4) This is the FC cassette from pSYN-FIX-030. In addition, there is a separate Fc expression cassette that was transfected into the cell line in plasmid pSYN-Fc-015 that encodes the same amino acid sequence, but contains a few noncoding changes. The second copy of Fc encoding sequence enables a better monomer: dimer ratio. atggagacagacacactcctgctatgggtactgctgctctgggttccaggttccactggtgacaaaactcacaca tgcccaccgtgcccagcacctgaactcctgggaggaccgtcagtcttcctcttccccccaaaacccaaggacacc ctcatgatctccoggacccctgaggtcacatgcgtggtggtggacgtgagccacgaagaccctgaggtcaagttc aactggtacgtggacggcgtggaggtgcataatgccaagacaaagccgcgggaggagcagtacaacagcacgtac cgtgtggtcagcgtcctcaccgtcctgcaccaggactggctgaatggcaaggagtacaagtgcaaggtctccaac aaagccctcccagcccccatcgagaaaaccatctccaaagccaaagggcagccccgagaaccacaggtgtacacc ctgcccccatcccgcgatgagctgaccaagaaccaggtcagcctgacctgcctggtcaaaggcttctatcccagc gacatcgccgtggagtgggagagcaatgggcagccggagaacaactacaagaccacgcctaccgtgttggactcc gacggctccttcttcctctacagcaagctcaccgtggacaagagcaggtggcagcaggggaacgtcttctcatgc tccgtgatgcatgaggctctgcacaaccactacacgcagaagagcctctccctgtctccgggtaaa

TABLE 5 Polypeptide Sequences: FIX-Fc FIX-Fc Monomer Hybrid: created by coexpressing FIX-Fc and Fc chains. A. FIX-Fc chain (SEQ ID NO: 2): (28 amino acid signal sequence underlined, 18 amino acid propeptide double underlined, Fc portion in italics.) The C-terminal lysine is not present in either subunit; this processing is often observed in recombinant proteins produced in mammalian cell culture, as well as with plasma derived proteins. FIXFC-SC SUBUNIT: FIX Signal Peptide: -46 MQRVNMIMAE SPGLITICLL GYLLSAEC FIX Propeptide: -18 TVFLDHENAN KILNRPKR   1 YNSGKLEEFV QGNLERECME EKCSFEEARE VFENTERTTE FWKQYVDGDQ  51 CESNPCLNGG SCKDDINSYE CWCPFGFEGK NCELDVTCNI KNGRCEQFCK 101 NSADNKVVCS CTEGYRLAEN QKSCEPAVPF PCGRVSVSQT SKMTRAETVF 151 PDVDYVNSTE AETILDNITQ STQSFNDFTR VVGGEDAKPG QFPWQVVLNG 201 KVDAFCGGSI VNEKWrVTAA HCVETGVKIT VVAGEHNIEE TEHTEQKRNV 251 IRIIPHHNYN AAINKYNHDI ALLELDEPLV LNSYVTPICI ADKEYTNIFL 301 KFGSGYVSGW GRVFHKGRSA LVLQYLRVPL VDRATCLRST KFTIYNNMFC 351 AGFHEGGRDS CQGDSGGPHV TEVEGTSFLT GIISWGEECA MKGKYGIYTK 401 VSRYVNWIKE KIKLTDKTHT CPPCPAPELL GGPSVFLEPP KPKDTLMISR 451 TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV 501 LTVLHQDWLN GKEYKCKVSN KALPAPLEKT ISKAKGQPRE PQVYTLPPSR 551 DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF 601 LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK B. Fc chain (SEQ ID NO: 4) 20 amino acid heterologous mouse Igκ light chain simal peptide (underlined): -20 METDTLLLWV LLLWVPGSTG Mature Fc sequence (corresponding to human IgG1 amino acids 221 to 447, EU numbering)   1 DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED  51 PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK 101 CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK 151 GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG 201 NVFSCSVMHE ALHNHYTQKS LSLSPGK

TABLE 6 Polynucleotide Sequence: FIVIII-Fc A. B-Domain Deleted FVIIIFc (i) B-Domain Deleted FVIIIFc Chain DNA Sequence (FVIII signal peptide underlined. Fc region in bold) (SEQ ID NO: 5, which encodes SEQ ID NO: 6)  661                                A TGCAAATAGA GCTCTCCACC TGCTTCTTTC  721 TGTGCCTTTT GCGATTCTCC 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 TTCAGGATGA ATCAGGAATC TTGGGACCTT 2101 TACTTTATGG GGAAGTTGGA GACACACTGT TGATTATATT TAAGAATCAA GCAAGCAGAC 2161 CATATAACAT CTACCCTCAC GGAATCACTG ATGTCCGTCC TTTGTATTCA AGGACATTAC 2221 CAAAAGGTGT AAAACATTTG AAGGATTTTC CAATTCTGCC AGGAGAAATA TTCAAATATA 2281 AATGGACAGT GACTCTAGAA GATGGGCCAA CTAAATCAGA TCCTCGGTGC CTGACCCGCT 2341 ATTACTCTAG TTTCGTTAAT ATGGAGAGAG ATCTAGCTTC AGGACTCATT GGCCCTCTCC 2401 TCATCTGCTA CAAAGAATCT GTAGATCAAA GAGGAAACCA GATAATGTCA CACAAGAGCA 2461 ATGTCATCCT GTTTTCTGTA TTTGATGAGA ACCGAAGCTG GTACCTCACA GAGAATATAC 2521 AACGCTTTCT CCCCAATCCA GCTGGAGTGC ACCTTGAGGA 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 TCCAAAACCC AGGTCTATGG ATTCTGGGGT 2821 GCCACAACTC AGACTTTCGG AACAGAGGCA TGACCGCCTT ACTGAAGGTT TCTAGTTGTG 2881 ACAAGAACAC TGGTCATTAT TACGAGGACA CTTATGAAGA TATTTCAGCA TACTTGCTCA 2941 GTAAAAACAA TGCCATTGAA CCAAGAACCT TCTCTCAAAA CCCACCAGTC TTGAAACGCC 3001 ATCAACGGGA AATAACTCGT ACTACTCTTC AGTCAGATCA AGACCAAATT GACTATGATG 3061 ATACCATATC AGTTGAAATG AAGAAGGAAG ATTTTGACAT TTATGATGAG GATGAAAATC 3121 AGAGCCCCCG CAGCTTTCAA AAGAAAACAC GACACTATTT TATTGCTGCA GTGGAGAGGC 3181 TCTGGGATTA TGGGATGACT AGCTCCCCAC ATGTTCTAAG AAACAGGGCT CAGAGTGGCA 3241 GTGTCCCTCA GTTCAAGAAA CTTGTTTTCC AGGAATTTAC TGATGGCTCC TTTACTCAGC 3301 CCTTATACCG TGGAGAACTA AATCAACATT TGGGACTCCT GGGGCCATAT ATAAGAGCAG 3361 AAGTTGAAGA TAATATCATG GTAACTTTCA GAAATCAGGC CTCTCGTCCC TATTCCTTCT 3421 ATTCTAGCCT TATTTCTTAT GAGGAAGATC AGAGGCAAGG AGCAGAACCT AGAAAAAACT 3481 TTGTCAAGCC TAATGAAACC AAAACTTACT TTTGGAAAGT CCAACATCAT ATGGCACCCA 3541 CTAAAGATGA CTTTGACTGC AAAGCCTGGG CTTATTTCTC TGATGTTGAC CTGGAAAAAG 3601 ATGTGCACTC AGGCCTGATT GGACCCCTTC TGGTCTGCCA CACTAACACA CTGAACCCTG 3661 CTCATGGGAG ACAACTGACA CTACAGGAAT TTGCTCTCTT TTTCACCATC TTTGATGAGA 3721 CCAAAAGCTG GTACTTCACT GAAAATATCG AAAGAAACTG CAGGGCTCCC TGCAATATCC 3781 AGATGGAAGA TCCCACTTTT AAAGAGAATT ATCGCTTCCA TGCAATCAAT GCCTACATAA 3841 TGGATACACT ACCTGGCTTA GTAATGGCTC AGGATCAAAG GATTCGATGG TATCTCCTCA 3901 GCATGGGCAG CAATGAAAAC ATCCATTCTA TTCATTTCAG TGGACATGTG TTCACTGTAC 3961 GAAAAAAAGA GGAGTATAAA ATCGCACTGT ACAATCTCTA TCCAGGTGTT TTTGACACAG 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 CTAACTACTC 4801 AGGGAGTAAA ATCTCTGCTT ACCAGCATGT ATGTGAAGGA GTTCCTCATC TCCAGCAGTC 4861 AAGATGGCCA TCAGTGGACT CTCTTTTTTC AGAATGGCAA AGTAAAGGTT TTTCAGGGAA 4921 ATCAAGACTC CTTCACACCT GTGGTGAACT CTCTAGACCC ACCGTTACTG ACTCGCTACC 4981 TTCGAATTCA CCCCCAGAGT TCCCTCCACC AGATTGCCCT GAGGATGGAG CTTCTCGCCT 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 Igκ singal peptide underlined) (SEQ ID NO: 3, which encodes SEQ ID NO: 4) 7981                                                  ATGGA GACAGACACA 8041 CTCCTGCTAT CGCTACTGCT GCTCTGGGTT CCAGCTTCCA CTGGTGACAA AACTCACACA 8101 TGCCCACCGT GCCCAGCACC TGAACTCCTG GGAGGACCGT CAGTCTTCCT CTTCCCCCCA 8161 AAACCCAAGC ACACCCTCAT GATCTCCCGC ACCCCTGAGG TCACATGCGT GGTGGTGGAC 8221 GTGAGCCACG AAGACCCTGA GGTCAAGTTC AACTGCTACG TGGACGGCGT GGAGGTGCAT 8281 AATGCCAAGA CAAACCCGCC GGAGGAGCAG TACAACAGCA CGTACCGTGT GGTCAGCGTC 8341 CTCACCGTCC TGCACCAGGA CTGGCTGAAT GGCAAGGAGT ACAAGTGCAA GGTCTCCAAC 8401 AAACCCCTCC CAGCCCCCAT CGAGAAAACC ATCTCCAAAG CCAAAGGGCA GCCCCGAGAA 8461 CCACAGGTGT ACACCCTGCC CCCATCCCGC GATGAGCTGA CCAAGAACCA GGTCAGCCTG 8521 ACCTGCCTCC TCAAAGGCTT CTATCCCAGC GACATCGCCG TGGAGTGGGA GAGCAATGCG 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 (i) Full-length FVIIIFc DNA Sequence (FVIII signal peptide underlined, Fc region in bold) (SEQ ID NO: 7, which encodes SEQ ID NO: 8)  661                                         ATG CAAATAGAGC TCTCCACCTG  721 CTTCTTTCTG TGCCTTTTGC GATTCTGCTT TAGTGCCACC AGAAGATACT ACCTGGGTGC  781 AGTGGAACTG TCATGGGACT ATATGCAAAG TGATCTCGCT GAGCTGCCTG TCGACGCAAG  841 ATTTCCTCCa AGAGTGCCAA AATCTTTTCC ATTCAACACC TCAGTCGTGT ACAAAAAGAC  901 TCTGTTTGTA GAATTCACGG ATCACCTTTT CAACATCGCT AAGCCAACCC CACCCTGGAT  961 GGGTCTGCTA GGTCCTACCA TCCAGGCTGA GGTTTATGAT ACAGTGGTCA TTACACTTAA 1021 GAACATGGCT TCCCATCCTG TCAGTCTTCA TGCTGTTGGT GTATCCTACT CGAAAGCTTC 1081 TGAGCGAGCT GAATATGATG ATCAGACCAG TCAAAGGGAG AAAGAAGATG ATAAAGTCTT 1141 CCCTGGTGGA AGCCATACAT ATGTCTGGCA GGTCCTGAAA GAGAATCCTC CAATGGCCTC 1201 TGACCCACTG TGCCTTACCT ACTCATATCT TTCTCATGTG GACCTGGTAA AAGACTTGAA 1261 TTCAGGCCTC ATTGGAGCCC TACTAGTATG TAGAGAAGGG AGTCTCGCCA ACCAAAAGAC 1321 ACACACCTTC CACAAATTTA TACTACTTTT TGCTGTATTT GATGAAGGGA AAAGTTGGCA 1381 CTCAGAAACA AAGAACTCCT TGATGCAGGA TAGGGATCCT GCATCTGCTC GGCCCTGGCC 1441 TAAAATCCAC ACAGTCAATG GTTATGTAAA CAGGTCTCTG CCAGGTCTGA TTGGATGCCA 1501 CAGGAAATCA GTCTATTGGC ATGTGATTGG AATGGGCACC ACTCCTCAAG TGCACTCAAT 1561 ATTCCTCGAA GGTCACACAT TTCTTGTGAG GAACCATCGC CAGGCGTCCT TGGAAATCTC- 1621 GCCAATAACT TTCCTTACTG CTCAAACACT CTTCATGGAC CTTGCACAGT 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 AACAATGCCC CTCAGCGGAT TGGTACGAAC 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 CAAGACCAAT GTCATCCTGT TTTCTGTATT TGATGAGAAC CGAAGCTGGT ACCTCACAGA 2521 GAATATACAA CGCTTTCTCC CCAATCCAGC TGGAGTGCAG CTTGAGGATC CAGAGTTCCA 2581 AGCCTCCAAC ATCATGCACA CCATCAATGG CTATGTTTTT GATAGTTTGC AGTTGTCAGT 2641 TTGTTTGCAT GACGTGGCAT ACTGGTACAT TCTAAGCATT CGAGCACAGA CTGACTTCCT 2701 TTCTGTCTTC TTCTCTGGAT ATACCTTCAA ACACAAAATG GTCTATGAAG ACACACTCAC 2761 CCTATTCCCA TTCTCAGGAG AAACTGTCTT CATGTCGATG GAAAACCCAG GTCTATGGAT 2821 TCTGGGGTGC CACAACTCAC ACTTTCCGAA 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 TATCATAGTC 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 ACAAAGCACT CATGGAAACA ACTCTCTGAA CTCTGGGCAA GGCCCCAGTC CAAAGCAATT 4141 AGTATCCTTA GGACCAGAAA AATCTGTCGA ACCTCAGAAT TTCTTCTCTG AGAAAAACAA 4201 AGTGGTAGTA GGAAAGGGTG AATTTACAAA GGACGTAGGA CTCAAAGAGA TGGTTTTTCC 4261 AAGCACCAGA AACCTATTTC TTACTAACTT GGATAATTTA CATGAAAATA ATACACACAA 4321 TCAAGAAAAA AAAATTCAGG AAGAAATAGA AAAGAAGGAA ACATTAATCC AAGAGAATGT 4381 AGTTTTGCCT CAGATACATA CAGTGACTGG CACTAAGAAT TTCATGAAGA AECTTTTCTT 4441 ACTGAGCACT AGGCAAAATG TAGAAGGTTC ATATGACGGG GCATATGCTC CAGTACTTCA 4501 AGATTTTAGC 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 GCAAATACAT 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 TCGCAAAGTT GAATTGCTTC CAAAAGTTCA CATTTATCAG AAGGACCTAT TCCCTACGGA 5281 AACTACCAAT GGGTCTCCTC GCCATCTGGA TCTCGTGGAA GGGAGCCTTC TTCAGCGAAC 5341 ACAGGGAGCG ATTAAGTGGA ATGAAGCAAA CAGACCTCCA AAAGTTCCCT TTCTGAGAGT 5401 AGCAACAGAA AGCTCTGCAA AGACTCCCTC CAAGCTATTG CATCCTCTTG CTTGGCATAA 5461 CCACTATGGT ACTCAGATAC CAAAAGAAGA GTGGAAATCC CAAGAGAAGT CACCAGAAAA 5521 AACAGCTTTT AAGAAAAAGG ATACCATTTT GTCCCTGAAC GCTTCTGAAA GCAATCATGC 5581 AATAGCAGCA ATAAATGAGG GAcAAAATAA GCCCGAAATA GAAGTCACCT GGGCAAAGCA 5641 AGCTAGGACT GAAACCCTGT GCTCTCAAAA CCCACCAGTC TTGAAACGCC ATCAACGGGA 5701 AATAACTCGT ACTACTCTTC AGTCAGATCA AGAGGAAATT CACTATGATG 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 TGCGACTCCT GGGGCCATAT ATAAGAGCAG AAGTTGAAGA 6061 TAATATCATG GTAACTTTCA GAAATCAGGC CTCTCCTCCC TATTCCTTCT ATTCTAGCCT 6121 TATTTCTTAT GAGGAAGATC ACACCCAAGC AGCAGAACCT AGAAAAAACT TTGTCAAGCC 6181 TAATGAAACC AAAACTTACT TTTGGAAAGT GCAACATCAT ATGGCACCCA CTAAAGATCA 6241 GTTTCACTGC AAAGCCTGGG CTTATTTCTC TGATGTTGAC CTGGAAAAAG ATCTGCACTC 6301 AGGCCTGATT GGACCCCTTC TCGTCTCCCA CACTAACACA CTGAACCCTC CTCATGGGAG 6361 ACAAGTGACA GTACAGGAAT TTGCTCTGTT TTTCACCATC TTTGATCAGA CCAAAAGCTG 6421 GTACTTCACT GAAAATATGG AAAGAAACTG CAGGGCTCCC TGCAATATCC AGATGGAAGA 6481 TCCCACTTTT AAAGAGAATT ATCGCTTCCA TCCAATCAAT GGCTACATAA TGGATACACT 6541 ACCTGGCTTA GTAATGGCTC AGGATCAAAG GATTCGATGG TATCTGCTCA CCATGGCCAG 6601 CAATGAAAAC ATCCATTCTA TTCATTTCAG TGGACATGTC TTCACTGTAC GAAAAAAAGA 6661 GGAGTATAAA ATGGCACTGT ACAATCTCTA TCCACCTCTT TTTGAGACAC TGGAAATGTT 6721 ACCATCCAAA GCTCGAATTT GGCGGGTGGA ATGCCTTATT GCCGACCATC TACATGCTGG 6781 GATGAGCACA CTTTTTCTGG TGTACAGCAA TAAGTGTCAG ACTCCCCTGG GAATGGCTTC 6841 TGCACACATT 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 ATAGCATTCC 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 GACTTCCACA 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 GGTGMACACC 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 (SEQ ID NO: 3)

TABLE 7 Polypeptide Sequence: FVIII-Fc A. D-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) (SEQ ID NO: 6) FVIII SIGNAL PEPTIDE: -19 MQIELSTCFFLCLLRFCFS FVIII MATURE POLYPEPTIDE SEQUENCE: ATRRYYLGAVELSWDYMQSDLGELPVDARFPPRVPKSFPFNTSVVYKKTLFVEFTDHLFNIAKPRPPW MGLLGPTIQAEVYDTVVITLKNMASHPVSLHAVGVSYWKASEGAEYDDQTSQREKEDDKVFPGGSHTY VWQVLKENGPMASDPLCLTYSYLSHVDLVKDLNSGLIGALLVCREGSLAKEKTQTLHKFILLFAVFDE GKSWHSETKNSLMQDRDAASARAWPKMHTVNGYVNRSLPGLIGCHRKSVYWHVIGMGTTPEVHSIFLE GHTFLVRNHRQASLEISPITFLTAQTLLMDLGQFLLFCHISSHQHDGMEAYVKVDSCPEEPQLRMKNN EEAEDYDDDLTDSEMDVVRFDDDNSPSFIQIRSVAKKHPKTWVHYIAAEEEDWDYAPLVLAPDDRSYK SQYLNNGPQRIGRKYKKVRFMAYTDETFKTREAIQHESGILGPLLYGEVGDTLLIIFKNQASRPYNIY PHGITDVRPLYSRRLPKGVKHLKDFPILPGEIFKYKWTVTVEDGPTKSDPRCLTRYYSSFVNMERDLA SGLIGPLLICYKESVDQRGNQIMSDKRNVILFSVFDENRSWYLTENIQRFLPNPAGVQLEDPEFQASN IMHSINGYVFDSLQLSVCLHEVAYWYILSIGAQTDFLSVFFSGYTFKHKMVYEDTLTLFPFSGETVFM SMENPGLWILGCHNSDFRNRGMTALLKVSSCDKNTGDYYEDSYEDISAYLLSKNNAIEPRSFSQNPPV LKRHQREITRTTLQSDQEEIDYDDTISVEMKKEDFDIYDEDENQSPRSFQKKTRHYFIAAVERLWDYG MSSSPHVLRNRAQSGSVPQFKKVVFQEFTDGSFTQPLYRGELNEHLGLLGPYIRAEVEDNIMVTFRNQ ASRPYSFYSSLISYEEDQRQGAEPRKNFVKPNETKTYFWKVQHHMAPTKDEFDCKAWAYFSDVDLEKD VHSGLIGPLLVCHTNTLNPAHGRQVTVQEFALFFTIFDETKSWYFTENMERNCRAPCNIQMEDPTFKE NYRFHAINGYIMDTLPGLVMAQDQRIRWYLLSMGSNENIHSIHFSGHVFTVRKKEEYKMALYNLYPGV FETVEMLPSKAGIWRVECLIGEHLHAGMSTLFLVYSNKGQTPLGMASGHIRDFQITASGQYGQWAPKL ARLHYSGSINAWSTKEPFSWIKVDLLAPMIIHGIKTQGARQKFSSLYISQFIIMYSLDGKKWQTYRGN STGTLMVFFGKVDSSGIKHNIFNPPIIARYIRLHPTHYSIRSTLRMELMGCDLNSCSMPLGMESKAIS DAQITASSYFTNMFATWSPSKARLHLQGRSNAWRPQVNNPKEWLQVDFQKTMKVTGVTTQGVKSLLTS MYVKEFLISSSQDGHQWTLFFQNGKVKVFQGNQDSFTPVVNSLDPPLLTRYLRIHPQSWVHQIALRME VLGCEAQDLYDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY VDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREP QVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDK SRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK ii) Fc chain (20 amino acid heterologous signal peptide from mouse Igκ chain underlined) (SEQ ID NO: 4) FC SIGNAL PEPTIDE: --20 METDTLLLWVLLLWVPGSTG FC SEQUENCE: DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK TKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD ELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFS CSVMHEALHNHYTQKSLSLSPGK 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 (SEQ ID NO: 8) FVIII SIGNAL PEPTIDE: -19 MQIELSTCFFLCLLRFCFS FVIII MATURE SEQUENCE: ATRRYYLGAVELSWDYMQSDLGELPVDARFPPRVPKSFPFNTSVVYKKTLFVEFTDHLFNIAKPRPPW MGLLGPTIQAEVYDTVVITLKNMASHPVSLHAVGVSYWKASEGAEYDDQTSQREKEDDKVFPGGSHTY VWQVLKENGPMASDPLCLTYSYLSHVDLVKDLNSGLIGALLVCREGSLAKEKTQTLHKFILLFAVFDE GKSWHSETKNSLMQDRDAASARAWPKMHTVNGYVNRSLPGLIGCHRKSVYWHVIGMGTTPEVHSIFLE GHTFLVRNHRQASLEISPITFLTAQTLLMDLGQFLLFCHISSHQHDGMEAYVKVDSCPEEPQLRMKNN EEAEDYDDDLTDSEMDVVRFDDDNSPSFIQIRSVAKKHPKTWVHYIAAEEEDWDYAPLVLAPDDRSYK SQYLNNGPQRIGRKYKKVRFMAYTDETFKTREAIQHESGILGPLLYGEVGDTLLIIFKNQASRPYNIY PHGITDVRPLYSRRLPKGVKHLKDFPILPGEIFKYKWTVTVEDGPTKSDPRCLTRYYSSFVNMERDLA SGLIGPLLICYKESVDQRGNQIMSDKRNVILFSVFDENRSWYLTENIQRFLPNPAGVQLEDPEFQASN IMHSINGYVFDSLQLSVCLHEVAYWYILSIGAQTDFLSVFFSGYTFKHKMVYEDTLTLFPFSGETVFM SMENPGLWILGCHNSDFRNRGMTALLKVSSCDKNTGDYYEDSYEDISAYLLSKNNAIEPRSFSQNSRH PSTRQKQFNATTIPENDIEKTDPWFAHRTPMPKIQNVSSSDLLMLLRQSPTPHGLSLSDLQEAKYETF SDDPSPGAIDSNNSLSEMTHFRPQLHHSGDMVFTPESGLQLRLNEKLGTTAATELKKLDFKVSSTSNN LISTIPSDNLAAGTDNTSSLGPPSMPVHYDSQLDTTLFGKKSSPLTESGGPLSLSEENNDSKLLESGL MNSQESSWGKNVSSTESGRLFKGKRAHGPALLTKDNALFKVSISLLKTNKTSNNSATNRKTHIDGPSL LIENSPSVWQNILESDTEFKKVTPLIHDRMLMDKNATALRLNHMSNKTTSSKNMEMVQQKKEGPIPPD AQNPDMSFFKMLFLPESARWIQRTHGKNSLNSGQGPSPKQLVSLGPEKSVEGQNFLSEKNKVVVKGKE FTKDVGLKEMVFPSSRNLFLTNLDNLHENNTHNQEKKIQEEIEKKETLIQENVVLPQIHTVTGTKNFM KNLFLLSTRQNVEGSYDGAYAPVLQDFRSLNDSTNRTKKHTAHFSKKGEEENLEGLGNQTKQIVEKYA CTTRISPNTSQQNFVTQRSKRALKQFRLPLEETELEKRIIVDDTSTQWSKNMKHLTPSTLTQIDYNEK EKGAITQSPLSDCLTRSHSIPQANRSPLPIAKVSSFPSIRPIYLTRVLFQDNSSHLPAASYRKKDSGV QESSHFLQGAKKNNLSLAILTLEMTGDQREVGSLGTSATNSVTYKKVENTVLPKPDLPKTSGKVELLP KVHIYQKDLFPTETSNGSPGHLDLVEGSLLQGTEGAIKWNEANRPGKVPFLRVATESSAKTPSKLLDP LAWDMHYGTQIPKEEWKSQEKSPEKTAFKKKDTILSLNACESNHAIAAINEGQNKPEIEVTWAKQGRT ERLCSQNPPVLKRHQREITRTTLQSDQEEIDYDDTISVEMKKEDFDIYDEDENQSPRSFQKKTRHYFI AAVERLWDYGMSSSPHVLRNRAQSGSVPQFKKVVFQEFTDGSFTQPLYRGELNEHLGLLGPYIRAEVE DNIMVTFRNQASRPYSFYSSLISYEEDQRQGAEPRKNFVKPNETKTYFWKVQHHMAPTKDEFDCKAWA YFSDVDLEKDVHSGLIGPLLVCHTNTLNPAHGRQVTVQEFALFFTIFDETKSWYFTENMERNCRAPCN IQMEDPTFKENYRFHAINGYIMDTLPGLVMAQDQRIRWYLLSMGSNENIHSIHFSGHVFTVRKKEEYK MALYNLYPGVFETVEMLPSKAGIWRVECLIGEHLHAGMSTLFLVYSNKCQTPLGMASGHIRDFQTTAS GQYGQWAPKLARLHYSGSINAWSTKEPFSWIKVDLLAPMIIHGIKTQGARQKFSSLYISQFIIMYSLD GKKWQTYRGNSTGTLMVFFGNVDSSGIKHNIFNPPIIARYIRLHPTHYSIRSTLRMELMGCDLNSCSM PLGMESKAISDAQITASSYFTNMFATWSPSKARLHLQGRSNAWRPQVNNPKEWLQVDFQKTMKVTGVT TQGVKSLLTSMYVKEFLISSSQDGHQWTLFFQNGKVKVFQGNQDSFTPVVNSLDPPLLTRYLRIHPQS WVHQIALRMEVLCCEAQDLYDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSH EDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVEHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF FLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK ii) Fc chain (20 amino acid heterologous signal peptide from mouse Igk chain underlined) (SEQ ID NO: 4) METDTLLLWVLLLWVPGSTG DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAK TKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRD ELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFS CSVMHEALHNHYTQKSLSLSPGK

TABLE 8 Additional Sequences SEQ ID NO: 9 >CTP peptide 1 DPRFQDSSSSKAPPPSLPSPSRLPGPSDTPIL SEQ ID NO: 10 >CTP peptide 2 SSSSKAPPPSLPSPSRLPGPSDTPILPQ SEQ ID NO: 11 >PAS peptide 1 ASPAAPAPASPAAPAPSAPA SEQ ID NO: 12 >PAS Peptide 2 AAPASPAPAAPSAPAPAAPS SEQ ID NO: 13 >PAS peptide 3 APSSPSPSAPSSPSPASPSS SEQ ID NO: 14 >PAS peptide 4 APSSPSPSAPSSPSPASPS SEQ ID NO: 15 >PAS peptide 5 SSPSAPSPSSPASPSPSSPA SEQ ID NO: 16 >PAS peptide 6 AASPAAPSAPPAAASPAAPSAPPA SEQ ID NO: 17 >PAS peptide 7 ASAAAPAAASAAASAPSAAA SEQ ID NO: 18 >Albumin Binding Peptide Core Sequence DICLPRWGCLW SEQ In NO: 19 >GFP Protein sequence (Genbank ID AAG34521,1) MSKGEELFTGVVPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTL VTTEGYGVQCFARYPDHMKQBDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLV NRIELKGIDFKEDGNILGEELEYNYNSHNVYTMADKQKNGIKVNFKIRHNIEDGSVQLAD HYQQNTPIGDGPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITHGMDELYKSR TSGSPGLQEFDIKLIDTVDLESCN SEQ ID NO: 20 >Example: Single-chain Human IgG1 Fc. (Fc sequences with Gly/Ser linker underlined.) DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVD GVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKGGGGSGGGGSGGG GSGGGGSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKT TPPVLDSDGSFELYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK SEQ ID NO: 21 >Mature human albumin protein sequence (derived from NCBI Ref. Sequence NP_000468) RGVERRDAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAKTCV ADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPR LVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADK AACLLPKLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKL VTDLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVE NDEMPADLPSLAADEVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLAKTYE TTLEKCCAAADPHECYAKVEDEFKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRYTKK VPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVT KCCTESLVNRRPCFSALEVDETYVPKEFNAETFTFHADICTLSEKERQIKKQTALVELVK HKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEEGKKLVAASQAALGL SEQ ID NO: 22 >Albumin binding peptide 1 RLIEDICLPRWGCLWEDD SEQ ID NO: 23 >Albumin binding peptide 2 QRLMEDICLPRWGCLWEDDF SEQ ID NO: 24 >Albumin binding peptide 3 QGLIGDICLPPWGCLWGDSVK SEQ ID NO: 25 >Albumin binding peptide 4 GEWWEDICLPRWGCLWEEED SEQ ID NO: 26 >Cysteine-containing peptide GGGSGCGGGS SEQ ID NO: 27 >Human LRP1 sequence (signal peptide and transmembrane segment underlined; NCBI Reference Sequence: CAA32112) MLTPPLLLLLPLLSALVAAATDAPKTCSPKQFACRDQITCISKGWRCDGERDCPDGSDEA PEICPQSKAQRCQPNEHNCLGTELCVPMSRLCNGVQDCMDGSDEGPHCRELQGNCSREGC QHHCVPTLDGPTCYCNSSFQLQADGKTCRDFDECSVYGTCSQLCTNTDGSFICGCVEGYL LQPDNRSCKAKNEPVDRPPVLLIANSQNILATYLSGAQVSTITPTSTRQTTAMDFSYANE TVCWVHVGDSAAQTQLKCARMPGLKGFVDEHTINISLSLHHVEQMAIDWLTGNEYFVDDI DDRIFVCNRNGDTCVTLLDLELYNPKGIALDPAMGKVFFTDYGQIPKVERCDMDGQNRTK LVDSKIVFPHGITLDLVSRLVYWADAYLDYIEVVDYEGKGRQTIIQGILIEHLYGLTVFE NYLYATNSDNANAQQKTSVIRVNRENSTEYQVVTRVDKGGALHIYHQRRQPRVRSHACEN DQYGKPGGCSDICLLANSHKARTCRCRSGESLGSDGKSCKKPEHELFLVYGKGRPGIIRG MDMGAKVPDEHMIPIENLMNPRALDFHAETGFIYFADTTSYLIGRQKIDGTERETILKDG IHNVEGVAVDWMGDNLYWTDDGPKKTISVARLEKAAQTRKTLIEGKMTHPRAIVVDPLNG WMYWTDWEEDPKDSRRGRLERAWMDGSHRDIFVTSKTVLWPNGLSLDIPAGRLYWVDAFY DRIETILLNGTDRKIVYEGPELNHAFGLCHHGNYLFWTEYRSGSVYRLERGVGGAPPTVT LLRSERPPIFEIRMYDAQQQQVGTNKCRVNNGGCSSLCLATPGSRQCACAEDQVLDADGV TCLANPSYVPPPQCQPGEFACANERCIQERWKCDGDNDCLDNSDEAPALCHQHTCPSDRF KCENNRCIPNRWLCDGDNDCGNSEDESNATCSARTCPPNQFSCASGRCIPISWTCDLDDD CGDRSDESASCAYPTCFPLTQFTCNNGRCININWRCDNDNDCGDNSDEAGCSHSCSSTQF KCNSGRCIPEHWTCDGDNDCGDYSDETRANCTNQATRPPGGCRTDEFQCRLDGLCIPLRW RCDGDTDCMDSSDEKSCEGVTHVCDPSVKRGCKDSARCTSKAWVCDGDNDCEDNSDEENC ESLACRPPSHPCANNTSVCLPPDKLCDGNDDCGDGSDEGELCDQCSLNNGGCSHNCSVAP GEGIVCSCPLGMELGPDNHTCQIQSYCAKELKCSQKCDQNKFSVKCSCYEGWVLEPDGES CRSLDPFKPFIIFSNRHEIRRIDLHKGDYSVLVPGLRNTIALDFHLSQSALYWTDVVEDK IYRGKLLDNGALTSFEVVIQYGLATPEGIAVDWIAGNIYWVESNLDQIEVAKLDGTLRTT LLAGDIEHPRAIALDPRDGILFWTDWDASLPRIEAASMSGAGRRTVHRETGSGGWPNGLT VDYLEKRILWIDARSDAIYSARYDGSGHMEVLRGHEFLSHPFAVTLYGGEVYWTDWRTNT LAKANKWTGHNVTVVQRTNTQPFDLQVYHPSRQPMAPNPCEANGGQGPCSHLCLINYNRT VSCACPHLMKLHKDNTTCYEFKKFLLYARQMEIRGVDLDAPYYNYIISFTVPDIDNVTVL DYDAREQRVYWSDVRTQAIKRAFINGTGVETVVSADLPNAHGLAVDWVSRNLFWTSYDTN KKQINVARLDGSFKNAVVQGLEQPHGLVVHPLRGKLYWTDGDNISMANMDGSNRTLLFSG QKGPVGLAIDFPESKLYWISEGNHTINRCNLDGSGLEVIDAMRSQLGKATALAIMGDKLW WADQVSEKMGTCSKADGSGSVVLRNSTTLVMHMKVYDESIQLDHKGTNPCSVNNGDCSQL CLPTSETTRSCMCTAGYSLRSGQQACEGVGSPLLYSVHEGIRGIPLDPNDKSDALVPVSG TSLAVGIDFHAENDTIYWVDMGLSTISRAKRDQTWREDVVTNGIGRVEGIAVDWIAGNIY WTDQGFDVIEVARLNGSFRYVVISQGLDKPRAITVHPEKGYLFWTEWGQYPRIERSRLDG TERVVLVNVSISWPNGISVDYQDGKLYWCDARTDKIERIDLETGENREVVLSSNNMDMFS VSVFEDFrYWSDRTHANGSIKRGSKDNATDSVPLRTGIGVQLKDIKVFNRDRQKGTNVCA VANGGCQQLCLYRGRGQRACACAEGMLAEDGASCREYAGYLLYSERTILKSIHLSDERNL NAPVQPFEDPEHMKNVIALAFDYRAGTSPGTPNRIFFSDIHRGNIQQINDDGERRITIVE NVGSVEGLAYHRGWDTLYWTSYTTSTITRHTVDQTRPGAFERETVITMSGDDHPRAFVLD ECQNLMFWTNWNEQHPSIMRAALSGANVLTLIEKDIRTPNGLAIDHRAEKLYFSDATLDK IERCEYDGSHRYVILKSEPVHPFGLAVYGEHIFWTDWVRRAVQRANKHVGSNMKLLRVDI PQQPMGIIAVANDTNSCELSPCRINNGGCQDLCLLTHQGHVNCSCRGGRILQDDLTCRAV NSSCRAQDEFECANGECINFSLTCDGVPHCKDKSDEKPSYCNSRRCKKTFRQCSNGRCVS NMLWCNGADDCGDGSDEIPCNKTACGVGEFRCRDGTCIGNSSRCNQFVDCEDASDEMNCS ATDCSSYFRLGVKGVLFQPCERTSLCYAPSWVCDGANDCGDYSDERDCPGVKRPRCPLNY FACPSGRCIPMSWTCDKEDDCEHGEDETHCNKFCSEAQFECQNHRCISKQWLCDGSDDCG DGSDEAAHCEGKTCGPSSFSCPGTHVCVPERWLCDGDKDCADGADESIAAGCLYNSTCDD REFMCQNRQCIPKHFVCDHDRDCADGSDESPECEYPTCGPSEFRCANGRCLSSRQWECDG ENDCHDQSDEAPKNPHCTSPEHKCNASSQFLCSSGRCVAEALLCNGQDDCGDSSDERGCH INECLSRKLEGCSQDCEDLKIGFKCRCRPGFRLKDDGRTCADVDECSTTFPCSQRCINTH GSYKCLCVEGYAPRGGDPHSCKAVTDEEPFLIFANRYYLRKLNLDGSNYTLLKQGLNNAV ALDFDYREQMTYWTDVTTQGSMIRRMHLNGSNVQVLHRTGLENPDGLAVDWVGGNLYWCD KGRDTIEVSKLNGAYRTVLVSSGLREPRALVVDVQNGYLYWTDWGDHSLIGRIGMDGSSR SVIVDTKITWPNGLTLDYVTERIYWADAREDYIEFASLDGSNRHVVLSQDIPHIFALTLF EDYVYWTDWETKSINRAHKTTGTNKTLLISTLHRPMDLHVFHALRQPDVPNHPCKVNNGG CSNLCLLSPGGGHKCACPTNFYLGSDGRTCVSNCTASQFVCKNDKCIPFWWKCDTEDDCG DHSDEPPDCPEFKCRPGQFQCSTGICTNPAFICDGDNDCQDNSDEANCDIEVCLPSQFKC TNTNRCIPGIFRCNGQDNCGDGEDERDCPEVTCAPNQFQCSITKRCIPRVWVCDRDNDCV DGSDEPANCTQMTCGVDEFRCKDSGRCIPARWKCDGEDDCGDGSDEPKEECDERTCEPYQ FRCKNNRCVPGRWQCDYDNDCGDNSDEESCTPRPCSESEFSCANGRCIAGRWKCDGDHDC ADGSDEKDCTPRCDMDQFQCKSGHCIPLRWRCDADADCMDGSDEEACGTGVRTCPLDEFQ CNNTLCKPLAWKCDGEDDCGDNSDENPEECARFVCPPNRPFRCKNDRVCLWIGRQCDGTD NCGDGTDEEDCEPPTAHTTHCKDKKEFLCRNQRCLSSSLRCNMFDDCGDGSDEEDCSIDP KLTSCATNASICGDEARCVRTEKAAYCACRSGFHTVPGQPGCQDINECLRFGTCSQLCNN TKGGHLCSCARNFMKTENTCKAEGSEYQVLYIADDNEIRSLFPGHPHSAYEQAFQGDESV RIDAMDVEVKAGRVYWTNWHTGTISYRSLPPAAPPTTSNRHRRQIDRGVTELNISGLKMP RGIAIDWVAGNVYWTDSGRDVIEVAQMKGENRKTLISGMIDEPHAIVVDPLRGTMYWSDW GNHPKIETAAMDGTLRETLVQDNIQWPTGLAVDYHNERLYWADAKLEVIGSIRLNGTDPI VAADSKRGLSHPFSIDVFEDYIYGVTYINNRVFKIHKFGHSPLVNLTGGLSHASDVVLYH QHKQPEVIVPCDRKKCEWLCLLSPSGPVCTCPNGKRLDNGTCVPVPSPTPPPDAPRPGTC NLQCFNGGSCFLNARRQPKCRCQPRYTGDKCELDQCWEHCRNGGTCAASPSGMPTCRCPT GFTGPKCTQQVCAGYCANNSTCTVNQGNQPQCRCLPGFLGDRCQYEQCSGYCENFGTCQM AADGSRQCRCTAYFEGSRCEVNKCSRCLEGACVVNKQSGDVTCNCTDGRVAPSCLTCVGH CSNGGSCTMNSKMMPECQCPPHMTGPRCEEHVFSQQQPGHIASILIPLLLLLLLVLVAGV VFWYKRRVQGAKGFQHQRMTNGAMNVEIGNPTYKMYEGGEPDDVGGLLDADFALDPDKPT NFTNPVYATLYMGGHGSRHSLASTDEKRELLGRGPEDEIGDPLA SEQ ID NO: 28 >Biotin Acceptor Peptide (BAP) LNDIFEAQKIEWH SEQ ID NO: 29 >Lipoate Acceptor Peptide 2 (LAP2) GFEIDKVWYDLDA SEQ ID NO: 30 >HAPylation motif, n = 1 to 400 (Gly4Ser)n SEQ ID NO: 31 >CTP DSSSSKAPPPSLPSPSRLPGPSDTPILPQ

Claims

1. A method of providing a clotting factor, comprising administering a fixed dose of a clotting factor to a subject in need thereof.

2. A method of reducing, ameliorating, or preventing one or more symptoms of a bleeding disease or disorder in a subject comprising administering a fixed dose of a clotting factor to a subject in need thereof.

3. (canceled)

4. The method of claim 1 wherein the clotting factor is a modified clotting factor comprising a clotting factor and a heterologous moiety.

5. (canceled)

6. (canceled)

7. The method of claim 4, wherein the heterologous moiety comprises albumin, albumin binding polypeptide, an FcRn binding partner, PAS, the β subunit of the C-terminal peptide (CTP) of human chorionic gonadotropin, polyethylene glycol (PEG), hydroxyethyl starch (HES), albumin-binding small molecules, or combinations thereof.

8. (canceled)

9. The method of claim 1, wherein the fixed dose is administered at regular intervals of every day, every two days, every three days, twice a week, every four days, every five days, every six days, every week, every eight days, every nine days, every 10 days, every 11 days, every 12 days, every 13 days, every two weeks, every three weeks, or every four weeks.

10-41. (canceled)

42. The method of claim 1, wherein the fixed dose is standard across all body weights.

43-46. (canceled)

47. The method of claim 1, wherein the fixed dose is stratified into two or more fixed dose amounts based on specified weight categories.

48-59. (canceled)

60. The method of claim 1, wherein the clotting factor is a long-acting FVIII polypeptide, wherein the long-acting FVIII polypeptide comprises a FVIII polypeptide and a first FcRn binding partner.

61. (canceled)

62. (canceled)

63. The method of claim 60, wherein the long-acting FVIII polypeptide further comprises a second FcRn binding partner.

64-70. (canceled)

71. The method of claim 1, wherein the clotting factor comprises FVIII polypeptide in the long acting polypeptide is a full-length FVIII or a B-domain deleted FVIII.

72. (canceled)

73. The method of claim 1, wherein the clotting factor comprises a FVIII polypeptide comprising an amino acid sequence that is at least 90% or 95% identical to a FVIII amino acid sequence shown in Table 7A or 7B without a signal sequence (amino acids 20 to 1457 of SEQ ID NO: 6 or amino acids 20 to 2351 of SEQ ID NO: 8).

74. (canceled)

75. The method of claim 60, wherein the first FcRn binding partner is at least 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to a Fc amino acid sequence shown in Table 5B without a signal sequence (amino acids 1 to 227 SEQ ID NO:4).

76. (canceled)

77. The method of claim 63, wherein the second FcRn binding partner is at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to a Fc amino acid sequence shown in Table 5B without a signal sequence (amino acids 1 to 227 SEQ ID NO:4).

78. The method of claim 60, wherein the long-acting FVIII polypeptide comprises a sequence at least 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the FVIII and Fc amino acid sequence shown in Table 7A or 7B without a signal sequence (amino acids 1 to 1665 of SEQ ID NO: 6 or amino acids 1 to 2559 of SEQ ID NO: 8).

79. The method of claim 63, wherein the fixed dose is standard across all body weights.

80. (canceled)

81. (canceled)

82. The method of claim 63, wherein the fixed dose is stratified into two or more dose amounts based on specified weight categories.

83-92. (canceled)

93. The method of claim 1, wherein the fixed dose of the clotting factor is to prevent one or more bleeding episodes.

94. (canceled)

95. (canceled)

96. The method of claim 1, wherein the fixed dose of the clotting factor is for perioperative management of a bleeding episode.

97-104. (canceled)

105. The method of claim 1, wherein the subject is in need of on-demand treatment.

106. (canceled)

107. (canceled)

108. The method of claim 1, wherein the subject is in need of surgical prophylaxis, peri-operative management, or treatment for surgery.

109-119. (canceled)

120. A pharmaceutical composition comprising a fixed dose of a modified clotting factor and a pharmaceutically acceptable carrier for use to reduce, ameliorate, or prevent one or more symptoms of a bleeding disease or disorder in a subject in need thereof

121. (canceled)

122. A kit comprising the pharmaceutical composition of claim 120 and instructions to administer the fixed dose of the modified clotting factor to the subject.

123-128. (canceled)

Patent History
Publication number: 20200030420
Type: Application
Filed: Jul 3, 2019
Publication Date: Jan 30, 2020
Inventors: Haiyan JIANG (Belmont, MA), Lei DIAO (Somerville, MA), Shuanglian LI (Lexington, MA), Paula COBB (Lincoln, MA)
Application Number: 16/502,352
Classifications
International Classification: A61K 38/48 (20060101); A61K 38/36 (20060101); C12N 9/64 (20060101);