Modified Coagulation Factors With Prolonged In Vivo Half-life

SCHULTE; Stefan ;   et al.

Patent Application Summary

U.S. patent application number 14/248661 was filed with the patent office on 2014-09-18 for modified coagulation factors with prolonged in vivo half-life. This patent application is currently assigned to CSL Behring GmbH. The applicant listed for this patent is CSL Behring GmbH. Invention is credited to Hubert METZNER, Stefan SCHULTE, Thomas WEIMER.

Application Number20140273096 14/248661
Document ID /
Family ID39155545
Filed Date2014-09-18

United States Patent Application 20140273096
Kind Code A1
SCHULTE; Stefan ;   et al. September 18, 2014

MODIFIED COAGULATION FACTORS WITH PROLONGED IN VIVO HALF-LIFE

Abstract

The present invention relates to nucleic acid sequences coding for modified coagulation factors, preferably coagulation factor VIII, and their derivatives; recombinant expression vectors containing such nucleic acid sequences; host cells transformed with such recombinant expression vectors; and recombinant polypeptides and derivatives coded for by said nucleic acid sequences, whereby said recombinant polypeptides and derivatives have biological activities and prolonged in vivo half-lives compared to the unmodified wild-type proteins. The invention also relates to corresponding sequences that result in improved in vitro stability. The present invention further relates to processes for the manufacture of such recombinant proteins and their derivatives. The invention also relates to a transfer vector for use in human gene therapy, which comprises such nucleic acid sequences.


Inventors: SCHULTE; Stefan; (Marburg, DE) ; WEIMER; Thomas; (Gladenbach, DE) ; METZNER; Hubert; (Marburg, DE)
Applicant:
Name City State Country Type

CSL Behring GmbH

Marburg

DE
Assignee: CSL Behring GmbH
Marburg
DE

Family ID: 39155545
Appl. No.: 14/248661
Filed: April 9, 2014

Related U.S. Patent Documents

Application Number Filing Date Patent Number
12520840 Jul 23, 2009 8754194
PCT/EP2007/011356 Dec 21, 2007
14248661
60879334 Jan 9, 2007

Current U.S. Class: 435/69.6 ; 435/320.1; 435/328; 435/352; 435/354; 435/356; 435/358; 435/365; 435/369; 536/23.4
Current CPC Class: C07K 14/745 20130101; C07K 2319/31 20130101; C07K 14/755 20130101; C07K 14/765 20130101; C07K 16/18 20130101; A61P 7/04 20180101
Class at Publication: 435/69.6 ; 536/23.4; 435/320.1; 435/328; 435/365; 435/356; 435/354; 435/352; 435/369; 435/358
International Class: C07K 14/755 20060101 C07K014/755; C07K 14/765 20060101 C07K014/765; C07K 16/18 20060101 C07K016/18

Foreign Application Data

Date Code Application Number
Dec 22, 2006 EP 06026747.3

Claims



1-32. (canceled)

33. A polynucleotide or a group of polynucleotides encoding a modified factor VIII (FVIII) polypeptide, comprising a FVIII polypeptide having an N-terminal amino acid and a C-terminal amino acid, and a half-life enhancing polypeptide (HLEP) inserted within the B-domain between the N-terminal amino acid and the C-terminal amino acid of the FVIII polypeptide, wherein the FVIII polypeptide is capable of being cleaved from the HLEP moiety during activation in vivo, wherein the modified FVIII polypeptide exhibits a prolonged half-life prior to activation during a bleeding event and a half-life substantially the same as that of an unmodified FVIII peptide following activation, and wherein the HLEP comprises albumin or an immunoglobulin constant region polypeptide.

34. The polynucleotide or group of polynucleotides according to claim 33, wherein the modified FVIII polypeptide has a prolonged functional or antigenic half-life as compared to a FVIII polypeptide lacking an inserted HLEP.

35. The polynucleotide or group of polynucleotides according to claim 33, wherein the modified FVIII polypeptide has an improved in vivo recovery as compared to the FVIII polypeptide lacking an inserted HLEP.

36. The polynucleotide or group of polynucleotides according to claim 33, wherein the modified FVIII polypeptide has increased stability in serum-free culture media and/or in animal protein-free culture media as compared to the FVIII polypeptide lacking an inserted HLEP.

37. The polynucleotide or group of polynucleotides according to claim 33, wherein the B-domain of FVIII or a part thereof is replaced with the HLEP.

38. The polynucleotide or group of polynucleotides according to claim 37, wherein more than 75% of the B-domain is deleted, or more than 75% of the B-domain is replaced by linker sequences.

39. The polynucleotide or group of polynucleotides according to claim 33, wherein the modified FVIII polypeptide has at least 10% of the biological activity of the FVIII polypeptide lacking an inserted HLEP.

40. The polynucleotide or group of polynucleotides according to claim 33, wherein the half-life enhancing polypeptide is albumin.

41. The polynucleotide or group of polynucleotides according to claim 33, wherein the B-domain of FVIII has been replaced partly or completely with human albumin.

42. The polynucleotide or group of polynucleotides according to claim 33, wherein the immunoglobulin constant region polypeptide is an immunoglobulin G Fc domain.

43. A plasmid or vector comprising the polynucleotide according to claim 33, or a group of plasmids or vectors comprising the group of polynucleotides according to claim 33.

44. The plasmid or vector, or the group of plasmids or vectors, according to claim 43, wherein the plasmid(s) or vector(s) are expression vector(s).

45. The vector, or the group of vectors, according to claim 43, wherein the vector(s) are transfer vector(s) for use in human gene therapy.

46. A host cell comprising the polynucleotide or group of polynucleotides according to claim 33.

47. A method of producing a modified FVIII polypeptide, comprising culturing the host cell according to claim 46 under conditions such that the modified FVIII polypeptide is expressed.
Description



FIELD OF THE INVENTION

[0001] The present invention relates to modified nucleic acid sequences coding for coagulation factors preferably coagulation factor VIII and their derivatives, recombinant expression vectors containing such nucleic acid sequences, host cells transformed with such recombinant expression vectors, recombinant polypeptides and derivatives coded for by said nucleic acid sequences which recombinant polypeptides and derivatives do have biological activities together with prolonged in vivo half-life and/or improved in vivo recovery compared to the unmodified wild-type protein. The invention also relates to corresponding sequences that result in improved in vitro stability. The present invention further relates to processes for the manufacture of such recombinant proteins and their derivatives. The invention also relates to a transfer vector for use in human gene therapy, which comprises such modified nucleic acid sequences.

BACKGROUND OF THE INVENTION

[0002] There are various bleeding disorders caused by deficiencies of blood coagulation factors. The most common disorders are hemophilia A and B, resulting from deficiencies of blood coagulation factor VIII and IX, respectively. Another known bleeding disorder is von Willebrand's disease.

[0003] Classic hemophilia or hemophilia A is an inherited bleeding disorder. It results from a chromosome X-linked deficiency of blood coagulation Factor VIII, and affects almost exclusively males with an incidence of between one and two individuals per 10.000. The X-chromosome defect is transmitted by female carriers who are not themselves hemophiliacs. The clinical manifestation of hemophilia A is an increased bleeding tendency. Before treatment with Factor VIII concentrates was introduced the mean life span for a person with severe hemophilia was less than 20 years. The use of concentrates of Factor VIII from plasma has considerably improved the situation for the hemophilia A patients increasing the mean life span extensively, giving most of them the possibility to live a more or less normal life. However, there have been certain problems with the plasma derived concentrates and their use, the most serious of which have been the transmission of viruses. So far, viruses causing hepatitis B, non-A non-B hepatitis and AIDS have hit the population seriously. Since then different virus inactivation methods and new highly purified Factor VIII concentrates have recently been developed which established a very high safety standard also for plasma derived Factor VIII.

[0004] The cloning of the cDNA for Factor VIII (Wood et al. 1984. Nature 312:330-336; Vehar et al. 1984. Nature 312:337-342) made it possible to express Factor VIII recombinantly leading to the development of several recombinant Factor VIII products, which were approved by the regulatory authorities between 1992 and 2003. The fact that the central B domain of the Factor VIII polypeptide chain residing between amino acids Arg-740 and Glu-1649 does not seem to be necessary for full biological activity has also led to the development of a B domain deleted Factor VIII.

[0005] The mature Factor VIII molecule consists of 2332 amino acids which can be grouped into three homologous A domains, two homologous C domains and a B Domain which are arranged in the order: A1-A2-B-A3-C1-C2. The complete amino acid sequence of mature human Factor VIII is shown in SEQ ID NO:2. During its secretion into plasma Factor VIII is processed intracellularly into a series of metal-ion linked heterodimers as single chain Factor VIII is cleaved at the B-A3 boundary and at different sites within the B-domain. This processing leads to heterogenoeous heavy chain molecules consisting of the A1, the A2 and various parts of the B-domain which have a molecular size ranging from 90 kDa to 200 kDa. The heavy chains are bound via a metal ion to the light chains, which consist of the A3, the C1 and the C2 domain (Saenko et al. 2002. Vox Sang. 83:89-96). In plasma this heterodimeric Factor VIII binds with high affinity to von Willebrand Factor (vWF), which protects it from premature catabolism. The half-life of non-activated Factor VIII bound to vWF is about 12 hours in plasma.

[0006] Coagulation Factor VIII is activated via proteolytic cleavage by FXa and thrombin at amino acids Arg372 and Arg740 within the heavy chain and at Arg1689 in the light chain resulting in the release of von Willebrand Factor and generating the activated Factor VIII heterotrimer which will form the tenase complex on phospholipid surfaces with FIXa and FX provided that Ca.sup.2+ is present. The heterotrimer consists of the A1 domain, a 50 kDa fragment, the A2 domain, a 43 kDa fragment and the light chain (A3-C1-C2), a 73 kDa fragment. Thus the active form of Factor VIII (Factor VIIIa) consists of an A1-subunit associated through the divalent metal ion linkage to a thrombin-cleaved A3-C1-C2 light chain and a free A2 subunit relatively loosely associated with the A1 and the A3 domain.

[0007] To avoid excessive coagulation, Factor VIIIa must be inactivated soon after activation. The inactivation of Factor VIIIa via activated Protein C (APC) by cleavage at Arg336 and Arg562 is not considered to be the major rate-limiting step. It is rather the dissociation of the non covalently attached A2 subunit from the heterotrimer which is thought to be the rate limiting step in Factor VIIIa inactivation after thrombin activation (Fay et al. 1991. J. Biol. Chem. 266 8957, Fay & Smudzin 1992. J. Biol. Chem. 267:13246-50). This is a rapid process, which explains the short half-life of Factor VIIIa in plasma, which is only 2.1 minutes (Saenko et al. 2002. Vox Sang. 83:89-96).

[0008] In severe hemophilia A patients undergoing prophylactic treatment Factor VIII has to be administered intravenously (i.v.) about 3 times per week due to the short plasma half-life of Factor VIII of about 12 hours. Each i.v. administration is cumbersome, associated with pain and entails the risk of an infection especially as this is mostly done at home by the patients themselves or by the parents of children being diagnosed for hemophilia A.

[0009] It would thus be highly desirable to create a Factor VIII with increased functional half-life allowing the manufacturing of pharmaceutical compositions containing Factor VIII, which have to be administered less frequently.

[0010] Several attempts have been made to prolong the half-life of non-activated Factor VIII either by reducing its interaction with cellular receptors (WO 03/093313A2, WO 02/060951A2), by covalently attaching polymers to Factor VIII (WO 94/15625, WO 97/11957 and U.S. Pat. No. 4,970,300) or by encapsulation of Factor VIII (WO 99/55306).

[0011] In WO 97/03193 it was speculated that the introduction of novel metal binding sites could stabilize Factor VIII and in particular mutants in which His or Met is substituted for any of Phe652, Tyr1786, Lys1818, Asp1840 and/or Asn1864. However no rationale was provided how to determine the success meaning the stabilization resulting from such modifications nor a rationale why the proposed amino acids were chosen. This approach remains speculative, as no further evidence was published since.

[0012] Another approach has been made in creating a Factor VIIIa, which is inactivation resistant by first covalently attaching the A2 domain to the A3 domain and secondly by mutating the APC cleavage sites (Pipe & Kaufman. 1997. PNAS 94:11851-11856, WO 97/40145 and WO 03/087355.). The underlying genetic construct was also used to produce transgenic animals as described in WO 02/072023A2. The instant variant showed still 38% of its peak activity 4 h after thrombin activation but lacks the vWF binding domain since by fusing the A2 to the A3 domain this particular domain was deleted. For the reason that vWF binding significantly prolongs half-life of FVIII in vivo, it is to be expected that half-life of the non-activated form of the instant FVIII variant is compromised. The inventors themselves recognized this and tried to overcome the problem by adding an antibody which stablizes the light chain in a conformation which retains some affinity for vWF.

[0013] Gale et al. 2002 (Protein Science 11:2091-2101) published the stabilization of FVa by covalently attaching the A3 domain to the A2 domain. They identified two neighbouring amino acids according to structural predictions, one on the A2 domain and the other being located on the A3 domain, and replaced these two amino acids with cysteine residues, which formed a disulfide bridge during export into the endoplasmatic reticulum. The same approach was used to covalently attach via disulfide bridges the A2 to the A3 domain of Factor VIII (WO 02/103024A2). Such Factor VIII mutants with covalently attached A3 and A2 domains, thus stabilizing FVIIIa, retained about 90% of their initial highest activity for 40 minutes after activation whereas the activity of wild type Factor VIII quickly diminished to 10% of its initial highest activity. The Factor VIII mutants retained their 90% activity for additional 3 h without any further loss of activity (Gale et al. 2003. J. Thromb. Haemost. 1:1966-1971).

[0014] WO2006/108590 discloses several stabilized FVIII mutants characterized by the insertion of different peptidic linkers substituting the thrombin activation site at Arg372 also stabilizing the activated form of FVIII. The level of FVIII activity increased concomitantly with the length of the linker reaching a maximum when 99 amino acids (L99) were inserted. Using a chromogenic assay method, the FVIII activity detected with FVIII L99 was similar to FVIII WT. Activated FVIII L99 was almost stable during more than 1 hour.

[0015] As none of the above described approaches has yet resulted in an improved FVIII molecule applicable in patients there is an ongoing need to develop modified coagulation factor VIII molecules which exhibit prolonged half-life.

[0016] In view of a potential thrombogenic risk it is more desirable to prolong the half-life of the non-activated form of FVIII than that of FVIIIa.

[0017] Another problem generally encountered with rec FVIII production is poor yield. Various methods known to the man of the art have been tried, but have not resolved such problem of poor yield.

DESCRIPTION OF THE INVENTION

[0018] It is an objective of this invention to provide blood coagulation molecules with enhanced in vivo half-life.

[0019] It is another objective of this invention to provide blood coagulation molecules with improved in vivo recovery.

[0020] Another objective of the invention is that these modified blood coagulation molecules can be expressed by mammalian cells and retain their biological activity in the expressed modified proteins.

[0021] Another objective of the invention is to provide an improved yield by increased expression and/or increased stability of the coagulation molecules in mammalian cell culture.

[0022] Yet another objective of the invention is to provide FVIII molecules with increased stability in mammalian cell culture in serum- and/or animal protein-free culture media, especially in the absence of vWF.

[0023] It was now surprisingly found that inserting heterologous polypeptides such as albumin into the FVIII molecule, preferably such that they replace the FVIII B domain almost completely or in part, not only permits expression and secretion of FVIII chimeric proteins from mammalian cells but also results in modified FVIII molecules that retain significant FVIII activity. In addition, such modified FVIII molecules exhibit prolonged in vivo half-life and/or improved in vivo recovery.

[0024] An additional potential benefit of those embodiments of the present invention in which FVIII is the coagulation factor and the A2 domain remains not covalently attached to the A3 domain after activation is that only the half-life of the non-activated form of FVIII is increased, whereas the half-life of the activated form of FVIII remains essentially the same, which might result in a decreased risk of thrombogenicity.

[0025] It was furthermore found that the FVIII molecules of the invention are more stable than wild-type FVIII in mammalian cell culture, especially in the absence of stabilizing von Willebrand factor (vWF) in serum- and/or animal protein-free culture media.

[0026] Such molecules can be generated by inserting a half-life enhancing protein (HLEP) moiety into the amino acid sequence of the blood coagulation factor, e.g. into the FVIII molecule. If FVIII is the blood coagulation factor the HLEP is preferably inserted into or replaces the B domain of FVIII or part of it.

[0027] HLEPs in the sense of the present invention are selected from a group consisting of members of the albumin family, which includes albumin, afamin, alpha-fetoprotein and the vitamin D binding protein, as well as portions of an immunoglobulin constant region and polypeptides capable of binding under physiological conditions to members of the albumin family as well as to portions of an immunoglobulin constant region. The most preferred HLEP is human albumin.

[0028] Also encompassed by the invention are other proteins in which HLEPs are inserted into other coagulation factors such as von Willebrand factor, factor V and prothrombin factors including factor VII, factor IX, factor X, protein C, protein S, protein Z and prothrombin. Similar to FVIII described above the particular HLEP, preferably albumin, is inserted in preferred embodiments at or in the vicinity of junction sites of domains or subunits of the coagulation factors above.

[0029] In the prior art fusions of coagulation factors to albumin (WO 01/79271), alpha-fetoprotein (WO 2005/024044) and immunoglobulin (WO 2004/101740) as half-life enhancing polypeptides have been described. These were taught to be attached to the carboxy- or the amino-terminus or to both termini of the respective therapeutic protein moiety, occasionally linked by peptidic linkers, preferably by linkers consisting of glycine and serine.

[0030] Ballance et al. (WO 01/79271) described N- or C-terminal fusion polypeptides of a multitude of different therapeutic polypeptides fused to human serum albumin. Long lists of potential fusion partners are described without disclosing experimental data for almost any of these polypeptides whether or not the respective albumin fusion proteins actually retain biological activity and have improved properties. Among said list of therapeutic polypeptides also Factor VIII is mentioned.

[0031] Contrary to prior art fusion proteins, the heterologous amino acid sequence in the modified coagulation factor of this invention is not fused to the very N-terminus or C-terminus of the coagulation factor, but inserted within an internal region of the amino acid sequence of the coagulation factor. Surprisingly, the insertion of even large polypeptides did not result in a complete loss of biological activity of the coagulation factor. Rather, the thus modified coagulation factor had biological activity, increased in vivo functional half-life, in vivo recovery and increased stability.

[0032] The present invention therefore relates to a modified coagulation factor having at an internal region of the coagulation factor an insertion of a half-life enhancing polypeptide (HLEP), characterized in that the modified coagulation factor has prolonged functional half-life compared to the functional half-life of the coagulation factor lacking said insertion, and/or compared to the functional half-life of the wild type coagulation factor.

[0033] The present invention also relates to the insertion of more than one HLEP wherein the HLEP, which is inserted several times, may be the same HLEP or may be a combination of different HLEPs. Also combinations of insertions of one or more HLEPs at an internal region of the coagulation factor with additional N- and/or C-terminal fusions of one or more HLEPs, which could be the same HLEP or a combination of different HLEPs are encompassed by the invention.

[0034] The present invention also relates to a modified coagulation factor having at an internal region of the coagulation factor an insertion of a half-life enhancing polypeptide (HLEP), characterized in that the modified coagulation factor has improved in vivo recovery compared to the in vivo recovery of the coagulation factor lacking said insertion, and/or compared to the in vivo recovery of the wild type coagulation factor.

[0035] In another aspect of the invention the modified coagulation factor has increased stability in serum-free culture media, compared to that of the coagulation factor lacking said insertion, and/or compared to the stability of the wild type coagulation factor. In another aspect of the invention the modified coagulation factor has increased stability in animal protein-free culture media, compared to that of the coagulation factor lacking said insertion, and/or compared to the stability of the wild type coagulation factor. The increased stability in serum-free and/or animal-free culture media is especially pronounced if stabilizing amounts of vWF are missing.

[0036] Animal protein-free media in the sense of the invention are media free from proteins or protein fragments derived from animals.

[0037] Another aspect of the invention are polynucleotides or sets of polynucleotides encoding the modified coagulation factor of the invention.

[0038] The invention further relates to plasmids or vectors comprising a polynucleotide described herein, to host cells comprising a polynucleotide or a plasmid or vector described herein.

[0039] Another aspect of the invention is a method of producing a modified coagulation factor, comprising: [0040] (a) culturing host cells of the invention under conditions such that the modified coagulation factor is expressed; and [0041] (b) optionally recovering the modified coagulation factor from the host cells or from the culture medium.

[0042] The invention further pertains to pharmaceutical compositions comprising a modified coagulation factor, a polynucleotide, or a plasmid or vector described herein.

[0043] Yet another aspect of the invention is the use of a modified coagulation factor, a polynucleotide, or a plasmid or vector, or of a host cell according to this invention for the manufacture of a medicament for the treatment or prevention of a blood coagulation disorder.

DETAILED DESCRIPTION OF THE INVENTION

[0044] The invention pertains to a modified coagulation factor comprising at an internal region between the N-terminal amino acid and the C-terminal amino acid of the primary translation polypeptide of the coagulation factor an insertion of a half-life enhancing polypeptide (HLEP), characterized in that the modified coagulation factor has prolonged functional half-life compared to the functional half-life of the coagulation factor lacking said insertion, and/or compared to the functional half-life of the wild type coagulation factor.

[0045] The "functional half-life" according to the present invention is the half-life of the biological function of the coagulation factor once it has been administered to a mammal and can be measured in vitro in blood samples taken at different time intervals from said mammal after the coagulation factors has been administered.

[0046] The phrases "insertion", "inserting" and "inserted" refer to the addition of amino acids at an internal position of the coagulation factor amino acid sequence. Other than in the case of N-terminal or C-terminal fusion proteins, the amino acids are according to this invention not added to the very N-terminus or C-terminus of the coagulation factor amino acid sequence, but inserted at an internal position within the amino acid sequence of the coagulation factor. "Insertion" encompasses not only the addition of amino acids (without deleting amino acids from the coagulation factor amino acid sequence), but also the replacement of one or more amino acids of the coagulation factor amino acid sequence with the amino acids to be "inserted". For example, a complete internal domain or a substantial part thereof may be replaced with the HLEP.

[0047] In one embodiment, the modified coagulation factor has the following structure:

N-L1-H-L2-C, [formula 1]

[0048] wherein

[0049] N is an N-terminal portion of a coagulation factor,

[0050] L1 and L2 independently are chemical bonds or linker sequences, which linker sequences can be different linker sequences or the same linker sequences,

[0051] H is a HLEP, and

[0052] C is a C-terminal portion of the coagulation factor.

[0053] Preferably, N comprises one or two or three or four or five protein domains that are present at the N-terminus of the wild type coagulation factor. C preferably comprises one or two or three or four or five protein domains that are present at the C-terminus of the wild type coagulation factor. In one embodiment, the wild type coagulation factor has substantially the structure N--C. In another embodiment, the wild type coagulation factor has substantially the structure N-D-C, wherein D represents a domain or a part thereof that is replaced with the HLEP in the modified coagulation factor or in other words D represents a deletion of a part of the wild type coagulation factor (i.e. a complete domain or part thereof) which is replaced with the HLEP in the modified coagulation factor. Preferred coagulation factor sequences are described infra. Usually, the length of N+C does not exceed that of the wild type coagulation factor.

[0054] L1 and L2 may independently be chemical bonds or linker sequences consisting of one or more amino acids, e.g. of 1 to 20, 1 to 15, 1 to 10, 1 to 5 or 1 to 3 (e.g. 1, 2 or 3) amino acids and which may be equal or different from each other. Usually, the linker sequences are not present at the corresponding position in the wild type coagulation factor. Examples of suitable amino acids present in L1 and L2 include Gly and Ser.

[0055] Preferred HLEP sequences are described infra. The modified coagulation factor of the invention may comprise more than one HLEP sequence, e.g. two or three HLEP sequences. These multiple HLEP sequences may be inserted in tandem, e.g. as successive repeats, or they may be present at different positions of the coagulation factor sequence including also fusions of HLEP sequences at the very N-terminus or at the very C-terminus or at both termini of the coagulation factor sequence, wherein at least one HLEP sequence must be inserted at an internal position within the coagulation factor sequence. In these embodiments, the modified coagulation factor may have one of the following structures:

N-L1-H-L2-I-L3-H-L4-C [formula 2]

N-L1-H-L2-C-L3-H [formula 3]

H-L1-N-L2-H-L3-C [formula 4]

H-L1-N-L2-H-L3-C-L4-H [formula 5]

[0056] wherein

[0057] N is an N-terminal portion of a coagulation factor,

[0058] L1, L2, L3 and L4 independently are chemical bonds or linker sequences, which linker sequences can be different linker sequences or the same linker sequences,

[0059] H is a HLEP,

[0060] I is an internal sequence of the coagulation factor and

[0061] C is a C-terminal portion of the coagulation factor.

[0062] Coagulation factors may be processed proteolytically at various stages. For example, as mentioned supra, during its secretion into plasma single chain Factor VIII is cleaved intracellularly at the B-A3 boundary and at different sites within the B-domain. The heavy chain is bound via a metal ion to the light chain having the domain structure A3-C1-C2. Factor VIII is activated via proteolytic cleavage at amino acids Arg372 and Arg740 within the heavy chain and at Arg1689 in the light chain generating the activated Factor VIII heterotrimer consisting of the A1 domain, the A2 domain, and the light chain (A3-C1-C2), a 73 kDa fragment. Thus the active form of Factor VIII (Factor VIIIa) consists of an A1-subunit associated through the divalent metal ion linkage to a thrombin-cleaved A3-C1-C2 light chain and a free A2 subunit relatively loosely associated with the A1 and the A3 domain.

[0063] Accordingly, the present invention encompasses also modified coagulation factors that are not present as single chain polypeptides but consist of several polypeptides (e.g. one or two or three) that are associated with each other via non-covalent linkages. By way of example, the structure of the modified coagulation factor may be as follows:

N-L1-H-L2 . . . C, [formula 6]

N-L1-H . . . L2-C, [formula 7]

N-L1 . . . H-L2-C, [formula 8]

N . . . L1-H-L2-C, [formula 9]

wherein " . . . " signifies a non-covalent linkage, and the meaning of N, L1, L2, H and C is as defined above. Cleaved forms analogous to those of formula 6 to formula 9 of polypeptides according to formula 2 to formula 5 are also encompassed by the invention.

[0064] Usually, the site of insertion is chosen such that the biological activity of the coagulation factor is retained in full or at least in part. Preferably, the biological activity of the modified coagulation factor of the invention is at least 25%, more preferably at least 50%, most preferably at least 75% of biological activity of the coagulation factor lacking the insertion or of the wild type form of the coagulation factor.

[0065] Generally, insertion between two domains of the coagulation factor or within the vicinity of the boundary between two domains is preferred. The two domains may be adjacent domains in the wild type coagulation factor or not.

[0066] When referring herein to an insertion between two domains (e.g. an "insertion between domain X and domain Y"), this preferably means an insertion exactly between the C-terminal amino acid of domain X and the N-terminal amino acid of domain Y. However, an "insertion between domain X and domain Y" in the sense of this invention may also include an insertion at an amino acid position up to n amino acids upstream to the C-terminal amino acid of domain X, or at an amino acid position up to n amino acids downstream to the N-terminal amino acid of domain Y. The figure n is an integer that should not be greater than 10%, preferably not greater than 5% of the total number of amino acids of the domain referred to. Usually, n is 20, preferably 15, more preferably 10, still more preferably 5 or less (e.g. 1, 2, 3, 4 or 5).

[0067] It is also preferred that the stability of the modified coagulation factor in serum-free medium is greater than that of the coagulation factor lacking the insertion and/or that of the wild type form of the coagulation factor. It is also preferred that the stability of the modified coagulation factor in animal protein-free medium is greater than that of the coagulation factor lacking the insertion and/or that of the wild type form of the coagulation factor. Preferably the increase in stability compared to the coagulation factor lacking the insertion and/or to the wild type form of the coagulation factor is at least 10%, more preferably at least 25%, most preferably at least 50%. The stability of the coagulation factor in those media can be determined as described in example 7.

[0068] The functional half-life according to the present invention is the half-life of the biological function of the coagulation factor once it has been administered to a mammal and is measured in vitro. The functional half-life of the modified coagulation factor according to the invention is greater than that of the coagulation factor lacking the modification as tested in the same species. The functional half-life is preferably increased by at least 25%, more preferably by at least 50%, and even more preferably by at least 100% compared to the coagulation factor lacking the modification and/or to the wild type form of the coagulation factor.

[0069] The functional half-life of a modified coagulation factor comprising a HLEP modification, can be determined by administering the respective modified coagulation factor (and in comparison that of the non-modified coagulation factor) to rats, rabbits or other experimental animal species intravenously or subcutaneously and following the elimination of the biological activity of said modified or respectively non-modified coagulation factor in blood samples drawn at appropriate intervals after application. Suitable test methods are the activity tests described herein.

[0070] As a surrogate marker for the half-life of biological activity also the levels of antigen of the modified or respectively non-modified coagulation factor can be measured. Thus also encompassed by the invention are modified coagulation factors having at an internal region between the N-terminal amino acid and the C-terminal amino acid of the primary translation polypeptide of the coagulation factor an insertion of a half-life enhancing polypeptide (HLEP), characterized in that the modified coagulation factor has a prolonged half-life of the coagulation factor antigen compared to the half-life of the coagulation factor antigen lacking said insertion. The "half-life of the coagulation factor antigen" according to the present invention is the half-life of the antigen of the coagulation factor once it has been administered to a mammal and is measured in vitro. Antigen test methods based on specific antibodies in an enzyme immunoassay format as known to the man of the art and commercially available (e.g. Dade Behring, Instrumentation Laboratory, Abbott Laboratories, Diagnostica Stago). Functional and antigen half-lives can be calculated using the time points of the beta phase of elimination according to the formula t.sub.1/2=ln 2/k, whereas k is the slope of the regression line.

[0071] Once a coagulation factor is activated in vivo during coagulation, it may be no longer desirable to maintain the increased half-life of the now activated coagulation factor as this might lead to thrombotic complications what is already the case for a wild type activated coagulation factor FVIIa (Aledort 2004. J Thromb Haemost 2:1700-1708) and what should be much more possibly threatening if the activated factor would have an increased half-life. It is therefore another objective of the present invention to provide long-lived coagulation factor molecules, which after endogenous activation in vivo or after availability of a cofactor in vivo do have a functional half-life comparable to that of an unmodified coagulation factor. This can be achieved by maintaining certain cleavage sites in the modified coagulation factor (see infra) leading to a proteolytic cleavage during activation which separates the coagulation factor from the HLEP. Accordingly, in one embodiment, the functional half-life of the endogenously activated modified coagulation factor is substantially the same as that of the activated non-modified coagulation factor lacking the modification, and/or it is substantially the same as that of the activated wild type coagulation factor (e.g. .+-.15%, preferably .+-.10%).

[0072] In another embodiment, the functional half-life of the endogenously activated modified coagulation factor is prolonged compared to that of the activated non-modified coagulation factor lacking the insertion, or compared to that of the activated wild type coagulation factor. The increase may be more than 15%, for example at least 20% or at least 50%. Again, such functional half-life values can be measured and calculated as described for functional half-lives supra. Increased half-lives of the endogenously activated modified coagulation factors may be beneficial in situations were only very low levels of the coagulation factors are available that therefore are not thrombogenic. Such situations may occur e.g. upon gene therapy treatment where often only low expression rates can be achieved. Therefore, such stabilized coagulation factors might be beneficial in e.g. gene therapy despite a thrombogenic risk connected to such coagulation factors if administered as proteins in high or physiologic doses.

[0073] Half-Life Enhancing Polypeptides (HLEPs)

[0074] A "half-life enhancing polypeptide" as used herein is selected from the group consisting of albumin, a member of the albumin-family, the constant region of immunoglobulin G and fragments thereof and polypeptides capable of binding under physiological conditions to albumin, to members of the albumin family as well as to portions of an immunoglobulin constant region. It may be a full-length half-life-enhancing protein described herein (e.g. albumin, a member of the albumin-family or the constant region of immunoglobulin G) or one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity or the biological activity of the coagulation factor. Such fragments may be of 10 or more amino acids in length or may include at least about 15, at least about 20, at least about 25, at least about 30, at least about 50, at least about 100, or more contiguous amino acids from the HLEP sequence or may include part or all of specific domains of the respective HLEP, as long as the HLEP fragment provides a functional half-life extension of at least 25% compared to a wild type coagulation factor.

[0075] The HLEP portion of the proposed coagulation factor insertion constructs of the invention may be a variant of a normal HLEP. The term "variants" includes insertions, deletions and substitutions, either conservative or non-conservative, where such changes do not substantially alter the active site, or active domain which confers the biological activities of the modified coagulation factors.

[0076] In particular, the proposed FVIII HLEP insertion or B domain replacement constructs of the invention may include naturally occurring polymorphic variants of HLEPs and fragments of HLEPs. The HLEP may be derived from any vertebrate, especially any mammal, for example human, monkey, cow, sheep, or pig. Non-mammalian HLEPs include, but are not limited to, hen and salmon.

[0077] Albumin as HLEP

[0078] The terms, "human serum albumin" (HSA) and "human albumin" (HA) and "albumin" (ALB) are used interchangeably in this application. The terms "albumin" and "serum albumin" are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).

[0079] As used herein, "albumin" refers collectively to albumin polypeptide or amino acid sequence, or an albumin fragment or variant, having one or more functional activities (e.g., biological activities) of albumin. In particular, "albumin" refers to human albumin or fragments thereof, especially the mature form of human albumin as shown in SEQ ID NO:3 herein or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof.

[0080] In particular, the proposed coagulation factor insertion constructs of the invention may include naturally occurring polymorphic variants of human albumin and fragments of human albumin. Generally speaking, an albumin fragment or variant will be at least 10, preferably at least 40, most preferably more than 70 amino acids long. The albumin variant may preferentially consist of or alternatively comprise at least one whole domain of albumin or fragments of said domains, for example domains 1 (amino acids 1-194 of SEQ ID NO:3), 2 (amino acids 195-387 of SEQ ID NO: 3), 3 (amino acids 388-585 of SEQ ID NO: 3), 1+2 (1-387 of SEQ ID NO: 3), 2+3 (195-585 of SEQ ID NO: 3) or 1+3 (amino acids 1-194 of SEQ ID NO: 3+amino acids 388-585 of SEQ ID NO: 3). Each domain is itself made up of two homologous subdomains namely 1-105, 120-194, 195-291, 316-387, 388-491 and 512-585, with flexible inter-subdomain linker regions comprising residues Lys106 to Glu119, Glu292 to Val315 and Glu492 to Ala511.

[0081] The albumin portion of the proposed coagulation factor insertion constructs of the invention may comprise at least one subdomain or domain of HA or conservative modifications thereof.

[0082] Afamin, Alpha-Fetoprotein and Vitamin D Binding Protein as HLEPs

[0083] Besides albumin, alpha-fetoprotein, another member of the albumin family, has been claimed to enhance the half-life of an attached therapeutic polypeptide in vivo (WO 2005/024044). The albumin family of proteins, evolutionarily related serum transport proteins, consists of albumin, alpha-fetoprotein (AFP; Beattie & Dugaiczyk 1982. Gene 20:415-422), afamin (AFM; Lichenstein et al. 1994. J. Biol. Chem. 269:18149-18154) and vitamin D binding protein (DBP; Cooke & David 1985. J. Clin. Invest. 76:2420-2424). Their genes represent a multigene cluster with structural and functional similarities mapping to the same chromosomal region in humans, mice and rat. The structural similarity of the albumin family members suggest their usability as HLEPs. It is therefore another object of the invention to use such albumin family members, fragments and variants thereof as HLEPs. The term "variants" includes insertions, deletions and substitutions, either conservative or non-conservative as long as the desired function is still present.

[0084] Albumin family members may comprise the full length of the respective protein AFP, AFM and DBP, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity. Such fragments may be of 10 or more amino acids in length or may include about 15, 20, 25, 30, 50, or more contiguous amino acids of the respective protein sequence or may include part or all of specific domains of the respective protein, as long as the HLEP fragments provide a half-life extension of at least 25%. Albumin family members of the insertion proteins of the invention may include naturally occurring polymorphic variants of AFP, AFM and DBP.

[0085] Immunoglobulins as HLEPs

[0086] Immunoglobulin G (IgG) constant regions (Fc) are known in the art to increase the half-life of therapeutic proteins (Dumont J A et al. 2006. BioDrugs 20:151-160). The IgG constant region of the heavy chain consists of 3 domains (CH1-CH3) and a hinge region. The immunoglobulin sequence may be derived from any mammal, or from subclasses IgG1, IgG2, IgG3 or IgG4, respectively. IgG and IgG fragments without an antigen-binding domain may also be used as HLEPs. The therapeutic polypeptide portion is connected to the IgG or the IgG fragments preferably via the hinge region of the antibody or a peptidic linker, which may even be cleavable. Several patents and patent applications describe the fusion of therapeutic proteins to immunoglobulin constant regions to enhance the therapeutic protein's in vivo half-lifes. US 2004/0087778 and WO 2005/001025 describe fusion proteins of Fc domains or at least portions of immunoglobulin constant regions with biologically active peptides that increase the half-life of the peptide, which otherwise would be quickly eliminated in vivo. Fc-IFN-.beta. fusion proteins were described that achieved enhanced biological activity, prolonged circulating half-life and greater solubility (WO 2006/000448). Fc-EPO proteins with a prolonged serum half-life and increased in vivo potency were disclosed (WO 2005/063808) as well as Fc fusions with G-CSF (WO 2003/076567), glucagon-like peptide-1 (WO 2005/000892), clotting factors (WO 2004/101740) and interleukin-10 (U.S. Pat. No. 6,403,077), all with half-life enhancing properties.

[0087] Coagulation Factors

[0088] The term "coagulation factor" as used herein denotes a blood coagulation factor or blood clotting factor. Coagulation factors include factor VIII, von Willebrand factor, prothrombin factors (comprising factor VII, Factor IX, factor X, protein C, protein S, protein Z and prothrombin) and coagulation factor V.

[0089] Coagulation factors of the present invention may also be variants of wild-type coagulation factors. The term "variants" includes insertions, deletions and substitutions, either conservative or non-conservative, where such changes do not substantially alter the active site, or active domain, which confers the biological activities of the respective coagulation factor.

[0090] FVIII

[0091] The terms "blood coagulation Factor VIII", "Factor VIII" and FVIII'' are used interchangeably herein. "Blood coagulation Factor VIII" includes wild type blood coagulation Factor VIII as well as derivatives of wild type blood coagulation Factor VIII having the procoagulant activity of wild type blood coagulation Factor VIII. Derivatives may have deletions, insertions and/or additions compared with the amino acid sequence of wild type Factor VIII. The term FVIII includes proteolytically processed forms of Factor VIII, e.g. the form before activation, comprising heavy chain and light chain.

[0092] The term "Factor VIII" includes any Factor VIII variants or mutants having at least 10%, preferably at least 25%, more preferably at least 50%, most preferably at least 75% of the biological activity of wild type factor VIII.

[0093] As non-limiting examples, Factor VIII molecules include Factor VIII mutants preventing or reducing APC cleavage (Amano 1998. Thromb. Haemost. 79:557-563), Factor VIII mutants further stabilizing the A2 domain (WO 97/40145), FVIII mutants resulting in increased expression (Swaroop et al. 1997. JBC 272:24121-24124), Factor VIII mutants reducing its immunogenicity (Lollar 1999. Thromb. Haemost. 82:505-508), FVIII reconstituted from differently expressed heavy and light chains (Oh et al. 1999. Exp. Mol. Med. 31:95-100), FVIII mutants reducing binding to receptors leading to catabolism of FVIII like HSPG (heparan sulfate proteoglycans) and/or LRP (low density lipoprotein receptor related protein) (Ananyeva et al. 2001. TCM, 11:251-257), disulfide bond-stabilized FVIII variants (Gale et al., 2006. J. Thromb. Hemost. 4:1315-1322), FVIII mutants with improved secretion properties (Miao et al., 2004. Blood 103:3412-3419), FVIII mutants with increased cofactor specific activity (Wakabayashi et al., 2005. Biochemistry 44:10298-304), FVIII mutants with improved biosynthesis and secretion, reduced ER chaperone interaction, improved ER-Golgi transport, increased activation or resistance to inactivation and improved half-life (summarized by Pipe 2004. Sem. Thromb. Hemost. 30:227-237). All of these factor VIII mutants and variants are incorporated herein by reference in their entirety.

[0094] A suitable test to determine the biological activity of Factor VIII is the one stage or the two stage coagulation assay (Rizza et al. 1982. Coagulation assay of FVIII:C and FIXa in Bloom ed. The Hemophilias. NY Churchchill Livingston 1992) or the chromogenic substrate FVIII:C assay (S. Rosen, 1984. Scand J Haematol 33: 139-145, suppl.). The content of these references is incorporated herein by reference.

[0095] The cDNA sequence and the amino acid sequence of the mature wild type form of human blood coagulation Factor VIII are shown in SEQ ID NO:1 and SEQ ID NO:2, respectively. The reference to an amino acid position of a specific sequence means the position of said amino acid in the FVIII wild-type protein and does not exclude the presence of mutations, e.g. deletions, insertions and/or substitutions at other positions in the sequence referred to. For example, a mutation in "Glu2004" referring to SEQ ID NO:2 does not exclude that in the modified homologue one or more amino acids at positions 1 through 2332 of SEQ ID NO:2 are missing.

[0096] FVIII Proteins with a HLEP Insertion

[0097] Modified FVIII proteins of the invention in the most general sense are characterized in that they comprise FVIII molecules with a HLEP integrated into the FVIII molecules such that the HLEP does not reduce the molar specific FVIII activity of the chimeric protein below about 10% of the molar specific FVIII activity of wild type FVIII. The insertion of the HLEP can take place in any place between the N-terminal and the C-terminal amino acid of the FVIII sequence. Preferentially the HLEP is integrated between domains of the wild-type FVIII protein.

[0098] The domains of FVIII comprise the following amino acid positions (amino acid numbers refer to SEQ ID NO:2):

[0099] A1: . . . 1-336

[0100] a1: . . . 337-372

[0101] A2: . . . 373-710

[0102] a2: . . . 711-740

[0103] B: . . . 741-1648

[0104] a3: . . . 1649-1689

[0105] A3: . . . 1690-2019

[0106] C1: . . . 2020-2172

[0107] C2: . . . 2173-2332

[0108] Preferred integration sites for a HLEP within the FVIII molecule are defined as such sites where the insertion of a HLEP moiety has the least negative effect on FVIII functional activity. Potential integration sites include, but are not limited to, the region between the C-terminus of acidic region 1 (a1) and the N-terminus of the A2 domain, the region between the C-terminus of the A3 domain and the N-terminus of the C1 domain, the region between the C-terminus of the C1 domain and the N-terminus of the C2 domain and preferably the region of the B domain, where the B domain may be replaced partially or in its entirety (FIG. 2).

[0109] In a preferred embodiment of the invention chimeric FVIII proteins of the invention are characterized in that they comprise FVIII molecules with partial or full deletion of the B domain and a HLEP integrated into the FVIII molecules such that the HLEP is inserted between a functional A1/A2 domain at its amino terminus and a functional A3/C1/C2 domain at its carboxy terminus.

[0110] It was found that it is possible to insert HLEPs or HLEP derivatives within the B domain (the FVIII sequence between the A2 and A3 domains [amino acids 741 to 1648] which seems dispensable for the biological function of FVIII (Pittman et al. 1992. Blood 81:2925-2935) to provide FVIII molecules with new and improved properties while retaining FVIII biological activity. The B domain has a length of about 900 amino acids and the HLEP may either be inserted at any place within the B domain without any deletion of the B domain or the B domain may be replaced by a HLEP partially or in its entirety. Partial deletion refers to deletions of at least 1 amino acid, preferably to deletions of 100 to 600 amino acids and most preferred to deletions of more than 600 amino acids of the B domain (FIG. 2e-h).

[0111] In a preferred embodiment of the invention most of the B domain is replaced by a HLEP, while a few amino acids of the amino and carboxy terminal sequence of the B domain containing processing sites important for cleavage and activation of the FVIII molecules of the invention are conserved (FIGS. 1a,b,d and 2h-i). Preferably about 1 to 20 amino acids, more preferably 3 to 10 amino acids, at the C- and at the N-terminus of the B domain, which are required to conserve the processing sites for thrombin at amino acid position 740 of the FVIII sequence (SEQ ID NO 2) and the protease cleaving between the B domain and the A3 domain during the secretion process, are maintained within the FVIII molecule of the invention (FIG. 1 and FIG. 2h). Alternatively, the amino acids retained from the B domain might be replaced by artificial cleavage sites. A PACE/Furin cleavage site (Nakayama 1997. Biochem. J. 327:625-635) may be used to guide the processing during secretion, and artificial thrombin cleavage sites as described in WO 2004/005347 (FIG. 1c) or other protease cleavage sites may be introduced for activation processing (FIG. 1e).

[0112] Another aspect of the invention is the insertion of more than one HLEP wherein the HLEP, which is inserted several times, may be the same HLEP or may be a combination of different HLEPs. Also combinations of insertions of one or more HLEPs into FVIII with additional N- and/or C-terminal fusions of one or more HLEPs, which could be the same HLEP or a combination of different HLEPs are encompassed by the invention.

[0113] Once a coagulation factor is endogenously activated during coagulation in vivo, it may be no longer desirable to maintain the increased functional half-life of the now activated coagulation factor as this might lead to thrombotic complications what is already the case for a wild type activated coagulation factor as FVIIa (Aledort 2004. J Thromb Haemost 2:1700-1708) and what should be much more relevant if the activated factor would have an increased functional half-life. It is therefore another objective of the present invention to provide long-lived coagulation factor VIII molecules, which after endogenous activation in vivo or after availability of a cofactor do have a functional half-life comparable to that of unmodified FVIII. This can by way of non-limiting example be achieved by maintaining the cleavage sites for thrombin at amino acid position 740 of the FVIII sequence (SEQ ID NO 2) and for the protease cleaving between the B domain and the A3 domain during the secretion process. With such FVIII-HLEP connecting sequences the activation of the FVIII chimeric protein of the invention will lead to a concomitant complete separation of FVIIIa from the HLEP moiety.

[0114] In yet another embodiment of the invention, however, one or more of the proteolytical cleavage sites, preferably the thrombin cleavage sites at Arg740 (e.g. FIG. 2i) and/or Arg372, are mutated or deleted in order to prevent cleavage and result in an insertion protein which displays improved properties like enhanced functional half-life even as an activated molecule.

[0115] In another embodiment of the invention the deletion of the B domain may be extended into the flanking acidic regions a2 and a3 (FIG. 2 k and l). With regard to a2 this region may be deleted in part (FIG. 2k) or completely. Therefore the HLEP moiety will not be released upon FVIII activation but instead remain attached to the A2 domain. Such an activated insertion protein will have an enhanced functional half-life. Acidic region a3 may be deleted in part (FIG. 2l) as long as the vWF binding properties of a3 remain unaffected.

[0116] In one embodiment of the invention another potential integration site within the FVIII molecule is represented by the region between the C-terminus of acidic region 1 (a1)) and the N-terminus of the A2 domain (FIG. 2a-d). FIG. 2a describes an integration scheme where an additional thrombin cleavage site has been introduced at the albumin C-terminus. In such an insertion protein the HLEP moiety will be cleaved off during endogenous FVIII activation in vivo and the activated FVIII molecule will have a functional half-life comparable to wild-type FVIII. In the case of an insertion protein as depicted in FIG. 2b the additional thrombin cleavage site at the HLEP C-terminus is lacking. Therefore the HLEP will not be released upon FVIII activation but instead remain attached to the A2 domain. Such an activated insertion protein will have an enhanced functional half-life. In the case of an insertion protein as depicted in FIG. 2c the thrombin cleavage site at Arg372 is lacking. Therefore the HLEP will not be released upon FVIII activation but instead remain attached to the A1 domain. Such an activated insertion protein will have an enhanced half-life. An insertion protein as depicted in FIG. 2d will keep A1 and A2 domains covalently linked and generate an insertion protein with functional half-life extension also of the activated form.

[0117] In another embodiment of the invention another potential integration site within the FVIII molecule is represented by the region between the C-terminus of the A3 domain and the N-terminus of the C1 domain (FIG. 2m). In such an insertion protein the HLEP moiety will be an integral component of the FVIII light chain and both the non-activated and the activated insertion protein will have enhanced functional half-lives.

[0118] In another embodiment of the invention another potential integration site within the FVIII molecule is represented by the region between the C-terminus of the C1 domain and the N-terminus of the C2 domain (FIG. 2n). In such an insertion protein the HLEP moiety will be an integral component of the FVIII light chain and both the non-activated and the activated insertion protein will have enhanced functional half-lives.

[0119] In another embodiment of the invention the FVIII proteins of the invention may be expressed as two separate chains (see infra).

[0120] The modified coagulation factor VIII according to this invention may be a single chain polypeptide, or it may be composed of two or three polypeptide chains that are associated via non-covalent linkages, due to proteolytic processing.

[0121] In another embodiment of the invention, the amino acids at or near the PACE/Furin cleavage site (Arg1648, e.g. FIG. 1a) are mutated or deleted in order to prevent cleavage by PACE/Furin. This is thought to result in a one-chain Factor VIII/HLEP fusion molecule with improved half-life.

[0122] In one embodiment of the invention, the modified FVIII of the invention exhibits an increased functional half-life compared to the corresponding FVIII form containing no integrated HLEP and/or to the wild type form FVIII. The functional half-life e.g. can be determined in vivo in animal models of hemophilia A, like FVIII knockout mice, in which one would expect a longer lasting hemostatic effect as compared to wild type FVIII. The hemostatic effect could be tested for example by determining time to arrest of bleeding after a tail clip.

[0123] The functional half-life is preferably increased by at least 25%, more preferably by at least 50%, and even more preferably by at least 100% compared to the form without inclusion of a HLEP and/or to the wild type form of FVIII.

[0124] In another embodiment of the invention, the modified FVIII of the invention exhibits an improved in vivo recovery compared to the corresponding FVIII form containing no integrated HLEP and/or to the wild type form FVIII. The in vivo recovery can be determined in vivo in normal animals or in animal models of hemophilia A, like FVIII knockout mice, in which one would expect an increased percentage of the modified FVIII of the invention be found by antigen or activity assays in the circulation shortly (5 to 10 min.) after i.v. administration compared to the corresponding FVIII form containing no integrated HLEP and/or to the wild type form FVIII.

[0125] The in vivo recovery is preferably increased by at least 10%, more preferably by at least 20%, and even more preferably by at least 40% compared to the form without inclusion of a HLEP and/or to the wild type form of FVIII.

[0126] In yet another embodiment of the invention immunoglobulin constant regions or portions thereof are used as HLEPs. Preferably the Fc region comprised of a CH2 and CH3 domain and a hinge region of an IgG, more preferably of an IgG1 or fragments or variants thereof are used, variants including mutations which enhance binding to the neonatal Fc receptor (FcRn). The Fc region is not used to generate monomeric or dimeric Fc insertions as described in the art, but rather is inserted into the FVIII molecule such that part of the FVIII molecule is fused to its N-terminus and another part is fused to its C-terminus (FIG. 2a-n). In a preferred embodiment of the invention an unfused Fc region is coexpressed from another expression vector or even from the same expression vector which through disulfide bridge linking forms a Fc heterodimer with the Fc region within the chimeric FVIII molecule.

[0127] In addition to the extension of functional half-life of FVIII, HLEP moieties as described in this invention may also be used for insertion into other multi-domain proteins for the same purpose of half-life extension.

[0128] Therefore the invention also encompasses other modified proteins, preferably modified coagulation factors, with insertions of HLEP moieties within their amino acid sequence.

[0129] Von Willebrand Factor

[0130] Von Willebrand factor (vWF) is a multimeric plasma glycoprotein with a prominent role in primary hemostasis. The mature protein consists of 2050 amino acids and is composed of homologous domains arranged in the order D'-D3-A1-A2-A3-D4-B1-B2-B3-C1-C2-CK. The amino acid sequence and the cDNA sequence of wild type vWF are disclosed in Collins et al. 1987. Proc Natl. Acad. Sci. USA 84:4393-4397. The term "von Willebrand factor" includes any mutants and variants of wild type vWF having at least 10%, preferably at least 25%, more preferably at least 50%, most preferably at least 75% of the biological activity of wild type vWF. The biological activity of wild type vWF can be determined by the man of the art using methods for ristocetin co-factor activity (Federici A B et al. 2004. Haematologica 89:77-85), binding of vWF to GP lba of the platelet glycoprotein complex Ib-V-IX (Sucker et al. 2006. Clin Appl Thromb Hemost. 12:305-310), or a collagen binding assay (Kailas & Talpsep. 2001. Annals of Hematology 80:466-471).

[0131] One or more HLEPs may be inserted into the vWF molecule. HLEP insertion is chosen as not to interfere with the binding capabilities of vWF to e.g. FVIII, platelets, Heparin or collagen. Suitable insertion sites include, but are not limited to, the D3-A1 junction, the D4-B1 junction, the C2-CK junction as well as A2, into which a HLEP moiety may be inserted upon partial or complete removal of the A2 domain. VWF functional activities may be assessed as described supra.

[0132] Prothrombin Factors

[0133] Prothrombin factors, including factor VII (FVII), factor IX (FIX), factor X (FX), protein C (PC), protein S, protein Z and prothrombin (PT) are a family of proteins characterized by a gla domain containing y-carboxylated glutamic acid residues and EGF- or Kringle domains on the light chain, which is separated from the heavy chain containing the trypsin protease domain (two laminin-G domains for protein S) by a short intervening sequence which is cleaved upon activation of the protein.

[0134] The amino acid sequences and the cDNA sequences of these coagulation factors are known in the art and are disclosed for example in the PubMed protein sequence library (http://www.ncbi.nlm.nih.gov/entrez/querv.fcgi?db=Protein) with accession numbers NP.sub.--000122 (FVII), NP.sub.--000124 (FIX), NP.sub.--000495 (FX), NP.sub.--000303 (PC), NP.sub.--000304 (Protein S), NP.sub.--003882 (Protein Z) and NP.sub.--000497 (Prothrombin).

[0135] Also prothrombin factors may be stabilized by the insertion of a HLEP moiety as described in this invention. Prothrombin factors include factor VII (FVII), factor IX (FIX), factor X (FX), protein C(PC), protein S, protein Z and prothrombin (PT). As described supra, prothrom bin factors are characterized by a gla domain containing y-carboxylated glutamic acid residues and EGF- or Kringle domains on the light chain, which is separated from the heavy chain containing the trypsin protease domain (two laminin-G domains for protein S) by a short intervening sequence which is cleaved upon activation of the protein. This peptide sequence is the preferred integration site for a HLEP moiety. Preferably, the HLEP is inserted such that the activation cleavage is not hampered by maintaining the natural activation sequence or by inserting artificial cleavage sites like a PACE/Furin cleavage site (Nakayama 1997. Biochem. J. 327:625-635), an artificial thrombin cleavage site (as described in WO 2004/005347) or another suitable protease cleavage site. The conservation of the activity of the respective prothrombin factor after HLEP insertion may be assessed by assays known to the man of the art. FVII activity may be determined using a commercially available chromogenic test kit (Chromogenix Coaset FVII) based on the method described by Seligsohn et al. (1978. Blood 52:978-988) and FVIIa activity can be determined using the STACLOT.RTM. FVIIa-rTF kit (Diagnostica Stago) based on the method described by Morissey et al. (1993. Blood 81:734-744). FIX activity may be assessed by a clotting assay as described by Chavin & Weidner (1984. J. Biol. Chem. 259:3387-3390). FX activity may be measured using a chromogenic assay as described by Van Wijk et al. (1981. Thromb. Res. 22:681-686). Protein C activity may be assessed by a chromogenic assay as supplied by Instrumentation Laboratory (HaemoslL Protein C) based on the method described by Comb et al. (1984. Blood 63:15-21) and protein S activity by a method described by Heeb et al. (2006. J. Thromb. Haemost. 4:385-391). Petrovan et al. (1999. Am. J. Clin. Pathol. 112:705-711 describe an activity assay for prothrombin and Tabatabai et al. (2001. Thromb. Haemost. 85:655-660) published a protein Z activity assay.

[0136] Coagulation factor V

[0137] Coagulation factor V (FV) is a high molecular weight plasma glycoprotein that participates as a cofactor in the activation of Prothrombin by factor Xa. It is homologous to factor VIII and Ceruloplasmin and has a similar domain structure of A1-A2-B-A3-C1-C2. The amino acid sequence and the cDNA sequence of wild type FV are disclosed for example in PubMed with accession numbers NP.sub.--000121 and NM.sub.--000130, respectively.

[0138] As described above for Factor VIII, HLEP moieties could be inserted into the FV molecule for half-life extension at comparable inter-domain sites, preferably into the B domain or replacing part or all of the B domain. The FV activity can be assessed as described by Bick et al. (1973. Beitr. Pathol. 150:311-315).

[0139] Polynucleotides

[0140] The invention further relates to a polynucleotide encoding a modified coagulation factor, preferably a modified FVIII variant as described in this application. The term "polynucleotide(s)" generally refers to any polyribonucleotide or polydeoxyribonucleotide that may be unmodified RNA or DNA or modified RNA or DNA. The polynucleotide may be single- or double-stranded DNA, single or double-stranded RNA. As used herein, the term "polynucleotide(s)" also includes DNAs or RNAs that comprise one or more modified bases and/or unusual bases, such as inosine. It will be appreciated that a variety of modifications may be made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term "polynucleotide(s)" as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including, for example, simple and complex cells.

[0141] The skilled person will understand that, due to the degeneracy of the genetic code, a given polypeptide can be encoded by different polynucleotides. These "variants" are encompassed by this invention.

[0142] Preferably, the polynucleotide of the invention is an isolated polynucleotide. The term "isolated" polynucleotide refers to a polynucleotide that is substantially free from other nucleic acid sequences, such as and not limited to other chromosomal and extrachromosomal DNA and RNA. Isolated polynucleotides may be purified from a host cell. Conventional nucleic acid purification methods known to skilled artisans may be used to obtain isolated polynucleotides. The term also includes recombinant polynucleotides and chemically synthesized polynucleotides.

[0143] The invention further relates to a group of polynucleotides which together encode the modified coagulation factor of the invention. A first polynucleotide in the group may encode the N-terminal part of the modified coagulation factor, and a second polynucleotide may encode the C-terminal part of the modified coagulation factor.

[0144] Yet another aspect of the invention is a plasmid or vector comprising a polynucleotide according to the invention. Preferably, the plasmid or vector is an expression vector. In a particular embodiment, the vector is a transfer vector for use in human gene therapy.

[0145] The invention also relates to a group of plasmids or vectors that comprise the above group of polynucleotides. A first plasmid or vector may contain said first polynucleotide, and a second plasmid or vector may contain said second polynucleotide. By way of example, and with reference to coagulation factor VIII, the coding sequences of the signal peptide, the A1 and A2 domains, the B domain sequence remainder and the HLEP may be cloned into the first expression vector and the coding sequences of A3, C1 and C2 with an appropriate signal peptide sequence may be cloned into the second expression vector (FIG. 20). Both expression vectors are cotransfected into a suitable host cell, which will lead to the expression of the light and heavy chains of the FVIII molecule of the invention and the formation of a functional protein.

[0146] Alternatively, the coding sequence of the FVIII signal peptide, the A1 and A2 domains are cloned into the first expression vector and the coding sequences of the HLEP, FVIII A3, C1 and C2 with an appropriate signal peptide sequence are cloned into the second expression vector (FIG. 2p). Both expression vectors are cotransfected into a suitable host cell, which will lead to the expression of the light and heavy chains of the FVIII molecule of the invention and the formation of a functional protein.

[0147] Alternatively, both coding sequences are cloned into one expression vector either using two separate promoter sequences or one promoter and an internal ribosome entry site (IRES) element to direct the expression of both FVIII chains.

[0148] Still another aspect of the invention is a host cell comprising a polynucleotide, a plasmid or vector of the invention, or a group of polynucleotides or a group of plasmids or vectors as described herein.

[0149] The host cells of the invention may be employed in a method of producing a modified coagulation factor, preferably a modified FVIII molecule, which is part of this invention. The method comprises: [0150] (a) culturing host cells of the invention under conditions such that the desired insertion protein is expressed; and [0151] (b) optionally recovering the desired insertion protein from the host cells or from the culture medium.

[0152] It is preferred to purify the modified coagulation factors of the present invention to .gtoreq.80% purity, more preferably .gtoreq.95% purity, and particularly preferred is a pharmaceutically pure state that is greater than 99.9% pure with respect to contaminating macromolecules, particularly other proteins and nucleic acids, and free of infectious and pyrogenic agents. Preferably, an isolated or purified modified coagulation factor of the invention is substantially free of other, non-related polypeptides.

[0153] The various products of the invention are useful as medicaments. Accordingly, the invention relates to a pharmaceutical composition comprising a modified coagulation factor, preferably the modified FVIII molecule as described herein, a polynucleotide of the invention, or a plasmid or vector of the invention.

[0154] The invention also concerns a method of treating an individual suffering from a blood coagulation disorder such as hemophilia A or B. The method comprises administering to said individual an efficient amount of the modified coagulation factor, preferably modified FVIII or FIX as described herein. In another embodiment, the method comprises administering to the individual an efficient amount of a polynucleotide of the invention or of a plasmid or vector of the invention. Alternatively, the method may comprise administering to the individual an efficient amount of the host cells of the invention described herein.

[0155] The invention also relates to polynucleotides and their use encoding the modified VWF and Prothrombin factor variants as described above.

[0156] Expression of the Proposed Mutants

[0157] The production of recombinant mutant proteins at high levels in suitable host cells requires the assembly of the above-mentioned modified cDNAs into efficient transcriptional units together with suitable regulatory elements in a recombinant expression vector that can be propagated in various expression systems according to methods known to those skilled in the art. Efficient transcriptional regulatory elements could be derived from viruses having animal cells as their natural hosts or from the chromosomal DNA of animal cells. Preferably, promoter-enhancer combinations derived from the Simian Virus 40, adenovirus, BK polyoma virus, human cytomegalovirus, or the long terminal repeat of Rous sarcoma virus, or promoter-enhancer combinations including strongly constitutively transcribed genes in animal cells like beta-actin or GRP78 can be used. In order to achieve stable high levels of mRNA transcribed from the cDNAs, the transcriptional unit should contain in its 3'-proximal part a DNA region encoding a transcriptional termination-polyadenylation sequence. Preferably, this sequence is derived from the Simian Virus 40 early transcriptional region, the rabbit beta-globin gene, or the human tissue plasminogen activator gene.

[0158] The cDNAs are then integrated into the genome of a suitable host cell line for expression of the Factor VIII proteins. Preferably this cell line should be an animal cell-line of vertebrate origin in order to ensure correct folding, disulfide bond formation, asparagine-linked glycosylation and other post-translational modifications as well as secretion into the cultivation medium. Examples on other post-translational modifications are tyrosine O-sulfation and proteolytic processing of the nascent polypeptide chain. Examples of cell lines that can be use are monkey COS-cells, mouse L-cells, mouse C127-cells, hamster BHK-21 cells, human embryonic kidney 293 cells, and hamster CHO-cells.

[0159] The recombinant expression vector encoding the corresponding cDNAs can be introduced into an animal cell line in several different ways. For instance, recombinant expression vectors can be created from vectors based on different animal viruses. Examples of these are vectors based on baculovirus, vaccinia virus, adenovirus, and preferably bovine papilloma virus.

[0160] The transcription units encoding the corresponding DNA's can also be introduced into animal cells together with another recombinant gene which may function as a dominant selectable marker in these cells in order to facilitate the isolation of specific cell clones which have integrated the recombinant DNA into their genome. Examples of this type of dominant selectable marker genes are Tn5 amino glycoside phosphotransferase, conferring resistance to geneticin (G418), hygromycin phosphotransferase, conferring resistance to hygromycin, and puromycin acetyl transferase, conferring resistance to puromycin. The recombinant expression vector encoding such a selectable marker can reside either on the same vector as the one encoding the cDNA of the desired protein, or it can be encoded on a separate vector which is simultaneously introduced and integrated to the genome of the host cell, frequently resulting in a tight physical linkage between the different transcription units.

[0161] Other types of selectable marker genes which can be used together with the cDNA of the desired protein are based on various transcription units encoding dihydrofolate reductase (dhfr). After introduction of this type of gene into cells lacking endogenous dhfr-activity, preferentially CHO-cells (DUKX-B11, DG-44), it will enable these to grow in media lacking nucleosides. An example of such a medium is Ham's F12 without hypoxanthine, thymidin, and glycine. These dhfr-genes can be introduced together with the Factor VIII cDNA transcriptional units into CHO-cells of the above type, either linked on the same vector or on different vectors, thus creating dhfr-positive cell lines producing recombinant protein.

[0162] If the above cell lines are grown in the presence of the cytotoxic dhfr-inhibitor methotrexate, new cell lines resistant to methotrexate will emerge. These cell lines may produce recombinant protein at an increased rate due to the amplified number of linked dhfr and the desired protein's transcriptional units. When propagating these cell lines in increasing concentrations of methotrexate (1-10000 nM), new cell lines can be obtained which produce the desired protein at very high rate.

[0163] The above cell lines producing the desired protein can be grown on a large scale, either in suspension culture or on various solid supports. Examples of these supports are micro carriers based on dextran or collagen matrices, or solid supports in the form of hollow fibres or various ceramic materials. When grown in cell suspension culture or on micro carriers the culture of the above cell lines can be performed either as a bath culture or as a perfusion culture with continuous production of conditioned medium over extended periods of time. Thus, according to the present invention, the above cell lines are well suited for the development of an industrial process for the production of the desired recombinant mutant proteins

[0164] Purification and Formulation

[0165] The recombinant mutant protein, which accumulates in the medium of secreting cells of the above types, can be concentrated and purified by a variety of biochemical and chromatographic methods, including methods utilizing differences in size, charge, hydrophobicity, solubility, specific affinity, etc. between the desired protein and other substances in the cell cultivation medium.

[0166] An example of such purification is the adsorption of the recombinant mutant protein to a monoclonal antibody, directed to e.g. a HLEP, preferably human albumin, or directed to the respective coagulation factor, which is immobilised on a solid support. After adsorption of the FVIII mutant to the support, washing and desorption, the protein can be further purified by a variety of chromatographic techniques based on the above properties. The order of the purification steps is chosen e.g. according to capacity and selectivity of the steps, stability of the support or other aspects. Preferred purification steps e.g. are but are not limited to ion exchange chromatography steps, immune affinity chromatography steps, affinity chromatography steps, hydrophobic interaction chromatography steps, dye chromatography steps, and size exclusion chromatography steps.

[0167] In order to minimize the theoretical risk of virus contaminations, additional steps may be included in the process that allow effective inactivation or elimination of viruses. Such steps e.g. are heat treatment in the liquid or solid state, treatment with solvents and/or detergents, radiation in the visible or UV spectrum, gamma-radiation or nanofiltration.

[0168] The modified polynucleotides (e.g. DNA) of this invention may also be integrated into a transfer vector for use in the human gene therapy.

[0169] The various embodiments described herein may be combined with each other. The present invention will be further described in more detail in the following examples thereof. This description of specific embodiments of the invention will be made in conjunction with the appended figures.

[0170] The insertion proteins as described in this invention can be formulated into pharmaceutical preparations for therapeutic use. The purified protein may be dissolved in conventional physiologically compatible aqueous buffer solutions to which there may be added, optionally, pharmaceutical excipients to provide pharmaceutical preparations.

[0171] Such pharmaceutical carriers and excipients as well as suitable pharmaceutical formulations are well known in the art (see for example "Pharmaceutical Formulation Development of Peptides and Proteins", Frokjaer et al., Taylor & Francis (2000) or "Handbook of Pharmaceutical Excipients", 3.sup.rd edition, Kibbe et al., Pharmaceutical Press (2000)). In particular, the pharmaceutical composition comprising the polypeptide variant of the invention may be formulated in lyophilized or stable liquid form. The polypeptide variant may be lyophilized by a variety of procedures known in the art. Lyophilized formulations are reconstituted prior to use by the addition of one or more pharmaceutically acceptable diluents such as sterile water for injection or sterile physiological saline solution.

[0172] Formulations of the composition are delivered to the individual by any pharmaceutically suitable means of administration. Various delivery systems are known and can be used to administer the composition by any convenient route. Preferentially, the compositions of the invention are administered systemically. For systemic use, insertion proteins of the invention are formulated for parenteral (e.g. intravenous, subcutaneous, intramuscular, intraperitoneal, intracerebral, intrapulmonar, intranasal or transdermal) or enteral (e.g., oral, vaginal or rectal) delivery according to conventional methods. The most preferential routes of administration are intravenous and subcutaneous administration. The formulations can be administered continuously by infusion or by bolus injection. Some formulations encompass slow release systems.

[0173] The insertion proteins of the present invention are administered to patients in a therapeutically effective dose, meaning a dose that is sufficient to produce the desired effects, preventing or lessening the severity or spread of the condition or indication being treated without reaching a dose which produces intolerable adverse side effects. The exact dose depends on many factors as e.g. the indication, formulation, mode of administration and has to be determined in preclinical and clinical trials for each respective indication.

[0174] The pharmaceutical composition of the invention may be administered alone or in conjunction with other therapeutic agents. These agents may be incorporated as part of the same pharmaceutical. One example of such an agent is von Willebrand factor.

[0175] FIG. 1 shows the replacement of FVIII B domain by albumin. cDNA organisation of FVIII wild-type (FVIII wt) and FVIII with the B domain replacement by albumin (FVIII-HA) are outlined. Transition sequences and the remaining amino acids of the B domain in the FVIII-HA constructs are shown. Amino acid numbering refers to the FVIII wild-type sequence as outlined in SEQ ID NO:2. The C1636S amino acid exchange in DNA pF8-1211 and the R740 deletion in pF8-1413 are indicated.

[0176] FIG. 2 schematically shows various embodiments of the cDNA encoding the modified Factor VIII polypeptides of the present invention. The HLEP may be inserted at various positions within the FVIII sequence, as described supra.

[0177] FIG. 3 shows the pharmacokinetic profile of two modified FVIII molecules with albumin integrated and partial deletion of the B-domain (DNA pF8-1211 and pF8-1413, see FIG. 1) in comparison to wild type FVIII (see example 5).

EXAMPLES

Example 1

Generation of Expression Vectors for FVIII Molecules with Albumin Replacing the FVIII B Domain

[0178] An expression plasmid based on pIRESpuro3 (BD Biosciences) containing the full length FVIII cDNA sequence in its multiple cloning site (pF8-FL) was first used to delete the majority of the B domain sequence and create a restriction site for insertion of foreign sequences. For that oligonucleotides We1356 and We1357 (SEQ ID NO. 5 and 6) were used in a PCR reaction using pF8-FL as a template to amplify a part of the A2 domain and the N-terminus of the B domain (fragment 1) and oligonucleotides We1358 and We1359 (SEQ ID NO. 7 and 8) were used in another PCR reaction using pF8-FL as a template to amplify the C-terminus of the B domain, the A3 domain and part of the C1 domain (fragment 2). Both fragments were gel purified. Fragment 1 was subsequently digested with restricion endonucleases PinAl and BamH1, fragment 2 was digested with restriction endonucleases PinAl and BspEl; both fragments were then purified and ligated into pF8-FL, where the BamH1/BspEl fragment encompassing part of the A2 domain, the B and A3 domains and part of the C1 domain had been removed. The resulting plasmid, pF8-DB, now basically contained a major B domain deletion with a remainder of N- and C-terminal B domain sequences joined by a PinAl site. Into this site a human albumin fragment was inserted, which had been generated by PCR amplification on albumin cDNA using primers We2502 and We2503 (SEQ ID NO. 9 and 10), PinAl digestion and purification. To remove the PinAl sites the resulting plasmid was subjected to two rounds of site-directed mutagenesis according to standard protocols (QuickChange XL Site Directed Mutagenesis Kit, Stratagene). For this oligonucleotides We2504 and We2505 (SEQ ID NO. 11 and 12) were used as mutagenic primers in the first round, and oligonucleotides We2506 and We2507 (SEQ ID NO. 13 and 14) were used in the second round of mutagenesis. The final expression plasmid was designated pF8-1210. In order to remove a free cysteine residue (amino acid 1636, SEQ ID NO. 2 and FIG. 1) site-directed mutagenesis was applied using oligonucleotides We2508 and We2509 (SEQ ID NO. 15 and 16) giving rise to plasmid pF8-1211.

[0179] Site directed mutagenesis was applied according to standard protocols (QuickChange XL Site Directed Mutagenesis Kit, Stratagene) to delete the arginine in position 740 in plasmid pF8-1211. For this oligonucleotides We2768 and We2769 (SEQ ID NO. 17 and 18) were used as mutagenic primers. The resulting expression plasmid was designated pF8-1413. A FVIII molecule where the B domain had been replaced by amino acid sequence RRGR was used as the wild-type FVIII control, the encoding plasmid was called pF8-457.

[0180] Using the protocols and plasmids described above and by applying molecular biology techniques known to those skilled in the art (and as described e.g. in Current Protocols in Molecular Biology, Ausubel F M et al. (eds.) including supplement 80, October 2007, John Wiley & Sons, Inc.; http://www.currentprotocols.com/WileyCDA/) other constructs can be made by the artisan with insertions of a HLEP molecule in positions described in FIG. 2 and linker sequences as shown exemplarily in FIGS. 1b-e.

Example 2

Generation of Expression Vectors for FVIII Molecules with an Immunoglobulin Constant Region Replacing the FVIII B Domain

[0181] The insertion of an IgG Fc domain into the FVIII molecule replacing the majority of the B domain was performed in analogy to the protocols and reference described above. The resulting plasmid was called pF8-1518 and the mature protein translated from this is shown in SEQ ID NO. 19.

[0182] As recycling of IgG by the neonatal Fc receptor only works with the Fc being dimeric pF8-1518 was cotransfected into HEK-293 cells with a plasmid encoding a human immunoglobulin G heavy chain region (p1335, SEQ ID No. 20). The coexpression of plasmids pF8-1518 and p1335 led to the expression of a functional FVIII molecule (table 1).

[0183] In another set of constructs FVIII heavy and light chains were expressed separately. For that pF8-1518 was mutated in that a stop codon was introduced at the very 3''-end of the IgG heavy chain sequence. The expression of such construct (pF8-1515) led to a FVIII heavy chain (A1 and A2 domain) with a few amino acids of the B domain followed by the IgG heavy chain (SEQ ID NO. 21). The FVIII light chain construct was also based on plasmid pF8-1518 in that the A1 and A2 domain coding sequences were replaced by a signal peptide. The expression of such construct (pF8-1517) led to a FVIII light chain with an IgG heavy chain attached to its N-terminus (SEQ ID NO. 22). The coexpression of plasmids pF8-1515 and pF8-1517 led to the expression of a functional FVIII molecule (table 1).

Example 3

Transfection and Expression of FVIII Mutants

[0184] Expression plasmids were grown up. in E. coli TOP10 (Invitrogen) and purified using standard protocols (Qiagen). HEK-293 cells were transfected using the Lipofectamine 2000 reagent (Invitrogen) and grown up in serum-free medium (Invitrogen 293 Express) in the presence of 4 .mu.g/ml Puromycin and optionally 0.5 IU/ml vWF. Transfected cell populations were spread through T-flasks into roller bottles or small scale fermenters from which supernatants were harvested for purification.

[0185] Table 1 lists expression data of a number of constructs outlined in FIGS. 1 and 2 and described in examples 1 and 2. Unless otherwise indicated, the HLEP used is albumin.

TABLE-US-00001 TABLE 1 Activity Antigen Ratio activity/ Construct [U/mL] [U/mL] antigen FIG. 2c 1.0 7.3 0.14 FIG. 2d 0.4 4.7 0.09 FIG. 2f 0.44 1.09 0.40 FIG. 2h 1.04 0.94 1.11 FIG. 2i 0.33 0.47 0.70 FIG. 2i (HLEP = 0.31 1.01 0.31 Afamin) FIG. 2i (HLEP = 0.53 1.16 0.46 Alpha-fetoprotein) FIG. 2o 0.22 0.75 0.30 pF8-1518 + p1335 1.19 1.78 0.67 (HLEP = Fc) pF8-1515 + pF8-1517 1.75 6.68 0.26 (HLEP = Fc)

Example 4

Purification of Factor VIII Mutants

[0186] To the expression supernatant containing the chimeric Factor VIII molecule a sufficient amount of an immune affinity resin was added to bind the FVIII activity almost completely. The immune affinity resin had been prepared by binding an appropriate anti-FVIII MAb covalently to Sephacryl S1000 resin used as a support. After washing of the resin it was filled into a chromatography column and washed again. Elution was done using a buffer containing 250 mM CaCl2 and 50% ethylene glycol.

[0187] The immune affinity chromatography (IAC) fractions containing FVIII:C activity were pooled, dialyzed against formulation buffer (excipients: sodium chloride, sucrose, histidine, calcium chloride, and Tween 80), and concentrated. Samples are either stored frozen or are freeze-dried using an appropriate freeze-drying cycle. Table 2 shows the results of a purification run using a FVIII mutant (pF8-1211 from HEK-293) and IAC as main purification step.

TABLE-US-00002 TABLE 2 Volume FVIII:C FVIII:Ag Total protein* Specific activity FVIII:C/FVIII:Ag Sample (mL) (IU/mL) (IU/mL) (mg/mL) (IU/mg) (IU/IU) Supernatant 890 3.3 1.92 1.72 1.9 1.72 IAC Eluate 26 52.2 30.6 0.036 1450 1.71 *determined by measurement of Optical density (OD) at 280 nm (OD.sub.280, 1% = 10.0)

[0188] Alternatively, the FVIII containing cell culture supernatant is concentrated/purified by a first ion exchange chromatography followed by further purification using immune affinity chromatography (IAC). In this case the eluate of the ion exchange chromatography is loaded onto an IAC column using the above mentioned resin.

Example 5

Analysis of Chimeric Factor VIII Activity and Antigen

[0189] For activity determination of FVIII:C in vitro either a clotting assay (e.g. Pathromtin SL reagent and FVIII deficient plasma delivered by Dade Behring, Germany) or a chromogenic assay (e.g. Coamatic FVIII:C assay delivered by Haemochrom) were used. The assays were performed according to the manufacturers instructions.

[0190] FVIII antigen (FVIII:Ag) was determined by an ELISA whose performance is known to those skilled in the art. Briefly, microplates were incubated with 100 .mu.L per well of the capture antibody (sheep anti-human FVIII IgG, Cedarlane CL20035K-C, diluted 1:200 in Buffer A [Sigma C3041]) for 2 hours at ambient temperature. After washing plates three times with buffer B (Sigma P3563), serial dilutions of the test sample in sample diluent buffer (Cedarlane) as well as serial dilutions of a FVIII preparation (ZLB Behring; 200-2 mU/mL) in sample diluent buffer (volumes per well: 100 .mu.L) were incubated for two hours at ambient temperature. After three wash steps with buffer B, 100 .mu.L of a 1:2 dilution in buffer B of the detection antibody (sheep anti-human FVIII IgG, Cedarlane CL20035K-D, peroxidase labelled) were added to each well and incubated for another hour at ambient temperature. After three wash steps with buffer B, 100 .mu.L of substrate solution (1:10 (v/v) TMB OUVF:TMB Buffer OUVG, Dade Behring) were added per well and incubated for 30 minutes at ambient temperature in the dark. Addition of 100 .mu.L stop solution (Dade Behring, OSFA) prepared the samples for reading in a suitable microplate reader at 450 nm wavelength. Concentrations of test samples were then calculated using the standard curve with the FVIII preparation as reference.

Example 6

Pharmacokinetics of Factor VIII Mutants in Rats

[0191] The FVIII mutants were administered intravenously to narcotized CD/Lewis rats (6 rats per substance) with a dose of 100 IU/kg body weight. Blood samples were drawn at appropriate intervals starting at 5 minutes after application of the test substances. FVIII antigen content was subsequently quantified by an ELISA assay specific for human Factor VIII or by a mixed ELISA specific for albumin and FVIII, respectively (see above). The mean values of the treatment groups were used to calculate in vivo recovery after 5 min. Half-lives for each protein were calculated using the time points of the beta phase of elimination according to the formula t.sub.1/2=ln 2/k, whereas k is the slope of the regression line. The result is depicted in FIG. 3.

[0192] The terminal half-life calculated for the chimeric FVIII-HA constructs between 2 and 24 h was 4.97 h for 1413 and 6.86 h for 1211, the terminal half-life calculated for wild type FVIII between 2 and 8 h was 2.17 h. Therefore, a clear increase of the terminal half-life is shown for the chimeric FVIII-HA molecules extending FVIII half-life 2-3-fold.

[0193] Bioavailabilities of the chimeric FVIII-HA constructs and wild-type FVIII are shown in table 3 displaying superior bioavailabilities of the FVIII-HA proteins of the invention.

TABLE-US-00003 TABLE 3 Increased in vivo recovery of FVIII-HA proteins compared with FVIII wild-type (Helixate) in vivo recovery increase in in vivo recovery [% of injected protein 5 compared to Helixate .RTM. min. after i.v. application] (wild-type FVIII) [%] 1211 73.5 123.5 1413 87.7 147.8 Helixate 59.4

Example 7

Functional Half-Life of a Factor VIII Mutant in Rats

[0194] The FVIII mutant pF8-1211 (expressed in HEK-293 cells and purified by IAC) as well as a control preparation (wild type FVIII Helixate NexGen) were administered intravenously to narcotized CD/Lewis rats (6 rats per substance) with a dose of 100 IU/kg body weight. Blood samples were drawn at appropriate intervals starting at 5 minutes after application of the test substances. FVIII antigen content was subsequently quantified for the control group using an ELISA assay specific for human Factor VIII (see example 4). In order to measure the FVIII:C activity of the FVIII mutant in rat plasma an assay was established determining specifically the FVIII mutant activity. In principle, the FVIII mutant was bound from the rat plasma sample to a microtiter plate via an antibody directed against human albumin and FVIII activity was then determined by a chromogenic FVIII:C assay (Coatest VIII:C/4). Briefly, 96-well microtiter plates were coated with the capture antibody (mouse anti-human albumin Mab 3E8, diluted to 5 .mu.g/mL in carbonate/bicarbonate buffer.) over night at ambient temperature. After washing the plates with wash buffer (PBST, =phosphate buffered saline containing 0.05% Tween 20, Sigma P3563), the plates were blocked using non-fat milk in PBS (Phosphate buffered saline) and washed again with wash buffer followed by dilution buffer (50 mM Tris.times.HCl, 100 mM NaCl, 0.05% Tween 20 pH 7.2). Samples were applied in 40 .mu.L volume per well and incubated for 1 h at 37.degree. C. Washing was done using dilution buffer containing 300 mM CaCl2 followed by dilution buffer. The FVIII:C activity determination was performed using Coatest VIII:C/4 reagents. 10 .mu.L dilution buffer and 50 .mu.L Coatest FIXa and FX reagent were applied into the wells and incubated for 5 min at 37.degree. C. Then, 25 .mu.L of CaCl2 solution were added and again incubated for 10 min at 37.degree. C. 50 .mu.L of substrate solution was added and furthermore incubated for 10 min at 37.degree. C. This step was followed by addition of 25 .mu.L of stopping solution (20% acetic acid). A microtiter plate reader was used to read the absorbance at 405 nm. FVIII:C concentrations of the samples were calculated using a standard curve prepared with the FVIII mutant pF8-1211 as reference.

[0195] The FVIII:C respectively FVIII antigen results of the treatment groups were used to calculate the terminal half-lives for the corresponding proteins. The terminal functional half-life calculated for the chimeric FVIII-HSA construct pF8-1211 between 2 and 24 h was 4.44 h, the terminal half-life of FVIII antigen calculated for wild type FVIII between 2 and 8 h was 2.75 h. Therefore, a clear increase of the functional half-life of FVIII:C activity was shown for the chimeric FVIII-HSA molecule (increase by 61% compared to terminal FVIII:Ag half-life of wild type FVIII).

Example 8

In Vitro Stability of FVIII Albumin Insertion Protein

[0196] Table 4 summarizes the results of an expression study of a FVIII albumin insertion protein in serum-free cell culture. HEK-293 cells were transfected in triplicate with pF8-1439 (FVIII albumin insertion) and pF8-457 (FVIII wild-type), respectively, seeded into T80 flasks with equal cell numbers and grown in the absence of stabilizing vWF. Culture supernatant was then harvested after 96, 120 and 144 hours and tested for FVIII activity and antigen content.

TABLE-US-00004 TABLE 4 Culture FVIII FVIII activity/ time antigen* SD** activity* SD** antigen [hrs] [mU/mL] [mU/mL] ratio pF8-457 96 679.0 48.9 1056.7 135.8 1.6 (FVIII wild type) pF8-1439 96 386.7 44.2 1060.0 115.3 2.7 (FVIII albumin) pF8-457 120 819.3 23.2 1720.0 65.6 2.1 (FVIII wild type) pF8-1439 120 389.3 74.9 1420.0 196.7 3.6 (FVIII albumin) pF8-457 144 595.7 59.9 1236.7 388.0 2.1 (FVIII wild type) pF8-1439 144 381.3 50.1 1583.3 226.8 4.2 (FVIII albumin) *mean value from triplicate experiment; **SD, standard deviation

[0197] The results demonstrate a stabilizing effect of albumin when present as an integral part of the FVIII molecule in cell culture. The productivity is not necessarily higher in the case of the insertion protein but the specific activity of the FVIII protein (expressed in the ratio activity/antigen) is significantly higher when the albumin is an integral part of the FVIII molecule (FIG. 3) compared to wild-type FVIII.

Sequence CWU 1

1

4917056DNAHomo Sapiens 1atgcaaatag agctctccac ctgcttcttt ctgtgccttt tgcgattctg ctttagtgcc 60accagaagat actacctggg tgcagtggaa ctgtcatggg actatatgca aagtgatctc 120ggtgagctgc ctgtggacgc aagatttcct cctagagtgc caaaatcttt tccattcaac 180acctcagtcg tgtacaaaaa gactctgttt gtagaattca cggatcacct tttcaacatc 240gctaagccaa ggccaccctg gatgggtctg ctaggtccta ccatccaggc tgaggtttat 300gatacagtgg tcattacact taagaacatg gcttcccatc ctgtcagtct tcatgctgtt 360ggtgtatcct actggaaagc ttctgaggga gctgaatatg atgatcagac cagtcaaagg 420gagaaagaag atgataaagt cttccctggt ggaagccata catatgtctg gcaggtcctg 480aaagagaatg gtccaatggc ctctgaccca ctgtgcctta cctactcata tctttctcat 540gtggacctgg taaaagactt gaattcaggc ctcattggag ccctactagt atgtagagaa 600gggagtctgg ccaaggaaaa gacacagacc ttgcacaaat ttatactact ttttgctgta 660tttgatgaag ggaaaagttg gcactcagaa acaaagaact ccttgatgca ggatagggat 720gctgcatctg ctcgggcctg gcctaaaatg cacacagtca atggttatgt aaacaggtct 780ctgccaggtc tgattggatg ccacaggaaa tcagtctatt ggcatgtgat tggaatgggc 840accactcctg aagtgcactc aatattcctc gaaggtcaca catttcttgt gaggaaccat 900cgccaggcgt ccttggaaat ctcgccaata actttcctta ctgctcaaac actcttgatg 960gaccttggac agtttctact gttttgtcat atctcttccc accaacatga tggcatggaa 1020gcttatgtca aagtagacag ctgtccagag gaaccccaac tacgaatgaa aaataatgaa 1080gaagcggaag actatgatga tgatcttact gattctgaaa tggatgtggt caggtttgat 1140gatgacaact ctccttcctt tatccaaatt cgctcagttg ccaagaagca tcctaaaact 1200tgggtacatt acattgctgc tgaagaggag gactgggact atgctccctt agtcctcgcc 1260cccgatgaca gaagttataa aagtcaatat ttgaacaatg gccctcagcg gattggtagg 1320aagtacaaaa aagtccgatt tatggcatac acagatgaaa cctttaagac tcgtgaagct 1380attcagcatg aatcaggaat cttgggacct ttactttatg gggaagttgg agacacactg 1440ttgattatat ttaagaatca agcaagcaga ccatataaca tctaccctca cggaatcact 1500gatgtccgtc ctttgtattc aaggagatta ccaaaaggtg taaaacattt gaaggatttt 1560ccaattctgc caggagaaat attcaaatat aaatggacag tgactgtaga agatgggcca 1620actaaatcag atcctcggtg cctgacccgc tattactcta gtttcgttaa tatggagaga 1680gatctagctt caggactcat tggccctctc ctcatctgct acaaagaatc tgtagatcaa 1740agaggaaacc agataatgtc agacaagagg aatgtcatcc tgttttctgt atttgatgag 1800aaccgaagct ggtacctcac agagaatata caacgctttc tccccaatcc agctggagtg 1860cagcttgagg atccagagtt ccaagcctcc aacatcatgc acagcatcaa tggctatgtt 1920tttgatagtt tgcagttgtc agtttgtttg catgaggtgg catactggta cattctaagc 1980attggagcac agactgactt cctttctgtc ttcttctctg gatatacctt caaacacaaa 2040atggtctatg aagacacact caccctattc ccattctcag gagaaactgt cttcatgtcg 2100atggaaaacc caggtctatg gattctgggg tgccacaact cagactttcg gaacagaggc 2160atgaccgcct tactgaaggt ttctagttgt gacaagaaca ctggtgatta ttacgaggac 2220agttatgaag atatttcagc atacttgctg agtaaaaaca atgccattga accaagaagc 2280ttctcccaga attcaagaca ccctagcact aggcaaaagc aatttaatgc caccacaatt 2340ccagaaaatg acatagagaa gactgaccct tggtttgcac acagaacacc tatgcctaaa 2400atacaaaatg tctcctctag tgatttgttg atgctcttgc gacagagtcc tactccacat 2460gggctatcct tatctgatct ccaagaagcc aaatatgaga ctttttctga tgatccatca 2520cctggagcaa tagacagtaa taacagcctg tctgaaatga cacacttcag gccacagctc 2580catcacagtg gggacatggt atttacccct gagtcaggcc tccaattaag attaaatgag 2640aaactgggga caactgcagc aacagagttg aagaaacttg atttcaaagt ttctagtaca 2700tcaaataatc tgatttcaac aattccatca gacaatttgg cagcaggtac tgataataca 2760agttccttag gacccccaag tatgccagtt cattatgata gtcaattaga taccactcta 2820tttggcaaaa agtcatctcc ccttactgag tctggtggac ctctgagctt gagtgaagaa 2880aataatgatt caaagttgtt agaatcaggt ttaatgaata gccaagaaag ttcatgggga 2940aaaaatgtat cgtcaacaga gagtggtagg ttatttaaag ggaaaagagc tcatggacct 3000gctttgttga ctaaagataa tgccttattc aaagttagca tctctttgtt aaagacaaac 3060aaaacttcca ataattcagc aactaataga aagactcaca ttgatggccc atcattatta 3120attgagaata gtccatcagt ctggcaaaat atattagaaa gtgacactga gtttaaaaaa 3180gtgacacctt tgattcatga cagaatgctt atggacaaaa atgctacagc tttgaggcta 3240aatcatatgt caaataaaac tacttcatca aaaaacatgg aaatggtcca acagaaaaaa 3300gagggcccca ttccaccaga tgcacaaaat ccagatatgt cgttctttaa gatgctattc 3360ttgccagaat cagcaaggtg gatacaaagg actcatggaa agaactctct gaactctggg 3420caaggcccca gtccaaagca attagtatcc ttaggaccag aaaaatctgt ggaaggtcag 3480aatttcttgt ctgagaaaaa caaagtggta gtaggaaagg gtgaatttac aaaggacgta 3540ggactcaaag agatggtttt tccaagcagc agaaacctat ttcttactaa cttggataat 3600ttacatgaaa ataatacaca caatcaagaa aaaaaaattc aggaagaaat agaaaagaag 3660gaaacattaa tccaagagaa tgtagttttg cctcagatac atacagtgac tggcactaag 3720aatttcatga agaacctttt cttactgagc actaggcaaa atgtagaagg ttcatatgac 3780ggggcatatg ctccagtact tcaagatttt aggtcattaa atgattcaac aaatagaaca 3840aagaaacaca cagctcattt ctcaaaaaaa ggggaggaag aaaacttgga aggcttggga 3900aatcaaacca agcaaattgt agagaaatat gcatgcacca caaggatatc tcctaataca 3960agccagcaga attttgtcac gcaacgtagt aagagagctt tgaaacaatt cagactccca 4020ctagaagaaa cagaacttga aaaaaggata attgtggatg acacctcaac ccagtggtcc 4080aaaaacatga aacatttgac cccgagcacc ctcacacaga tagactacaa tgagaaggag 4140aaaggggcca ttactcagtc tcccttatca gattgcctta cgaggagtca tagcatccct 4200caagcaaata gatctccatt acccattgca aaggtatcat catttccatc tattagacct 4260atatatctga ccagggtcct attccaagac aactcttctc atcttccagc agcatcttat 4320agaaagaaag attctggggt ccaagaaagc agtcatttct tacaaggagc caaaaaaaat 4380aacctttctt tagccattct aaccttggag atgactggtg atcaaagaga ggttggctcc 4440ctggggacaa gtgccacaaa ttcagtcaca tacaagaaag ttgagaacac tgttctcccg 4500aaaccagact tgcccaaaac atctggcaaa gttgaattgc ttccaaaagt tcacatttat 4560cagaaggacc tattccctac ggaaactagc aatgggtctc ctggccatct ggatctcgtg 4620gaagggagcc ttcttcaggg aacagaggga gcgattaagt ggaatgaagc aaacagacct 4680ggaaaagttc cctttctgag agtagcaaca gaaagctctg caaagactcc ctccaagcta 4740ttggatcctc ttgcttggga taaccactat ggtactcaga taccaaaaga agagtggaaa 4800tcccaagaga agtcaccaga aaaaacagct tttaagaaaa aggataccat tttgtccctg 4860aacgcttgtg aaagcaatca tgcaatagca gcaataaatg agggacaaaa taagcccgaa 4920atagaagtca cctgggcaaa gcaaggtagg actgaaaggc tgtgctctca aaacccacca 4980gtcttgaaac gccatcaacg ggaaataact cgtactactc ttcagtcaga tcaagaggaa 5040attgactatg atgataccat atcagttgaa atgaagaagg aagattttga catttatgat 5100gaggatgaaa atcagagccc ccgcagcttt caaaagaaaa cacgacacta ttttattgct 5160gcagtggaga ggctctggga ttatgggatg agtagctccc cacatgttct aagaaacagg 5220gctcagagtg gcagtgtccc tcagttcaag aaagttgttt tccaggaatt tactgatggc 5280tcctttactc agcccttata ccgtggagaa ctaaatgaac atttgggact cctggggcca 5340tatataagag cagaagttga agataatatc atggtaactt tcagaaatca ggcctctcgt 5400ccctattcct tctattctag ccttatttct tatgaggaag atcagaggca aggagcagaa 5460cctagaaaaa actttgtcaa gcctaatgaa accaaaactt acttttggaa agtgcaacat 5520catatggcac ccactaaaga tgagtttgac tgcaaagcct gggcttattt ctctgatgtt 5580gacctggaaa aagatgtgca ctcaggcctg attggacccc ttctggtctg ccacactaac 5640acactgaacc ctgctcatgg gagacaagtg acagtacagg aatttgctct gtttttcacc 5700atctttgatg agaccaaaag ctggtacttc actgaaaata tggaaagaaa ctgcagggct 5760ccctgcaata tccagatgga agatcccact tttaaagaga attatcgctt ccatgcaatc 5820aatggctaca taatggatac actacctggc ttagtaatgg ctcaggatca aaggattcga 5880tggtatctgc tcagcatggg cagcaatgaa aacatccatt ctattcattt cagtggacat 5940gtgttcactg tacgaaaaaa agaggagtat aaaatggcac tgtacaatct ctatccaggt 6000gtttttgaga cagtggaaat gttaccatcc aaagctggaa tttggcgggt ggaatgcctt 6060attggcgagc atctacatgc tgggatgagc acactttttc tggtgtacag caataagtgt 6120cagactcccc tgggaatggc ttctggacac attagagatt ttcagattac agcttcagga 6180caatatggac agtgggcccc aaagctggcc agacttcatt attccggatc aatcaatgcc 6240tggagcacca aggagccctt ttcttggatc aaggtggatc tgttggcacc aatgattatt 6300cacggcatca agacccaggg tgcccgtcag aagttctcca gcctctacat ctctcagttt 6360atcatcatgt atagtcttga tgggaagaag tggcagactt atcgaggaaa ttccactgga 6420accttaatgg tcttctttgg caatgtggat tcatctggga taaaacacaa tatttttaac 6480cctccaatta ttgctcgata catccgtttg cacccaactc attatagcat tcgcagcact 6540cttcgcatgg agttgatggg ctgtgattta aatagttgca gcatgccatt gggaatggag 6600agtaaagcaa tatcagatgc acagattact gcttcatcct actttaccaa tatgtttgcc 6660acctggtctc cttcaaaagc tcgacttcac ctccaaggga ggagtaatgc ctggagacct 6720caggtgaata atccaaaaga gtggctgcaa gtggacttcc agaagacaat gaaagtcaca 6780ggagtaacta ctcagggagt aaaatctctg cttaccagca tgtatgtgaa ggagttcctc 6840atctccagca gtcaagatgg ccatcagtgg actctctttt ttcagaatgg caaagtaaag 6900gtttttcagg gaaatcaaga ctccttcaca cctgtggtga actctctaga cccaccgtta 6960ctgactcgct accttcgaat tcacccccag agttgggtgc accagattgc cctgaggatg 7020gaggttctgg gctgcgaggc acaggacctc tactga 705622332PRTHomo Sapiens 2Ala Thr Arg Arg Tyr Tyr Leu Gly Ala Val Glu Leu Ser Trp Asp Tyr 1 5 10 15 Met Gln Ser Asp Leu Gly Glu Leu Pro Val Asp Ala Arg Phe Pro Pro 20 25 30 Arg Val Pro Lys Ser Phe Pro Phe Asn Thr Ser Val Val Tyr Lys Lys 35 40 45 Thr Leu Phe Val Glu Phe Thr Asp His Leu Phe Asn Ile Ala Lys Pro 50 55 60 Arg Pro Pro Trp Met Gly Leu Leu Gly Pro Thr Ile Gln Ala Glu Val 65 70 75 80 Tyr Asp Thr Val Val Ile Thr Leu Lys Asn Met Ala Ser His Pro Val 85 90 95 Ser Leu His Ala Val Gly Val Ser Tyr Trp Lys Ala Ser Glu Gly Ala 100 105 110 Glu Tyr Asp Asp Gln Thr Ser Gln Arg Glu Lys Glu Asp Asp Lys Val 115 120 125 Phe Pro Gly Gly Ser His Thr Tyr Val Trp Gln Val Leu Lys Glu Asn 130 135 140 Gly Pro Met Ala Ser Asp Pro Leu Cys Leu Thr Tyr Ser Tyr Leu Ser 145 150 155 160 His Val Asp Leu Val Lys Asp Leu Asn Ser Gly Leu Ile Gly Ala Leu 165 170 175 Leu Val Cys Arg Glu Gly Ser Leu Ala Lys Glu Lys Thr Gln Thr Leu 180 185 190 His Lys Phe Ile Leu Leu Phe Ala Val Phe Asp Glu Gly Lys Ser Trp 195 200 205 His Ser Glu Thr Lys Asn Ser Leu Met Gln Asp Arg Asp Ala Ala Ser 210 215 220 Ala Arg Ala Trp Pro Lys Met His Thr Val Asn Gly Tyr Val Asn Arg 225 230 235 240 Ser Leu Pro Gly Leu Ile Gly Cys His Arg Lys Ser Val Tyr Trp His 245 250 255 Val Ile Gly Met Gly Thr Thr Pro Glu Val His Ser Ile Phe Leu Glu 260 265 270 Gly His Thr Phe Leu Val Arg Asn His Arg Gln Ala Ser Leu Glu Ile 275 280 285 Ser Pro Ile Thr Phe Leu Thr Ala Gln Thr Leu Leu Met Asp Leu Gly 290 295 300 Gln Phe Leu Leu Phe Cys His Ile Ser Ser His Gln His Asp Gly Met 305 310 315 320 Glu Ala Tyr Val Lys Val Asp Ser Cys Pro Glu Glu Pro Gln Leu Arg 325 330 335 Met Lys Asn Asn Glu Glu Ala Glu Asp Tyr Asp Asp Asp Leu Thr Asp 340 345 350 Ser Glu Met Asp Val Val Arg Phe Asp Asp Asp Asn Ser Pro Ser Phe 355 360 365 Ile Gln Ile Arg Ser Val Ala Lys Lys His Pro Lys Thr Trp Val His 370 375 380 Tyr Ile Ala Ala Glu Glu Glu Asp Trp Asp Tyr Ala Pro Leu Val Leu 385 390 395 400 Ala Pro Asp Asp Arg Ser Tyr Lys Ser Gln Tyr Leu Asn Asn Gly Pro 405 410 415 Gln Arg Ile Gly Arg Lys Tyr Lys Lys Val Arg Phe Met Ala Tyr Thr 420 425 430 Asp Glu Thr Phe Lys Thr Arg Glu Ala Ile Gln His Glu Ser Gly Ile 435 440 445 Leu Gly Pro Leu Leu Tyr Gly Glu Val Gly Asp Thr Leu Leu Ile Ile 450 455 460 Phe Lys Asn Gln Ala Ser Arg Pro Tyr Asn Ile Tyr Pro His Gly Ile 465 470 475 480 Thr Asp Val Arg Pro Leu Tyr Ser Arg Arg Leu Pro Lys Gly Val Lys 485 490 495 His Leu Lys Asp Phe Pro Ile Leu Pro Gly Glu Ile Phe Lys Tyr Lys 500 505 510 Trp Thr Val Thr Val Glu Asp Gly Pro Thr Lys Ser Asp Pro Arg Cys 515 520 525 Leu Thr Arg Tyr Tyr Ser Ser Phe Val Asn Met Glu Arg Asp Leu Ala 530 535 540 Ser Gly Leu Ile Gly Pro Leu Leu Ile Cys Tyr Lys Glu Ser Val Asp 545 550 555 560 Gln Arg Gly Asn Gln Ile Met Ser Asp Lys Arg Asn Val Ile Leu Phe 565 570 575 Ser Val Phe Asp Glu Asn Arg Ser Trp Tyr Leu Thr Glu Asn Ile Gln 580 585 590 Arg Phe Leu Pro Asn Pro Ala Gly Val Gln Leu Glu Asp Pro Glu Phe 595 600 605 Gln Ala Ser Asn Ile Met His Ser Ile Asn Gly Tyr Val Phe Asp Ser 610 615 620 Leu Gln Leu Ser Val Cys Leu His Glu Val Ala Tyr Trp Tyr Ile Leu 625 630 635 640 Ser Ile Gly Ala Gln Thr Asp Phe Leu Ser Val Phe Phe Ser Gly Tyr 645 650 655 Thr Phe Lys His Lys Met Val Tyr Glu Asp Thr Leu Thr Leu Phe Pro 660 665 670 Phe Ser Gly Glu Thr Val Phe Met Ser Met Glu Asn Pro Gly Leu Trp 675 680 685 Ile Leu Gly Cys His Asn Ser Asp Phe Arg Asn Arg Gly Met Thr Ala 690 695 700 Leu Leu Lys Val Ser Ser Cys Asp Lys Asn Thr Gly Asp Tyr Tyr Glu 705 710 715 720 Asp Ser Tyr Glu Asp Ile Ser Ala Tyr Leu Leu Ser Lys Asn Asn Ala 725 730 735 Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Arg Ser Thr Arg 740 745 750 Gln Lys Gln Phe Asn Ala Thr Thr Ile Pro Glu Asn Asp Ile Glu Lys 755 760 765 Thr Asp Pro Trp Phe Ala His Arg Thr Pro Met Pro Lys Ile Gln Asn 770 775 780 Val Ser Ser Ser Asp Leu Leu Met Leu Leu Arg Gln Ser Pro Thr Pro 785 790 795 800 His Gly Leu Ser Leu Ser Asp Leu Gln Glu Ala Lys Tyr Glu Thr Phe 805 810 815 Ser Asp Asp Pro Ser Pro Gly Ala Ile Asp Ser Asn Asn Ser Leu Ser 820 825 830 Glu Met Thr His Phe Arg Pro Gln Leu His His Ser Gly Asp Met Val 835 840 845 Phe Thr Pro Glu Ser Gly Leu Gln Leu Arg Leu Asn Glu Lys Leu Gly 850 855 860 Thr Thr Ala Ala Thr Glu Leu Lys Lys Leu Asp Phe Lys Val Ser Ser 865 870 875 880 Thr Ser Asn Asn Leu Ile Ser Thr Ile Pro Ser Asp Asn Leu Ala Ala 885 890 895 Gly Thr Asp Asn Thr Ser Ser Leu Gly Pro Pro Ser Met Pro Val His 900 905 910 Tyr Asp Ser Gln Leu Asp Thr Thr Leu Phe Gly Lys Lys Ser Ser Pro 915 920 925 Leu Thr Glu Ser Gly Gly Pro Leu Ser Leu Ser Glu Glu Asn Asn Asp 930 935 940 Ser Lys Leu Leu Glu Ser Gly Leu Met Asn Ser Gln Glu Ser Ser Trp 945 950 955 960 Gly Lys Asn Val Ser Ser Thr Glu Ser Gly Arg Leu Phe Lys Gly Lys 965 970 975 Arg Ala His Gly Pro Ala Leu Leu Thr Lys Asp Asn Ala Leu Phe Lys 980 985 990 Val Ser Ile Ser Leu Leu Lys Thr Asn Lys Thr Ser Asn Asn Ser Ala 995 1000 1005 Thr Asn Arg Lys Thr His Ile Asp Gly Pro Ser Leu Leu Ile Glu 1010 1015 1020 Asn Ser Pro Ser Val Trp Gln Asn Ile Leu Glu Ser Asp Thr Glu 1025 1030 1035 Phe Lys Lys Val Thr Pro Leu Ile His Asp Arg Met Leu Met Asp 1040 1045 1050 Lys Asn Ala Thr Ala Leu Arg Leu Asn His Met Ser Asn Lys Thr 1055 1060 1065 Thr Ser Ser Lys Asn Met Glu Met Val Gln Gln Lys Lys Glu Gly 1070 1075 1080 Pro Ile Pro Pro Asp Ala Gln Asn Pro Asp Met Ser Phe Phe Lys 1085 1090 1095 Met Leu Phe Leu Pro Glu Ser Ala Arg Trp Ile Gln Arg Thr His 1100 1105 1110 Gly Lys Asn Ser Leu Asn Ser Gly Gln Gly Pro Ser Pro Lys Gln 1115 1120 1125 Leu Val Ser Leu Gly Pro Glu Lys Ser Val Glu Gly Gln Asn Phe 1130 1135 1140 Leu Ser Glu Lys Asn Lys Val Val Val Gly Lys Gly Glu Phe Thr 1145 1150 1155 Lys Asp Val Gly Leu Lys Glu Met Val Phe Pro Ser Ser Arg Asn 1160 1165 1170 Leu Phe Leu Thr Asn Leu Asp Asn Leu His Glu Asn Asn Thr His 1175 1180 1185 Asn Gln Glu Lys Lys Ile Gln Glu Glu Ile Glu Lys Lys Glu Thr 1190 1195 1200 Leu Ile Gln Glu Asn

Val Val Leu Pro Gln Ile His Thr Val Thr 1205 1210 1215 Gly Thr Lys Asn Phe Met Lys Asn Leu Phe Leu Leu Ser Thr Arg 1220 1225 1230 Gln Asn Val Glu Gly Ser Tyr Asp Gly Ala Tyr Ala Pro Val Leu 1235 1240 1245 Gln Asp Phe Arg Ser Leu Asn Asp Ser Thr Asn Arg Thr Lys Lys 1250 1255 1260 His Thr Ala His Phe Ser Lys Lys Gly Glu Glu Glu Asn Leu Glu 1265 1270 1275 Gly Leu Gly Asn Gln Thr Lys Gln Ile Val Glu Lys Tyr Ala Cys 1280 1285 1290 Thr Thr Arg Ile Ser Pro Asn Thr Ser Gln Gln Asn Phe Val Thr 1295 1300 1305 Gln Arg Ser Lys Arg Ala Leu Lys Gln Phe Arg Leu Pro Leu Glu 1310 1315 1320 Glu Thr Glu Leu Glu Lys Arg Ile Ile Val Asp Asp Thr Ser Thr 1325 1330 1335 Gln Trp Ser Lys Asn Met Lys His Leu Thr Pro Ser Thr Leu Thr 1340 1345 1350 Gln Ile Asp Tyr Asn Glu Lys Glu Lys Gly Ala Ile Thr Gln Ser 1355 1360 1365 Pro Leu Ser Asp Cys Leu Thr Arg Ser His Ser Ile Pro Gln Ala 1370 1375 1380 Asn Arg Ser Pro Leu Pro Ile Ala Lys Val Ser Ser Phe Pro Ser 1385 1390 1395 Ile Arg Pro Ile Tyr Leu Thr Arg Val Leu Phe Gln Asp Asn Ser 1400 1405 1410 Ser His Leu Pro Ala Ala Ser Tyr Arg Lys Lys Asp Ser Gly Val 1415 1420 1425 Gln Glu Ser Ser His Phe Leu Gln Gly Ala Lys Lys Asn Asn Leu 1430 1435 1440 Ser Leu Ala Ile Leu Thr Leu Glu Met Thr Gly Asp Gln Arg Glu 1445 1450 1455 Val Gly Ser Leu Gly Thr Ser Ala Thr Asn Ser Val Thr Tyr Lys 1460 1465 1470 Lys Val Glu Asn Thr Val Leu Pro Lys Pro Asp Leu Pro Lys Thr 1475 1480 1485 Ser Gly Lys Val Glu Leu Leu Pro Lys Val His Ile Tyr Gln Lys 1490 1495 1500 Asp Leu Phe Pro Thr Glu Thr Ser Asn Gly Ser Pro Gly His Leu 1505 1510 1515 Asp Leu Val Glu Gly Ser Leu Leu Gln Gly Thr Glu Gly Ala Ile 1520 1525 1530 Lys Trp Asn Glu Ala Asn Arg Pro Gly Lys Val Pro Phe Leu Arg 1535 1540 1545 Val Ala Thr Glu Ser Ser Ala Lys Thr Pro Ser Lys Leu Leu Asp 1550 1555 1560 Pro Leu Ala Trp Asp Asn His Tyr Gly Thr Gln Ile Pro Lys Glu 1565 1570 1575 Glu Trp Lys Ser Gln Glu Lys Ser Pro Glu Lys Thr Ala Phe Lys 1580 1585 1590 Lys Lys Asp Thr Ile Leu Ser Leu Asn Ala Cys Glu Ser Asn His 1595 1600 1605 Ala Ile Ala Ala Ile Asn Glu Gly Gln Asn Lys Pro Glu Ile Glu 1610 1615 1620 Val Thr Trp Ala Lys Gln Gly Arg Thr Glu Arg Leu Cys Ser Gln 1625 1630 1635 Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr 1640 1645 1650 Thr Leu Gln Ser Asp Gln Glu Glu Ile Asp Tyr Asp Asp Thr Ile 1655 1660 1665 Ser Val Glu Met Lys Lys Glu Asp Phe Asp Ile Tyr Asp Glu Asp 1670 1675 1680 Glu Asn Gln Ser Pro Arg Ser Phe Gln Lys Lys Thr Arg His Tyr 1685 1690 1695 Phe Ile Ala Ala Val Glu Arg Leu Trp Asp Tyr Gly Met Ser Ser 1700 1705 1710 Ser Pro His Val Leu Arg Asn Arg Ala Gln Ser Gly Ser Val Pro 1715 1720 1725 Gln Phe Lys Lys Val Val Phe Gln Glu Phe Thr Asp Gly Ser Phe 1730 1735 1740 Thr Gln Pro Leu Tyr Arg Gly Glu Leu Asn Glu His Leu Gly Leu 1745 1750 1755 Leu Gly Pro Tyr Ile Arg Ala Glu Val Glu Asp Asn Ile Met Val 1760 1765 1770 Thr Phe Arg Asn Gln Ala Ser Arg Pro Tyr Ser Phe Tyr Ser Ser 1775 1780 1785 Leu Ile Ser Tyr Glu Glu Asp Gln Arg Gln Gly Ala Glu Pro Arg 1790 1795 1800 Lys Asn Phe Val Lys Pro Asn Glu Thr Lys Thr Tyr Phe Trp Lys 1805 1810 1815 Val Gln His His Met Ala Pro Thr Lys Asp Glu Phe Asp Cys Lys 1820 1825 1830 Ala Trp Ala Tyr Phe Ser Asp Val Asp Leu Glu Lys Asp Val His 1835 1840 1845 Ser Gly Leu Ile Gly Pro Leu Leu Val Cys His Thr Asn Thr Leu 1850 1855 1860 Asn Pro Ala His Gly Arg Gln Val Thr Val Gln Glu Phe Ala Leu 1865 1870 1875 Phe Phe Thr Ile Phe Asp Glu Thr Lys Ser Trp Tyr Phe Thr Glu 1880 1885 1890 Asn Met Glu Arg Asn Cys Arg Ala Pro Cys Asn Ile Gln Met Glu 1895 1900 1905 Asp Pro Thr Phe Lys Glu Asn Tyr Arg Phe His Ala Ile Asn Gly 1910 1915 1920 Tyr Ile Met Asp Thr Leu Pro Gly Leu Val Met Ala Gln Asp Gln 1925 1930 1935 Arg Ile Arg Trp Tyr Leu Leu Ser Met Gly Ser Asn Glu Asn Ile 1940 1945 1950 His Ser Ile His Phe Ser Gly His Val Phe Thr Val Arg Lys Lys 1955 1960 1965 Glu Glu Tyr Lys Met Ala Leu Tyr Asn Leu Tyr Pro Gly Val Phe 1970 1975 1980 Glu Thr Val Glu Met Leu Pro Ser Lys Ala Gly Ile Trp Arg Val 1985 1990 1995 Glu Cys Leu Ile Gly Glu His Leu His Ala Gly Met Ser Thr Leu 2000 2005 2010 Phe Leu Val Tyr Ser Asn Lys Cys Gln Thr Pro Leu Gly Met Ala 2015 2020 2025 Ser Gly His Ile Arg Asp Phe Gln Ile Thr Ala Ser Gly Gln Tyr 2030 2035 2040 Gly Gln Trp Ala Pro Lys Leu Ala Arg Leu His Tyr Ser Gly Ser 2045 2050 2055 Ile Asn Ala Trp Ser Thr Lys Glu Pro Phe Ser Trp Ile Lys Val 2060 2065 2070 Asp Leu Leu Ala Pro Met Ile Ile His Gly Ile Lys Thr Gln Gly 2075 2080 2085 Ala Arg Gln Lys Phe Ser Ser Leu Tyr Ile Ser Gln Phe Ile Ile 2090 2095 2100 Met Tyr Ser Leu Asp Gly Lys Lys Trp Gln Thr Tyr Arg Gly Asn 2105 2110 2115 Ser Thr Gly Thr Leu Met Val Phe Phe Gly Asn Val Asp Ser Ser 2120 2125 2130 Gly Ile Lys His Asn Ile Phe Asn Pro Pro Ile Ile Ala Arg Tyr 2135 2140 2145 Ile Arg Leu His Pro Thr His Tyr Ser Ile Arg Ser Thr Leu Arg 2150 2155 2160 Met Glu Leu Met Gly Cys Asp Leu Asn Ser Cys Ser Met Pro Leu 2165 2170 2175 Gly Met Glu Ser Lys Ala Ile Ser Asp Ala Gln Ile Thr Ala Ser 2180 2185 2190 Ser Tyr Phe Thr Asn Met Phe Ala Thr Trp Ser Pro Ser Lys Ala 2195 2200 2205 Arg Leu His Leu Gln Gly Arg Ser Asn Ala Trp Arg Pro Gln Val 2210 2215 2220 Asn Asn Pro Lys Glu Trp Leu Gln Val Asp Phe Gln Lys Thr Met 2225 2230 2235 Lys Val Thr Gly Val Thr Thr Gln Gly Val Lys Ser Leu Leu Thr 2240 2245 2250 Ser Met Tyr Val Lys Glu Phe Leu Ile Ser Ser Ser Gln Asp Gly 2255 2260 2265 His Gln Trp Thr Leu Phe Phe Gln Asn Gly Lys Val Lys Val Phe 2270 2275 2280 Gln Gly Asn Gln Asp Ser Phe Thr Pro Val Val Asn Ser Leu Asp 2285 2290 2295 Pro Pro Leu Leu Thr Arg Tyr Leu Arg Ile His Pro Gln Ser Trp 2300 2305 2310 Val His Gln Ile Ala Leu Arg Met Glu Val Leu Gly Cys Glu Ala 2315 2320 2325 Gln Asp Leu Tyr 2330 3585PRTHomo Sapiens 3Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp Leu Gly Glu 1 5 10 15 Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln Tyr Leu Gln 20 25 30 Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu Val Thr Glu 35 40 45 Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn Cys Asp Lys 50 55 60 Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val Ala Thr Leu 65 70 75 80 Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys Gln Glu Pro 85 90 95 Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn Pro Asn Leu 100 105 110 Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr Ala Phe His 115 120 125 Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu Ile Ala Arg 130 135 140 Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe Ala Lys Arg 145 150 155 160 Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp Lys Ala Ala 165 170 175 Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly Lys Ala Ser 180 185 190 Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys Phe Gly Glu 195 200 205 Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln Arg Phe Pro 210 215 220 Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp Leu Thr Lys 225 230 235 240 Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys Ala Asp Asp 245 250 255 Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln Asp Ser Ile Ser 260 265 270 Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu Leu Glu Lys Ser His 275 280 285 Cys Ile Ala Glu Val Glu Asn Asp Glu Met Pro Ala Asp Leu Pro Ser 290 295 300 Leu Ala Ala Asp Phe Val Glu Ser Lys Asp Val Cys Lys Asn Tyr Ala 305 310 315 320 Glu Ala Lys Asp Val Phe Leu Gly Met Phe Leu Tyr Glu Tyr Ala Arg 325 330 335 Arg His Pro Asp Tyr Ser Val Val Leu Leu Leu Arg Leu Ala Lys Thr 340 345 350 Tyr Glu Thr Thr Leu Glu Lys Cys Cys Ala Ala Ala Asp Pro His Glu 355 360 365 Cys Tyr Ala Lys Val Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro 370 375 380 Gln Asn Leu Ile Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu 385 390 395 400 Tyr Lys Phe Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro 405 410 415 Gln Val Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys 420 425 430 Val Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 435 440 445 Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu His 450 455 460 Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr Glu Ser 465 470 475 480 Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val Asp Glu Thr 485 490 495 Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr Phe His Ala Asp 500 505 510 Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile Lys Lys Gln Thr Ala 515 520 525 Leu Val Glu Leu Val Lys His Lys Pro Lys Ala Thr Lys Glu Gln Leu 530 535 540 Lys Ala Val Met Asp Asp Phe Ala Ala Phe Val Glu Lys Cys Cys Lys 545 550 555 560 Ala Asp Asp Lys Glu Thr Cys Phe Ala Glu Glu Gly Lys Lys Leu Val 565 570 575 Ala Ala Ser Gln Ala Ala Leu Gly Leu 580 585 42043PRTArtificialAmino acid sequence of mature human FVIII with B domain replacement by human albumin 4Ala Thr Arg Arg Tyr Tyr Leu Gly Ala Val Glu Leu Ser Trp Asp Tyr 1 5 10 15 Met Gln Ser Asp Leu Gly Glu Leu Pro Val Asp Ala Arg Phe Pro Pro 20 25 30 Arg Val Pro Lys Ser Phe Pro Phe Asn Thr Ser Val Val Tyr Lys Lys 35 40 45 Thr Leu Phe Val Glu Phe Thr Asp His Leu Phe Asn Ile Ala Lys Pro 50 55 60 Arg Pro Pro Trp Met Gly Leu Leu Gly Pro Thr Ile Gln Ala Glu Val 65 70 75 80 Tyr Asp Thr Val Val Ile Thr Leu Lys Asn Met Ala Ser His Pro Val 85 90 95 Ser Leu His Ala Val Gly Val Ser Tyr Trp Lys Ala Ser Glu Gly Ala 100 105 110 Glu Tyr Asp Asp Gln Thr Ser Gln Arg Glu Lys Glu Asp Asp Lys Val 115 120 125 Phe Pro Gly Gly Ser His Thr Tyr Val Trp Gln Val Leu Lys Glu Asn 130 135 140 Gly Pro Met Ala Ser Asp Pro Leu Cys Leu Thr Tyr Ser Tyr Leu Ser 145 150 155 160 His Val Asp Leu Val Lys Asp Leu Asn Ser Gly Leu Ile Gly Ala Leu 165 170 175 Leu Val Cys Arg Glu Gly Ser Leu Ala Lys Glu Lys Thr Gln Thr Leu 180 185 190 His Lys Phe Ile Leu Leu Phe Ala Val Phe Asp Glu Gly Lys Ser Trp 195 200 205 His Ser Glu Thr Lys Asn Ser Leu Met Gln Asp Arg Asp Ala Ala Ser 210 215 220 Ala Arg Ala Trp Pro Lys Met His Thr Val Asn Gly Tyr Val Asn Arg 225 230 235 240 Ser Leu Pro Gly Leu Ile Gly Cys His Arg Lys Ser Val Tyr Trp His 245 250 255 Val Ile Gly Met Gly Thr Thr Pro Glu Val His Ser Ile Phe Leu Glu 260 265 270 Gly His Thr Phe Leu Val Arg Asn His Arg Gln Ala Ser Leu Glu Ile 275 280 285 Ser Pro Ile Thr Phe Leu Thr Ala Gln Thr Leu Leu Met Asp Leu Gly 290 295 300 Gln Phe Leu Leu Phe Cys His Ile Ser Ser His Gln His Asp Gly Met 305 310 315 320 Glu Ala Tyr Val Lys Val Asp Ser Cys Pro Glu Glu Pro Gln Leu Arg 325 330 335 Met Lys Asn Asn Glu Glu Ala Glu Asp Tyr Asp Asp Asp Leu Thr Asp 340 345 350 Ser Glu Met Asp Val Val Arg Phe Asp Asp Asp Asn Ser Pro Ser Phe 355 360 365 Ile Gln Ile Arg Ser Val Ala Lys Lys His Pro Lys Thr Trp Val His 370 375 380 Tyr Ile Ala Ala Glu Glu Glu Asp Trp Asp Tyr Ala Pro Leu Val Leu 385 390 395 400 Ala Pro Asp Asp Arg Ser Tyr Lys Ser Gln Tyr Leu Asn Asn Gly Pro 405 410 415 Gln Arg Ile Gly Arg Lys Tyr Lys Lys Val Arg Phe Met Ala Tyr Thr 420 425 430 Asp Glu Thr Phe Lys Thr Arg Glu Ala Ile Gln His Glu Ser Gly Ile 435 440 445 Leu Gly Pro Leu Leu Tyr Gly Glu Val Gly Asp Thr Leu Leu Ile Ile 450 455 460 Phe Lys Asn Gln Ala Ser Arg Pro Tyr Asn Ile Tyr Pro His Gly Ile 465 470 475 480 Thr Asp Val Arg Pro Leu Tyr Ser Arg Arg Leu Pro Lys Gly Val Lys 485 490 495 His Leu Lys Asp Phe Pro Ile Leu Pro Gly Glu Ile Phe Lys Tyr Lys 500 505 510 Trp Thr Val Thr Val Glu Asp Gly Pro Thr Lys Ser Asp Pro

Arg Cys 515 520 525 Leu Thr Arg Tyr Tyr Ser Ser Phe Val Asn Met Glu Arg Asp Leu Ala 530 535 540 Ser Gly Leu Ile Gly Pro Leu Leu Ile Cys Tyr Lys Glu Ser Val Asp 545 550 555 560 Gln Arg Gly Asn Gln Ile Met Ser Asp Lys Arg Asn Val Ile Leu Phe 565 570 575 Ser Val Phe Asp Glu Asn Arg Ser Trp Tyr Leu Thr Glu Asn Ile Gln 580 585 590 Arg Phe Leu Pro Asn Pro Ala Gly Val Gln Leu Glu Asp Pro Glu Phe 595 600 605 Gln Ala Ser Asn Ile Met His Ser Ile Asn Gly Tyr Val Phe Asp Ser 610 615 620 Leu Gln Leu Ser Val Cys Leu His Glu Val Ala Tyr Trp Tyr Ile Leu 625 630 635 640 Ser Ile Gly Ala Gln Thr Asp Phe Leu Ser Val Phe Phe Ser Gly Tyr 645 650 655 Thr Phe Lys His Lys Met Val Tyr Glu Asp Thr Leu Thr Leu Phe Pro 660 665 670 Phe Ser Gly Glu Thr Val Phe Met Ser Met Glu Asn Pro Gly Leu Trp 675 680 685 Ile Leu Gly Cys His Asn Ser Asp Phe Arg Asn Arg Gly Met Thr Ala 690 695 700 Leu Leu Lys Val Ser Ser Cys Asp Lys Asn Thr Gly Asp Tyr Tyr Glu 705 710 715 720 Asp Ser Tyr Glu Asp Ile Ser Ala Tyr Leu Leu Ser Lys Asn Asn Ala 725 730 735 Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Arg Ser Thr Arg 740 745 750 Gln Lys Gln Asp Ala His Lys Ser Glu Val Ala His Arg Phe Lys Asp 755 760 765 Leu Gly Glu Glu Asn Phe Lys Ala Leu Val Leu Ile Ala Phe Ala Gln 770 775 780 Tyr Leu Gln Gln Cys Pro Phe Glu Asp His Val Lys Leu Val Asn Glu 785 790 795 800 Val Thr Glu Phe Ala Lys Thr Cys Val Ala Asp Glu Ser Ala Glu Asn 805 810 815 Cys Asp Lys Ser Leu His Thr Leu Phe Gly Asp Lys Leu Cys Thr Val 820 825 830 Ala Thr Leu Arg Glu Thr Tyr Gly Glu Met Ala Asp Cys Cys Ala Lys 835 840 845 Gln Glu Pro Glu Arg Asn Glu Cys Phe Leu Gln His Lys Asp Asp Asn 850 855 860 Pro Asn Leu Pro Arg Leu Val Arg Pro Glu Val Asp Val Met Cys Thr 865 870 875 880 Ala Phe His Asp Asn Glu Glu Thr Phe Leu Lys Lys Tyr Leu Tyr Glu 885 890 895 Ile Ala Arg Arg His Pro Tyr Phe Tyr Ala Pro Glu Leu Leu Phe Phe 900 905 910 Ala Lys Arg Tyr Lys Ala Ala Phe Thr Glu Cys Cys Gln Ala Ala Asp 915 920 925 Lys Ala Ala Cys Leu Leu Pro Lys Leu Asp Glu Leu Arg Asp Glu Gly 930 935 940 Lys Ala Ser Ser Ala Lys Gln Arg Leu Lys Cys Ala Ser Leu Gln Lys 945 950 955 960 Phe Gly Glu Arg Ala Phe Lys Ala Trp Ala Val Ala Arg Leu Ser Gln 965 970 975 Arg Phe Pro Lys Ala Glu Phe Ala Glu Val Ser Lys Leu Val Thr Asp 980 985 990 Leu Thr Lys Val His Thr Glu Cys Cys His Gly Asp Leu Leu Glu Cys 995 1000 1005 Ala Asp Asp Arg Ala Asp Leu Ala Lys Tyr Ile Cys Glu Asn Gln 1010 1015 1020 Asp Ser Ile Ser Ser Lys Leu Lys Glu Cys Cys Glu Lys Pro Leu 1025 1030 1035 Leu Glu Lys Ser His Cys Ile Ala Glu Val Glu Asn Asp Glu Met 1040 1045 1050 Pro Ala Asp Leu Pro Ser Leu Ala Ala Asp Phe Val Glu Ser Lys 1055 1060 1065 Asp Val Cys Lys Asn Tyr Ala Glu Ala Lys Asp Val Phe Leu Gly 1070 1075 1080 Met Phe Leu Tyr Glu Tyr Ala Arg Arg His Pro Asp Tyr Ser Val 1085 1090 1095 Val Leu Leu Leu Arg Leu Ala Lys Thr Tyr Glu Thr Thr Leu Glu 1100 1105 1110 Lys Cys Cys Ala Ala Ala Asp Pro His Glu Cys Tyr Ala Lys Val 1115 1120 1125 Phe Asp Glu Phe Lys Pro Leu Val Glu Glu Pro Gln Asn Leu Ile 1130 1135 1140 Lys Gln Asn Cys Glu Leu Phe Glu Gln Leu Gly Glu Tyr Lys Phe 1145 1150 1155 Gln Asn Ala Leu Leu Val Arg Tyr Thr Lys Lys Val Pro Gln Val 1160 1165 1170 Ser Thr Pro Thr Leu Val Glu Val Ser Arg Asn Leu Gly Lys Val 1175 1180 1185 Gly Ser Lys Cys Cys Lys His Pro Glu Ala Lys Arg Met Pro Cys 1190 1195 1200 Ala Glu Asp Tyr Leu Ser Val Val Leu Asn Gln Leu Cys Val Leu 1205 1210 1215 His Glu Lys Thr Pro Val Ser Asp Arg Val Thr Lys Cys Cys Thr 1220 1225 1230 Glu Ser Leu Val Asn Arg Arg Pro Cys Phe Ser Ala Leu Glu Val 1235 1240 1245 Asp Glu Thr Tyr Val Pro Lys Glu Phe Asn Ala Glu Thr Phe Thr 1250 1255 1260 Phe His Ala Asp Ile Cys Thr Leu Ser Glu Lys Glu Arg Gln Ile 1265 1270 1275 Lys Lys Gln Thr Ala Leu Val Glu Leu Val Lys His Lys Pro Lys 1280 1285 1290 Ala Thr Lys Glu Gln Leu Lys Ala Val Met Asp Asp Phe Ala Ala 1295 1300 1305 Phe Val Glu Lys Cys Cys Lys Ala Asp Asp Lys Glu Thr Cys Phe 1310 1315 1320 Ala Glu Glu Gly Lys Lys Leu Val Ala Ala Ser Gln Ala Ala Leu 1325 1330 1335 Gly Leu Gly Arg Thr Glu Arg Leu Cys Ser Gln Asn Pro Pro Val 1340 1345 1350 Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr Thr Leu Gln Ser 1355 1360 1365 Asp Gln Glu Glu Ile Asp Tyr Asp Asp Thr Ile Ser Val Glu Met 1370 1375 1380 Lys Lys Glu Asp Phe Asp Ile Tyr Asp Glu Asp Glu Asn Gln Ser 1385 1390 1395 Pro Arg Ser Phe Gln Lys Lys Thr Arg His Tyr Phe Ile Ala Ala 1400 1405 1410 Val Glu Arg Leu Trp Asp Tyr Gly Met Ser Ser Ser Pro His Val 1415 1420 1425 Leu Arg Asn Arg Ala Gln Ser Gly Ser Val Pro Gln Phe Lys Lys 1430 1435 1440 Val Val Phe Gln Glu Phe Thr Asp Gly Ser Phe Thr Gln Pro Leu 1445 1450 1455 Tyr Arg Gly Glu Leu Asn Glu His Leu Gly Leu Leu Gly Pro Tyr 1460 1465 1470 Ile Arg Ala Glu Val Glu Asp Asn Ile Met Val Thr Phe Arg Asn 1475 1480 1485 Gln Ala Ser Arg Pro Tyr Ser Phe Tyr Ser Ser Leu Ile Ser Tyr 1490 1495 1500 Glu Glu Asp Gln Arg Gln Gly Ala Glu Pro Arg Lys Asn Phe Val 1505 1510 1515 Lys Pro Asn Glu Thr Lys Thr Tyr Phe Trp Lys Val Gln His His 1520 1525 1530 Met Ala Pro Thr Lys Asp Glu Phe Asp Cys Lys Ala Trp Ala Tyr 1535 1540 1545 Phe Ser Asp Val Asp Leu Glu Lys Asp Val His Ser Gly Leu Ile 1550 1555 1560 Gly Pro Leu Leu Val Cys His Thr Asn Thr Leu Asn Pro Ala His 1565 1570 1575 Gly Arg Gln Val Thr Val Gln Glu Phe Ala Leu Phe Phe Thr Ile 1580 1585 1590 Phe Asp Glu Thr Lys Ser Trp Tyr Phe Thr Glu Asn Met Glu Arg 1595 1600 1605 Asn Cys Arg Ala Pro Cys Asn Ile Gln Met Glu Asp Pro Thr Phe 1610 1615 1620 Lys Glu Asn Tyr Arg Phe His Ala Ile Asn Gly Tyr Ile Met Asp 1625 1630 1635 Thr Leu Pro Gly Leu Val Met Ala Gln Asp Gln Arg Ile Arg Trp 1640 1645 1650 Tyr Leu Leu Ser Met Gly Ser Asn Glu Asn Ile His Ser Ile His 1655 1660 1665 Phe Ser Gly His Val Phe Thr Val Arg Lys Lys Glu Glu Tyr Lys 1670 1675 1680 Met Ala Leu Tyr Asn Leu Tyr Pro Gly Val Phe Glu Thr Val Glu 1685 1690 1695 Met Leu Pro Ser Lys Ala Gly Ile Trp Arg Val Glu Cys Leu Ile 1700 1705 1710 Gly Glu His Leu His Ala Gly Met Ser Thr Leu Phe Leu Val Tyr 1715 1720 1725 Ser Asn Lys Cys Gln Thr Pro Leu Gly Met Ala Ser Gly His Ile 1730 1735 1740 Arg Asp Phe Gln Ile Thr Ala Ser Gly Gln Tyr Gly Gln Trp Ala 1745 1750 1755 Pro Lys Leu Ala Arg Leu His Tyr Ser Gly Ser Ile Asn Ala Trp 1760 1765 1770 Ser Thr Lys Glu Pro Phe Ser Trp Ile Lys Val Asp Leu Leu Ala 1775 1780 1785 Pro Met Ile Ile His Gly Ile Lys Thr Gln Gly Ala Arg Gln Lys 1790 1795 1800 Phe Ser Ser Leu Tyr Ile Ser Gln Phe Ile Ile Met Tyr Ser Leu 1805 1810 1815 Asp Gly Lys Lys Trp Gln Thr Tyr Arg Gly Asn Ser Thr Gly Thr 1820 1825 1830 Leu Met Val Phe Phe Gly Asn Val Asp Ser Ser Gly Ile Lys His 1835 1840 1845 Asn Ile Phe Asn Pro Pro Ile Ile Ala Arg Tyr Ile Arg Leu His 1850 1855 1860 Pro Thr His Tyr Ser Ile Arg Ser Thr Leu Arg Met Glu Leu Met 1865 1870 1875 Gly Cys Asp Leu Asn Ser Cys Ser Met Pro Leu Gly Met Glu Ser 1880 1885 1890 Lys Ala Ile Ser Asp Ala Gln Ile Thr Ala Ser Ser Tyr Phe Thr 1895 1900 1905 Asn Met Phe Ala Thr Trp Ser Pro Ser Lys Ala Arg Leu His Leu 1910 1915 1920 Gln Gly Arg Ser Asn Ala Trp Arg Pro Gln Val Asn Asn Pro Lys 1925 1930 1935 Glu Trp Leu Gln Val Asp Phe Gln Lys Thr Met Lys Val Thr Gly 1940 1945 1950 Val Thr Thr Gln Gly Val Lys Ser Leu Leu Thr Ser Met Tyr Val 1955 1960 1965 Lys Glu Phe Leu Ile Ser Ser Ser Gln Asp Gly His Gln Trp Thr 1970 1975 1980 Leu Phe Phe Gln Asn Gly Lys Val Lys Val Phe Gln Gly Asn Gln 1985 1990 1995 Asp Ser Phe Thr Pro Val Val Asn Ser Leu Asp Pro Pro Leu Leu 2000 2005 2010 Thr Arg Tyr Leu Arg Ile His Pro Gln Ser Trp Val His Gln Ile 2015 2020 2025 Ala Leu Arg Met Glu Val Leu Gly Cys Glu Ala Gln Asp Leu Tyr 2030 2035 2040 521DNAArtificialPrimer 5cagcttgagg atccagagtt c 21630DNAArtificialPrimer 6gtgaccggtc ttttgcctag tgctagggtg 30726DNAArtificialPrimer 7gtgaccggta ggactgaaag gctgtg 26823DNAArtificialPrimer 8gattgatccg gaataatgaa gtc 23934DNAArtificialPrimer 9gcgaaccggt caggatgcac acaagagtga ggtt 341030DNAArtificialPrimer 10cgcaccggtt aagcctaagg cagcttgact 301129DNAArtificialPrimer 11ctagcactag gcaaaagcag gatgcacac 291229DNAArtificialPrimer 12gtgtgcatcc tgcttttgcc tagtgctag 291329DNAArtificialPrimer 13ctgccttagg cttaggtagg actgaaagg 291429DNAArtificialPrimer 14cctttcagtc ctacctaagc ctaaggcag 291533DNAArtificialPrimer 15ggactgaaag gctgtcctct caaaacccac cag 331633DNAArtificialPrimer 16ctggtgggtt ttgagaggac agcctttcag tcc 331736DNAArtificialPrimer 17caatgccatt gaaccaagct tctcccagaa ttcaag 361836DNAArtificialPrimer 18cttgaattct gggagaagct tggttcaatg gcattg 36191671PRTArtificialFVIII with human albumin replacing part of the B-domain 19Ala Thr Arg Arg Tyr Tyr Leu Gly Ala Val Glu Leu Ser Trp Asp Tyr 1 5 10 15 Met Gln Ser Asp Leu Gly Glu Leu Pro Val Asp Ala Arg Phe Pro Pro 20 25 30 Arg Val Pro Lys Ser Phe Pro Phe Asn Thr Ser Val Val Tyr Lys Lys 35 40 45 Thr Leu Phe Val Glu Phe Thr Asp His Leu Phe Asn Ile Ala Lys Pro 50 55 60 Arg Pro Pro Trp Met Gly Leu Leu Gly Pro Thr Ile Gln Ala Glu Val 65 70 75 80 Tyr Asp Thr Val Val Ile Thr Leu Lys Asn Met Ala Ser His Pro Val 85 90 95 Ser Leu His Ala Val Gly Val Ser Tyr Trp Lys Ala Ser Glu Gly Ala 100 105 110 Glu Tyr Asp Asp Gln Thr Ser Gln Arg Glu Lys Glu Asp Asp Lys Val 115 120 125 Phe Pro Gly Gly Ser His Thr Tyr Val Trp Gln Val Leu Lys Glu Asn 130 135 140 Gly Pro Met Ala Ser Asp Pro Leu Cys Leu Thr Tyr Ser Tyr Leu Ser 145 150 155 160 His Val Asp Leu Val Lys Asp Leu Asn Ser Gly Leu Ile Gly Ala Leu 165 170 175 Leu Val Cys Arg Glu Gly Ser Leu Ala Lys Glu Lys Thr Gln Thr Leu 180 185 190 His Lys Phe Ile Leu Leu Phe Ala Val Phe Asp Glu Gly Lys Ser Trp 195 200 205 His Ser Glu Thr Lys Asn Ser Leu Met Gln Asp Arg Asp Ala Ala Ser 210 215 220 Ala Arg Ala Trp Pro Lys Met His Thr Val Asn Gly Tyr Val Asn Arg 225 230 235 240 Ser Leu Pro Gly Leu Ile Gly Cys His Arg Lys Ser Val Tyr Trp His 245 250 255 Val Ile Gly Met Gly Thr Thr Pro Glu Val His Ser Ile Phe Leu Glu 260 265 270 Gly His Thr Phe Leu Val Arg Asn His Arg Gln Ala Ser Leu Glu Ile 275 280 285 Ser Pro Ile Thr Phe Leu Thr Ala Gln Thr Leu Leu Met Asp Leu Gly 290 295 300 Gln Phe Leu Leu Phe Cys His Ile Ser Ser His Gln His Asp Gly Met 305 310 315 320 Glu Ala Tyr Val Lys Val Asp Ser Cys Pro Glu Glu Pro Gln Leu Arg 325 330 335 Met Lys Asn Asn Glu Glu Ala Glu Asp Tyr Asp Asp Asp Leu Thr Asp 340 345 350 Ser Glu Met Asp Val Val Arg Phe Asp Asp Asp Asn Ser Pro Ser Phe 355 360 365 Ile Gln Ile Arg Ser Val Ala Lys Lys His Pro Lys Thr Trp Val His 370 375 380 Tyr Ile Ala Ala Glu Glu Glu Asp Trp Asp Tyr Ala Pro Leu Val Leu 385 390 395 400 Ala Pro Asp Asp Arg Ser Tyr Lys Ser Gln Tyr Leu Asn Asn Gly Pro 405 410 415 Gln Arg Ile Gly Arg Lys Tyr Lys Lys Val Arg Phe Met Ala Tyr Thr 420 425 430 Asp Glu Thr Phe Lys Thr Arg Glu Ala Ile Gln His Glu Ser Gly Ile 435 440 445 Leu Gly Pro Leu Leu Tyr Gly Glu Val Gly Asp Thr Leu Leu Ile Ile 450 455 460 Phe Lys Asn Gln Ala Ser Arg Pro Tyr Asn Ile Tyr Pro His Gly Ile 465 470 475 480 Thr Asp Val Arg Pro Leu Tyr Ser Arg Arg Leu Pro Lys Gly Val Lys 485 490 495 His Leu Lys Asp Phe Pro Ile Leu Pro Gly Glu Ile Phe Lys Tyr Lys 500 505 510 Trp Thr Val Thr Val Glu Asp Gly Pro Thr Lys Ser Asp Pro Arg Cys 515 520 525 Leu Thr Arg Tyr Tyr Ser Ser Phe Val Asn Met Glu Arg Asp Leu Ala 530 535

540 Ser Gly Leu Ile Gly Pro Leu Leu Ile Cys Tyr Lys Glu Ser Val Asp 545 550 555 560 Gln Arg Gly Asn Gln Ile Met Ser Asp Lys Arg Asn Val Ile Leu Phe 565 570 575 Ser Val Phe Asp Glu Asn Arg Ser Trp Tyr Leu Thr Glu Asn Ile Gln 580 585 590 Arg Phe Leu Pro Asn Pro Ala Gly Val Gln Leu Glu Asp Pro Glu Phe 595 600 605 Gln Ala Ser Asn Ile Met His Ser Ile Asn Gly Tyr Val Phe Asp Ser 610 615 620 Leu Gln Leu Ser Val Cys Leu His Glu Val Ala Tyr Trp Tyr Ile Leu 625 630 635 640 Ser Ile Gly Ala Gln Thr Asp Phe Leu Ser Val Phe Phe Ser Gly Tyr 645 650 655 Thr Phe Lys His Lys Met Val Tyr Glu Asp Thr Leu Thr Leu Phe Pro 660 665 670 Phe Ser Gly Glu Thr Val Phe Met Ser Met Glu Asn Pro Gly Leu Trp 675 680 685 Ile Leu Gly Cys His Asn Ser Asp Phe Arg Asn Arg Gly Met Thr Ala 690 695 700 Leu Leu Lys Val Ser Ser Cys Asp Lys Asn Thr Gly Asp Tyr Tyr Glu 705 710 715 720 Asp Ser Tyr Glu Asp Ile Ser Ala Tyr Leu Leu Ser Lys Asn Asn Ala 725 730 735 Ile Glu Pro Ser Phe Ser Gln Asn Ser Arg His Pro Ser Thr Arg Gln 740 745 750 Lys Gln Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu 755 760 765 Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu 770 775 780 Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser 785 790 795 800 His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 805 810 815 Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr 820 825 830 Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn 835 840 845 Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro 850 855 860 Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln 865 870 875 880 Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Val Thr Lys Asn Gln Val 885 890 895 Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val 900 905 910 Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro 915 920 925 Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr 930 935 940 Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val 945 950 955 960 Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Ser 965 970 975 Gln Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr 980 985 990 Thr Leu Gln Ser Asp Gln Glu Glu Ile Asp Tyr Asp Asp Thr Ile Ser 995 1000 1005 Val Glu Met Lys Lys Glu Asp Phe Asp Ile Tyr Asp Glu Asp Glu 1010 1015 1020 Asn Gln Ser Pro Arg Ser Phe Gln Lys Lys Thr Arg His Tyr Phe 1025 1030 1035 Ile Ala Ala Val Glu Arg Leu Trp Asp Tyr Gly Met Ser Ser Ser 1040 1045 1050 Pro His Val Leu Arg Asn Arg Ala Gln Ser Gly Ser Val Pro Gln 1055 1060 1065 Phe Lys Lys Val Val Phe Gln Glu Phe Thr Asp Gly Ser Phe Thr 1070 1075 1080 Gln Pro Leu Tyr Arg Gly Glu Leu Asn Glu His Leu Gly Leu Leu 1085 1090 1095 Gly Pro Tyr Ile Arg Ala Glu Val Glu Asp Asn Ile Met Val Thr 1100 1105 1110 Phe Arg Asn Gln Ala Ser Arg Pro Tyr Ser Phe Tyr Ser Ser Leu 1115 1120 1125 Ile Ser Tyr Glu Glu Asp Gln Arg Gln Gly Ala Glu Pro Arg Lys 1130 1135 1140 Asn Phe Val Lys Pro Asn Glu Thr Lys Thr Tyr Phe Trp Lys Val 1145 1150 1155 Gln His His Met Ala Pro Thr Lys Asp Glu Phe Asp Cys Lys Ala 1160 1165 1170 Trp Ala Tyr Phe Ser Asp Val Asp Leu Glu Lys Asp Val His Ser 1175 1180 1185 Gly Leu Ile Gly Pro Leu Leu Val Cys His Thr Asn Thr Leu Asn 1190 1195 1200 Pro Ala His Gly Arg Gln Val Thr Val Gln Glu Phe Ala Leu Phe 1205 1210 1215 Phe Thr Ile Phe Asp Glu Thr Lys Ser Trp Tyr Phe Thr Glu Asn 1220 1225 1230 Met Glu Arg Asn Cys Arg Ala Pro Cys Asn Ile Gln Met Glu Asp 1235 1240 1245 Pro Thr Phe Lys Glu Asn Tyr Arg Phe His Ala Ile Asn Gly Tyr 1250 1255 1260 Ile Met Asp Thr Leu Pro Gly Leu Val Met Ala Gln Asp Gln Arg 1265 1270 1275 Ile Arg Trp Tyr Leu Leu Ser Met Gly Ser Asn Glu Asn Ile His 1280 1285 1290 Ser Ile His Phe Ser Gly His Val Phe Thr Val Arg Lys Lys Glu 1295 1300 1305 Glu Tyr Lys Met Ala Leu Tyr Asn Leu Tyr Pro Gly Val Phe Glu 1310 1315 1320 Thr Val Glu Met Leu Pro Ser Lys Ala Gly Ile Trp Arg Val Glu 1325 1330 1335 Cys Leu Ile Gly Glu His Leu His Ala Gly Met Ser Thr Leu Phe 1340 1345 1350 Leu Val Tyr Ser Asn Lys Cys Gln Thr Pro Leu Gly Met Ala Ser 1355 1360 1365 Gly His Ile Arg Asp Phe Gln Ile Thr Ala Ser Gly Gln Tyr Gly 1370 1375 1380 Gln Trp Ala Pro Lys Leu Ala Arg Leu His Tyr Ser Gly Ser Ile 1385 1390 1395 Asn Ala Trp Ser Thr Lys Glu Pro Phe Ser Trp Ile Lys Val Asp 1400 1405 1410 Leu Leu Ala Pro Met Ile Ile His Gly Ile Lys Thr Gln Gly Ala 1415 1420 1425 Arg Gln Lys Phe Ser Ser Leu Tyr Ile Ser Gln Phe Ile Ile Met 1430 1435 1440 Tyr Ser Leu Asp Gly Lys Lys Trp Gln Thr Tyr Arg Gly Asn Ser 1445 1450 1455 Thr Gly Thr Leu Met Val Phe Phe Gly Asn Val Asp Ser Ser Gly 1460 1465 1470 Ile Lys His Asn Ile Phe Asn Pro Pro Ile Ile Ala Arg Tyr Ile 1475 1480 1485 Arg Leu His Pro Thr His Tyr Ser Ile Arg Ser Thr Leu Arg Met 1490 1495 1500 Glu Leu Met Gly Cys Asp Leu Asn Ser Cys Ser Met Pro Leu Gly 1505 1510 1515 Met Glu Ser Lys Ala Ile Ser Asp Ala Gln Ile Thr Ala Ser Ser 1520 1525 1530 Tyr Phe Thr Asn Met Phe Ala Thr Trp Ser Pro Ser Lys Ala Arg 1535 1540 1545 Leu His Leu Gln Gly Arg Ser Asn Ala Trp Arg Pro Gln Val Asn 1550 1555 1560 Asn Pro Lys Glu Trp Leu Gln Val Asp Phe Gln Lys Thr Met Lys 1565 1570 1575 Val Thr Gly Val Thr Thr Gln Gly Val Lys Ser Leu Leu Thr Ser 1580 1585 1590 Met Tyr Val Lys Glu Phe Leu Ile Ser Ser Ser Gln Asp Gly His 1595 1600 1605 Gln Trp Thr Leu Phe Phe Gln Asn Gly Lys Val Lys Val Phe Gln 1610 1615 1620 Gly Asn Gln Asp Ser Phe Thr Pro Val Val Asn Ser Leu Asp Pro 1625 1630 1635 Pro Leu Leu Thr Arg Tyr Leu Arg Ile His Pro Gln Ser Trp Val 1640 1645 1650 His Gln Ile Ala Leu Arg Met Glu Val Leu Gly Cys Glu Ala Gln 1655 1660 1665 Asp Leu Tyr 1670 20240PRTArtificialAmino acid sequence of a human immunoglobulin G heavy chain region (240 amino acids 1-19 human IgG signal peptide, 20-35 human IgG hinge region and 36-240 human IgG heavy chain) 20Met Glu Phe Gly Leu Ser Trp Leu Phe Leu Val Ala Ile Leu Lys Gly 1 5 10 15 Val Gln Cys Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu 20 25 30 Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr 35 40 45 Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val 50 55 60 Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val 65 70 75 80 Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser 85 90 95 Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu 100 105 110 Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala 115 120 125 Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro 130 135 140 Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Val Thr Lys Asn Gln 145 150 155 160 Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala 165 170 175 Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr 180 185 190 Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu 195 200 205 Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser 210 215 220 Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser 225 230 235 240 21975PRTArtificialmature human FVIII heavy chain with partial B domain replacement by human immunoglobulin G heavy chain region 21Ala Thr Arg Arg Tyr Tyr Leu Gly Ala Val Glu Leu Ser Trp Asp Tyr 1 5 10 15 Met Gln Ser Asp Leu Gly Glu Leu Pro Val Asp Ala Arg Phe Pro Pro 20 25 30 Arg Val Pro Lys Ser Phe Pro Phe Asn Thr Ser Val Val Tyr Lys Lys 35 40 45 Thr Leu Phe Val Glu Phe Thr Asp His Leu Phe Asn Ile Ala Lys Pro 50 55 60 Arg Pro Pro Trp Met Gly Leu Leu Gly Pro Thr Ile Gln Ala Glu Val 65 70 75 80 Tyr Asp Thr Val Val Ile Thr Leu Lys Asn Met Ala Ser His Pro Val 85 90 95 Ser Leu His Ala Val Gly Val Ser Tyr Trp Lys Ala Ser Glu Gly Ala 100 105 110 Glu Tyr Asp Asp Gln Thr Ser Gln Arg Glu Lys Glu Asp Asp Lys Val 115 120 125 Phe Pro Gly Gly Ser His Thr Tyr Val Trp Gln Val Leu Lys Glu Asn 130 135 140 Gly Pro Met Ala Ser Asp Pro Leu Cys Leu Thr Tyr Ser Tyr Leu Ser 145 150 155 160 His Val Asp Leu Val Lys Asp Leu Asn Ser Gly Leu Ile Gly Ala Leu 165 170 175 Leu Val Cys Arg Glu Gly Ser Leu Ala Lys Glu Lys Thr Gln Thr Leu 180 185 190 His Lys Phe Ile Leu Leu Phe Ala Val Phe Asp Glu Gly Lys Ser Trp 195 200 205 His Ser Glu Thr Lys Asn Ser Leu Met Gln Asp Arg Asp Ala Ala Ser 210 215 220 Ala Arg Ala Trp Pro Lys Met His Thr Val Asn Gly Tyr Val Asn Arg 225 230 235 240 Ser Leu Pro Gly Leu Ile Gly Cys His Arg Lys Ser Val Tyr Trp His 245 250 255 Val Ile Gly Met Gly Thr Thr Pro Glu Val His Ser Ile Phe Leu Glu 260 265 270 Gly His Thr Phe Leu Val Arg Asn His Arg Gln Ala Ser Leu Glu Ile 275 280 285 Ser Pro Ile Thr Phe Leu Thr Ala Gln Thr Leu Leu Met Asp Leu Gly 290 295 300 Gln Phe Leu Leu Phe Cys His Ile Ser Ser His Gln His Asp Gly Met 305 310 315 320 Glu Ala Tyr Val Lys Val Asp Ser Cys Pro Glu Glu Pro Gln Leu Arg 325 330 335 Met Lys Asn Asn Glu Glu Ala Glu Asp Tyr Asp Asp Asp Leu Thr Asp 340 345 350 Ser Glu Met Asp Val Val Arg Phe Asp Asp Asp Asn Ser Pro Ser Phe 355 360 365 Ile Gln Ile Arg Ser Val Ala Lys Lys His Pro Lys Thr Trp Val His 370 375 380 Tyr Ile Ala Ala Glu Glu Glu Asp Trp Asp Tyr Ala Pro Leu Val Leu 385 390 395 400 Ala Pro Asp Asp Arg Ser Tyr Lys Ser Gln Tyr Leu Asn Asn Gly Pro 405 410 415 Gln Arg Ile Gly Arg Lys Tyr Lys Lys Val Arg Phe Met Ala Tyr Thr 420 425 430 Asp Glu Thr Phe Lys Thr Arg Glu Ala Ile Gln His Glu Ser Gly Ile 435 440 445 Leu Gly Pro Leu Leu Tyr Gly Glu Val Gly Asp Thr Leu Leu Ile Ile 450 455 460 Phe Lys Asn Gln Ala Ser Arg Pro Tyr Asn Ile Tyr Pro His Gly Ile 465 470 475 480 Thr Asp Val Arg Pro Leu Tyr Ser Arg Arg Leu Pro Lys Gly Val Lys 485 490 495 His Leu Lys Asp Phe Pro Ile Leu Pro Gly Glu Ile Phe Lys Tyr Lys 500 505 510 Trp Thr Val Thr Val Glu Asp Gly Pro Thr Lys Ser Asp Pro Arg Cys 515 520 525 Leu Thr Arg Tyr Tyr Ser Ser Phe Val Asn Met Glu Arg Asp Leu Ala 530 535 540 Ser Gly Leu Ile Gly Pro Leu Leu Ile Cys Tyr Lys Glu Ser Val Asp 545 550 555 560 Gln Arg Gly Asn Gln Ile Met Ser Asp Lys Arg Asn Val Ile Leu Phe 565 570 575 Ser Val Phe Asp Glu Asn Arg Ser Trp Tyr Leu Thr Glu Asn Ile Gln 580 585 590 Arg Phe Leu Pro Asn Pro Ala Gly Val Gln Leu Glu Asp Pro Glu Phe 595 600 605 Gln Ala Ser Asn Ile Met His Ser Ile Asn Gly Tyr Val Phe Asp Ser 610 615 620 Leu Gln Leu Ser Val Cys Leu His Glu Val Ala Tyr Trp Tyr Ile Leu 625 630 635 640 Ser Ile Gly Ala Gln Thr Asp Phe Leu Ser Val Phe Phe Ser Gly Tyr 645 650 655 Thr Phe Lys His Lys Met Val Tyr Glu Asp Thr Leu Thr Leu Phe Pro 660 665 670 Phe Ser Gly Glu Thr Val Phe Met Ser Met Glu Asn Pro Gly Leu Trp 675 680 685 Ile Leu Gly Cys His Asn Ser Asp Phe Arg Asn Arg Gly Met Thr Ala 690 695 700 Leu Leu Lys Val Ser Ser Cys Asp Lys Asn Thr Gly Asp Tyr Tyr Glu 705 710 715 720 Asp Ser Tyr Glu Asp Ile Ser Ala Tyr Leu Leu Ser Lys Asn Asn Ala 725 730 735 Ile Glu Pro Ser Phe Ser Gln Asn Ser Arg His Pro Ser Thr Arg Gln 740 745 750 Lys Gln Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu 755 760 765 Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu 770 775 780 Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser 785 790 795 800 His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu 805 810 815 Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr 820 825 830 Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn 835 840 845 Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro

Ala Pro 850 855 860 Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln 865 870 875 880 Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Val Thr Lys Asn Gln Val 885 890 895 Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val 900 905 910 Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro 915 920 925 Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr 930 935 940 Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val 945 950 955 960 Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser 965 970 975 22917PRTArtificialmature human FVIII light chain attached to human immunoglobulin G heavy chain region 22Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly 1 5 10 15 Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile 20 25 30 Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu 35 40 45 Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His 50 55 60 Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg 65 70 75 80 Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys 85 90 95 Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu 100 105 110 Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr 115 120 125 Thr Leu Pro Pro Ser Arg Glu Glu Val Thr Lys Asn Gln Val Ser Leu 130 135 140 Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp 145 150 155 160 Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val 165 170 175 Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp 180 185 190 Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His 195 200 205 Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Ser Gln Asn 210 215 220 Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr Thr Leu 225 230 235 240 Gln Ser Asp Gln Glu Glu Ile Asp Tyr Asp Asp Thr Ile Ser Val Glu 245 250 255 Met Lys Lys Glu Asp Phe Asp Ile Tyr Asp Glu Asp Glu Asn Gln Ser 260 265 270 Pro Arg Ser Phe Gln Lys Lys Thr Arg His Tyr Phe Ile Ala Ala Val 275 280 285 Glu Arg Leu Trp Asp Tyr Gly Met Ser Ser Ser Pro His Val Leu Arg 290 295 300 Asn Arg Ala Gln Ser Gly Ser Val Pro Gln Phe Lys Lys Val Val Phe 305 310 315 320 Gln Glu Phe Thr Asp Gly Ser Phe Thr Gln Pro Leu Tyr Arg Gly Glu 325 330 335 Leu Asn Glu His Leu Gly Leu Leu Gly Pro Tyr Ile Arg Ala Glu Val 340 345 350 Glu Asp Asn Ile Met Val Thr Phe Arg Asn Gln Ala Ser Arg Pro Tyr 355 360 365 Ser Phe Tyr Ser Ser Leu Ile Ser Tyr Glu Glu Asp Gln Arg Gln Gly 370 375 380 Ala Glu Pro Arg Lys Asn Phe Val Lys Pro Asn Glu Thr Lys Thr Tyr 385 390 395 400 Phe Trp Lys Val Gln His His Met Ala Pro Thr Lys Asp Glu Phe Asp 405 410 415 Cys Lys Ala Trp Ala Tyr Phe Ser Asp Val Asp Leu Glu Lys Asp Val 420 425 430 His Ser Gly Leu Ile Gly Pro Leu Leu Val Cys His Thr Asn Thr Leu 435 440 445 Asn Pro Ala His Gly Arg Gln Val Thr Val Gln Glu Phe Ala Leu Phe 450 455 460 Phe Thr Ile Phe Asp Glu Thr Lys Ser Trp Tyr Phe Thr Glu Asn Met 465 470 475 480 Glu Arg Asn Cys Arg Ala Pro Cys Asn Ile Gln Met Glu Asp Pro Thr 485 490 495 Phe Lys Glu Asn Tyr Arg Phe His Ala Ile Asn Gly Tyr Ile Met Asp 500 505 510 Thr Leu Pro Gly Leu Val Met Ala Gln Asp Gln Arg Ile Arg Trp Tyr 515 520 525 Leu Leu Ser Met Gly Ser Asn Glu Asn Ile His Ser Ile His Phe Ser 530 535 540 Gly His Val Phe Thr Val Arg Lys Lys Glu Glu Tyr Lys Met Ala Leu 545 550 555 560 Tyr Asn Leu Tyr Pro Gly Val Phe Glu Thr Val Glu Met Leu Pro Ser 565 570 575 Lys Ala Gly Ile Trp Arg Val Glu Cys Leu Ile Gly Glu His Leu His 580 585 590 Ala Gly Met Ser Thr Leu Phe Leu Val Tyr Ser Asn Lys Cys Gln Thr 595 600 605 Pro Leu Gly Met Ala Ser Gly His Ile Arg Asp Phe Gln Ile Thr Ala 610 615 620 Ser Gly Gln Tyr Gly Gln Trp Ala Pro Lys Leu Ala Arg Leu His Tyr 625 630 635 640 Ser Gly Ser Ile Asn Ala Trp Ser Thr Lys Glu Pro Phe Ser Trp Ile 645 650 655 Lys Val Asp Leu Leu Ala Pro Met Ile Ile His Gly Ile Lys Thr Gln 660 665 670 Gly Ala Arg Gln Lys Phe Ser Ser Leu Tyr Ile Ser Gln Phe Ile Ile 675 680 685 Met Tyr Ser Leu Asp Gly Lys Lys Trp Gln Thr Tyr Arg Gly Asn Ser 690 695 700 Thr Gly Thr Leu Met Val Phe Phe Gly Asn Val Asp Ser Ser Gly Ile 705 710 715 720 Lys His Asn Ile Phe Asn Pro Pro Ile Ile Ala Arg Tyr Ile Arg Leu 725 730 735 His Pro Thr His Tyr Ser Ile Arg Ser Thr Leu Arg Met Glu Leu Met 740 745 750 Gly Cys Asp Leu Asn Ser Cys Ser Met Pro Leu Gly Met Glu Ser Lys 755 760 765 Ala Ile Ser Asp Ala Gln Ile Thr Ala Ser Ser Tyr Phe Thr Asn Met 770 775 780 Phe Ala Thr Trp Ser Pro Ser Lys Ala Arg Leu His Leu Gln Gly Arg 785 790 795 800 Ser Asn Ala Trp Arg Pro Gln Val Asn Asn Pro Lys Glu Trp Leu Gln 805 810 815 Val Asp Phe Gln Lys Thr Met Lys Val Thr Gly Val Thr Thr Gln Gly 820 825 830 Val Lys Ser Leu Leu Thr Ser Met Tyr Val Lys Glu Phe Leu Ile Ser 835 840 845 Ser Ser Gln Asp Gly His Gln Trp Thr Leu Phe Phe Gln Asn Gly Lys 850 855 860 Val Lys Val Phe Gln Gly Asn Gln Asp Ser Phe Thr Pro Val Val Asn 865 870 875 880 Ser Leu Asp Pro Pro Leu Leu Thr Arg Tyr Leu Arg Ile His Pro Gln 885 890 895 Ser Trp Val His Gln Ile Ala Leu Arg Met Glu Val Leu Gly Cys Glu 900 905 910 Ala Gln Asp Leu Tyr 915 2331PRTArtificialLinker 23Asn Asn Ala Ile Glu Pro Ser Phe Ser Gln Asn Ser Arg His Pro Ser 1 5 10 15 Thr Arg Gln Lys Gln Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 30 2432PRTArtificialLinker 24Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Pro 1 5 10 15 Ser Thr Arg Gln Lys Gln Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 30 2530PRTArtificialLinker 25Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Pro 1 5 10 15 Ser Thr Arg Gln Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 30 2628PRTArtificialLinker 26Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Pro 1 5 10 15 Ser Thr Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 2725PRTArtificialLinker 27Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Arg His Asp 1 5 10 15 Ala His Lys Ser Glu Val Ala His Arg 20 25 2823PRTArtificialLinker 28Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Asp Ala His 1 5 10 15 Lys Ser Glu Val Ala His Arg 20 2920PRTArtificialLinker 29Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Asp Ala His Lys Ser Glu 1 5 10 15 Val Ala His Arg 20 3032PRTArtificialLinker 30Asn Asn Ala Ile Glu Pro Arg Ser Phe Ser Gln Asn Ser Gly Gly Ser 1 5 10 15 Gly Gly Ser Gly Gly Ser Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 30 3127PRTArtificialLinker 31Asn Asn Ala Ile Glu Pro Arg Ser Val Ala Lys Lys His Pro Lys Thr 1 5 10 15 Trp Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 3227PRTArtificialLinker 32Asn Asn Ala Ile Glu Pro Arg Ser Phe Gln Lys Lys Thr Arg His Tyr 1 5 10 15 Phe Asp Ala His Lys Ser Glu Val Ala His Arg 20 25 3321PRTArtificialLinker 33Asn Asn Ala Ile Glu Pro Arg Ala Val Gly Gly Asp Ala His Lys Ser 1 5 10 15 Glu Val Ala His Arg 20 3436PRTArtificialLinker 34Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Arg Thr Glu Arg Leu 1 5 10 15 Cys Ser Gln Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr 20 25 30 Arg Thr Thr Leu 35 3536PRTArtificialLinker 35Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Arg Thr Glu Arg Leu 1 5 10 15 Ser Ser Gln Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr 20 25 30 Arg Thr Thr Leu 35 3634PRTArtificialLinker 36Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Thr Glu Arg Leu Cys Ser 1 5 10 15 Gln Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr 20 25 30 Thr Leu 3734PRTArtificialLinker 37Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Thr Glu Arg Leu Ser Ser 1 5 10 15 Gln Asn Pro Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr 20 25 30 Thr Leu 3831PRTArtificialLinker 38Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Leu Cys Ser Gln Asn Pro 1 5 10 15 Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr Thr Leu 20 25 30 3931PRTArtificialLinker 39Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Leu Ser Ser Gln Asn Pro 1 5 10 15 Pro Val Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr Thr Leu 20 25 30 4029PRTArtificialLinker 40Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Ser Gln Asn Pro Pro Val 1 5 10 15 Leu Lys Arg His Gln Arg Glu Ile Thr Arg Thr Thr Leu 20 25 4124PRTArtificialLinker 41Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Val Leu Lys Arg His Gln 1 5 10 15 Arg Glu Ile Thr Arg Thr Thr Leu 20 4221PRTArtificialLinker 42Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Arg His Gln Arg Glu Ile 1 5 10 15 Thr Arg Thr Thr Leu 20 4336PRTArtificialLinker 43Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Arg Thr Glu Arg Leu 1 5 10 15 Cys Ser Gln Asn Pro Pro Val Leu Lys Arg His Arg Arg Glu Ile Thr 20 25 30 Arg Thr Thr Leu 35 4429PRTArtificialLinker 44Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Ser Gln Asn Pro Pro Val 1 5 10 15 Leu Lys Arg His Arg Arg Glu Ile Thr Arg Thr Thr Leu 20 25 4521PRTArtificialLinker 45Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Arg His Arg Arg Glu Ile 1 5 10 15 Thr Arg Thr Thr Leu 20 4636PRTArtificialLinker 46Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Gly Ser Gly Gly Ser 1 5 10 15 Gly Gly Ser Gly Gly Ser Gly Gly Ser Arg His Arg Arg Glu Ile Thr 20 25 30 Arg Thr Thr Leu 35 4733PRTArtificialLinker 47Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Gly Ser Gly Gly Ser 1 5 10 15 Gly Gly Ser Gly Gly Ser Arg His Arg Arg Glu Ile Thr Arg Thr Thr 20 25 30 Leu 4827PRTArtificialLinker 48Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Gly Ser Gly Gly Ser 1 5 10 15 Arg His Arg Arg Glu Ile Thr Arg Thr Thr Leu 20 25 4927PRTArtificialLinker 49Val Ala Ala Ser Gln Ala Ala Leu Gly Leu Gly Gly Ser Gly Gly Ser 1 5 10 15 Arg His Arg Arg Glu Ile Thr Arg Thr Thr Leu 20 25

* * * * *

References


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed