AU2013202564B2 - Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life - Google Patents

Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life Download PDF

Info

Publication number
AU2013202564B2
AU2013202564B2 AU2013202564A AU2013202564A AU2013202564B2 AU 2013202564 B2 AU2013202564 B2 AU 2013202564B2 AU 2013202564 A AU2013202564 A AU 2013202564A AU 2013202564 A AU2013202564 A AU 2013202564A AU 2013202564 B2 AU2013202564 B2 AU 2013202564B2
Authority
AU
Australia
Prior art keywords
modified
fviii
vwf
complex
wild
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2013202564A
Other versions
AU2013202564A1 (en
Inventor
Ulrich Kronthaler
Wiegand Lang
Holger Lind
Hubert Metzner
Stefan Schulte
Thomas Weimer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CSL Behring GmbH Deutschland
Original Assignee
CSL Behring GmbH Deutschland
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2009262476A external-priority patent/AU2009262476C1/en
Application filed by CSL Behring GmbH Deutschland filed Critical CSL Behring GmbH Deutschland
Priority to AU2013202564A priority Critical patent/AU2013202564B2/en
Publication of AU2013202564A1 publication Critical patent/AU2013202564A1/en
Application granted granted Critical
Publication of AU2013202564B2 publication Critical patent/AU2013202564B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to modified nucleic acid sequences coding for coagulation factor VIII (FVIII) and for von Willebrand factor (VWF) as well as complexes thereof 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 FVIII sequences that result in improved expression yield. 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.

Description

5 Factor Vill, von Willebrand factor or complexes thereof with prolonged in vivo half life Field of the invention: 10 The present invention relates to modified nucleic acid sequences coding for coagulation factor Vill (FVIII) and for von Willebrand factor (VWF) as well as complexes thereof 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 15 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 FVIll sequences that result in improved expression yield. The present invention further relates to processes for the manufacture of such recombinant proteins and their derivatives. The invention also relates to a transfer 20 vector for use in human gene therapy, which comprises such modified nucleic acid sequences. Background of the invention: 25 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 Vill and IX, respectively. Another known bleeding disorder is von Willebrand's disease. 30 In plasma FVIll exists mostly as a noncovalent complex with VWF and its coagulant function is to accelerate factor IXa dependent conversion of factor X to Xa Due to the complex formation of FVIII and VWF it was assumed for a long time that FViII and VWF functions are two functions of the same molecule. Only in the seventies it became clear that FVIi and VWF are separate molecules that form a complex under physiologic conditions. In the eighties then the dissociation constant of about 0.2 nmol/L was determined (Leyte et al., Biochem J 1989, 257: 679-683) and the DNA sequence of both molecules was studied. 5 Classic hemophilia or hemophilia A is an inherited bleeding disorder. It results from a chromosome X-linked deficiency of blood coagulation FVIll, 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 10 tendency. Before treatment with FVIIl concentrates was introduced the mean life span for a person with severe hemophilia was less than 20 years. The use of concentrates of FVIII 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 15 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 FVIll concentrates have recently been developed which established a very high safety standard also for plasma derived FVIl 20 The cloning of the cDNA for FVIlI (Wood et al. 1984. Nature 312:330-336; Vehar et al. 1984. Nature 312:337-342) made it possible to express FVIII recombinantly leading to the development of several recombinant FVIll products, which were approved by the regulatory authorities between 1992 and 2003. The fact that the central B domain of the FVIll 25 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 FVIII. The mature FVIII molecule consists of 2332 amino acids which can be grouped into three 30 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 FVIll is shown in SEQ ID NO:15. During its secretion into plasma FVIII is processed intracellularly into a series of metal-ion linked heterodimers as single chain FVuII is cleaved at the B-A3 boundary and at different sites within the B-domain. This processing leads to heterogeneous heavy chain molecules consisting of the Al, 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 FVIII binds 5 with high affinity to von Willebrand Factor (VWF), which protects it from premature catabolism. The half-life of non-activated FVIII bound to VWF is about 12 hours in plasma. Coagulation FVIII 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 10 release of von Willebrand Factor and generating the activated FVIII heterotrimer which will form the tenase complex on phospholipid surfaces with FIXa and FX provided that Ca 2 is present. The heterotrimer consists of the Al 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 FVIII (FVllla) consists of an Al-subunit associated through the divalent metal ion linkage 15 to a thrombin-cleaved A3-C1-C2 light chain and a free A2 subunit relatively loosely associated with the Al and the A3 domain. To avoid excessive coagulation, FVIIIa must be inactivated soon after activation. The inactivation of FVIlla via activated Protein C (APC) by cleavage at Arg336 and Arg562 is 20 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 FVIIIa 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 FVlila in plasma, which is only 2.1 minutes (Saenko et al. 25 2002. Vox Sang. 83:89-96). In severe hemophilia A patients undergoing prophylactic treatment FVIII has to be administered intravenously (i.v.) about 3 times per week due to the short plasma half-life of FVIll of about 12 to 14 hours. Each Lv. administration is cumbersome, associated with pain 30 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.
It would thus be highly desirable to create a FVIlI with increased functional half-life allowing the manufacturing of pharmaceutical compositions containing FVill, which have to be administered less frequently. 5 Several attempts have been made to prolong the half-life of non-activated FVIll either by reducing its interaction with cellular receptors (WO 03/093313A2, WO 021060951A2), by covalently attaching polymers to FVIII (WO 94/15625, WO 97111957 and US 4970300), by encapsulation of FVIII (WO 99/55306), by introduction of novel metal binding sites (WO 97/03193), by covalently attaching the A2 domain to the A3 domain either by peptidic (WO 10 97140145 and WO 031087355) or disulfide linkage (WO 02/103024A2) or by covalently attaching the Al domain to the A2 domain (W02006/108590). Another approach to enhance the functional half-life of FVII or VWF is by PEGylation of FVIl (WO 2007/126808, WO 2006/053299, WO 2004/075923) or by PEGylation of VWF 15 (WO 2006/071801) which pegylated VWF by having an increased half-life would indirectly also enhance the half-life of FVIII present in plasma. As none of the above described approaches has yet resulted in an approved FVIll pharmaceutical and as introducing mutations into the FVIII wild-type sequence or 20 introducing chemical modifications entails at least a theoretical risk of creating immunogenic FVIII variants there is an ongoing need to develop modified coagulation factor Vill molecules which exhibit prolonged half-life. In view of a potential thrombogenic risk it is more desirable to prolong the half-life of the 25 non-activated form of FRll than that of FVIlla. VWF, which is missing, functionally defect or only available in reduced quantity in different forms of von Willebrand disease (VWD), is a multimeric adhesive glycoprotein present in the plasma of mammals, which has multiple physiological functions. During primary 30 hemostasis VWF acts as a mediator between specific receptors on the platelet surface and components of the extracellular matrix such as collagen. Moreover, VWF serves as a carrier and stabilizing protein for procoagulant FVIll. VWF is synthesized in endothelial cells and megakaryocytes as a 2813 amino acid precursor molecule. The amino acid sequence and the cDNA sequence of wild-type VWF are disclosed in Collins et al. 1987, Proc Nail. Acad. Sci. USA 84:4393-4397. The precursor polypeptide, pre-pro-VWF, consists of a 22-residue signal peptide, a 741- residue pro-peptide and the 2050-residue polypeptide found in mature plasma VWF (Fischer et al, FEBS Left. 351: 345-348, 1994), After cleavage of the signal peptide in the endoplasmatic reticulum a C-terminal disulfide 5 bridge is formed between two monomers of VWF. During further transport through the secretary pathway 12 N-linked and 10 O-linked carbohydrate side chains are added. More important, VWF dimers are multimerized via N-terminal disulfide bridges and the propeptide of 741 amino acids length is cleaved off by the enzyme PACE/furin in the late Golgi apparatus. The propeptide as well as the high-molecular-weight multimers of VWF 10 (VWF-HMWM) are stored in the Weibel-Pallade bodies of endothelial cells or in the a Granules of platelets. Once secreted into plasma the protease ADAMTS13 cleaves VWF within the Al domain of VWF. Plasma VWF therefore consists of a whole range of multimers ranging from single 15 dimers of 500 kDa to multimers consisting of up to more than 20 dimers of a molecular weight of over 10,000 kDa. The VWF-HMWM hereby having the strongest hemostatic activity, which can be measured in ristocetin cofactor activity (VWF:RCo). The higher the ratio of VWF:RCoNWF antigen, the higher the relative amount of high molecular weight multimers, 20 Defects in VWF are causal to von Willebrand disease (VWD), which is characterized by a more or less pronounced bleeding phenotype. AND type 3 is the most severe form in which VWF is completely missing, VWD type 1 relates to a quantitative loss of VWF and its phenotype can be very mild. VWD type 2 relates to qualitative defects of VWF and can be 25 as severe as VWD type 3. VWD type 2 has many sub forms some of them being associated with the loss or the decrease of high molecular weight multimers. Von VWD type 2a is characterized by a loss of both intermediate and large multimers. VWD type 2B is characterized by a loss of highest-molecular-weight multimers. VWD is the most frequent inherited bleeding disorder in humans and can be treated by 30 replacement therapy with concentrates containing VWF of plasmatic or recombinant origin. VWF can be prepared from human plasma as for example described in EP 05503991. EP 0784632 describes a method for isolating recombinant V'WF.
In plasma FVIII binds with high affinity to von VWF, which protects it from premature catabolism and thus, plays in addition to its role in primary hemostasis a crucial role to regulate plasma levels of FVIII and as a consequence is also a central factor to control secondary hemostasis. The half-life of non-activated FVIII bound to VWF is about 12 to 14 5 hours in plasma. In von Willebrand disease type 3, where no or almost no VWF is present, the half-life of FVIII is only about 6 hours, leading to symptoms of mild to moderate hemophilia A in such patients due to decreased concentrations of FVIII. The stabilizing effect of VWF on FVIII has also been used to aid recombinant expression of FVIII in CHO cells (Kaufman et at. 1989, Mol Cell Biol). 10 Until today the standard treatment of Hemophilia A and VWD involves frequent intravenous infusions of preparations of FVIII and VWF concentrates or of concentrates comprising a complex of FVIll and VWF derived from the plasmas of human donors or in case of FVIII that of pharmaceutical preparations based on recombinant FVIII. While these replacement 15 therapies are generally effective, e.g. in severe hemophilia A patients undergoing prophylactic treatment FVIll has to be administered intravenously (iv.) about 3 times per week due to the short plasma half life of FVIil of about 12 hours. Already above levels of 1% of the FVIll activity in non-hemophiliacs, e.g. by a raise of FVIII levels by 0,01 U/ml, severe hemophilia A is turned into moderate hemophilia A. In prophylactic therapy dosing 20 regimes are designed such that the trough levels of FVIll activity do not fall below levels of 2-3% of the FRll activity in non-hemophiliacs. Each i.v. administration is cumbersome, associated with pain and entails the risk of an infection especially as this is mostly done in home treatment by the patients themselves or by the parents of children being diagnosed for hemophilia A. In addition the frequent i.v. injections inevitably result in scar formation, 25 interfering with future infusions. As prophylactic treatment in severe hemophilia is started early in life, with children often being less than 2 years old, it is even more difficult to inject FVIII 3 times per week into the veins of such small patients. For a limited period, implantation of port systems may offer an alternative. Despite the fact that repeated infections may occur and ports can cause inconvenience during physical exercise, they are 30 nevertheless typically considered as favorable as compared to intravenous injections. The in vivo half-life of human VWF in the human circulation is approximately 12 to 20 hours. In prophylactic treatment of VWD e.g. of type 3 it would also be highly desirable to find ways to prolong the functional half-life of VWF.
-It Another approach to enhance the functional half-life of VWF is by PEGylation (WO 2006/071801) which pegylated VWF by having an increased half-life would indirectly also enhance the half-life of FVIll present in plasma. 5 However the chemical conjugation of PEG or other molecules to therapeutic proteins always entails the risk, that the specific activity is reduced due to shielding of important interaction sites with other proteins, chemical conjugation adds an additional step in the manufacture of such proteins decreasing final yields and making manufacture more expensive. Also the long term effects on human health are not known as currently known 10 PEGylated therapeutic proteins do not need to be administrated lifelong as it would be the case for a VWF to be administered in prophylaxis of von Willebrand disease or in for a FVIII to be administered in hemophilia A. It would thus be highly desirable to obtain a long-lived VWF which is not chemically 15 modified. In the prior art fusions of coagulation factors to albumin (WO 01/79271), alpha-fetoprotein (WO 20051024044) and immunoglobulin (WO 2004/101740) as half-life enhancing polypeptides have been described. These were taught to be attached to the carboxy- or the 20 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. 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 25 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 FVIll and VWF are mentioned. 30 A C-terminal fusion would not have been seriously considered by the man skilled in the art as the C2 domain of FVIll at the very C-terminal part of FVIll between amino acid 2303 and 2332 of FVIII comprises a platelet membrane binding site which is essential for FVIII function. This is why there are many amino acid mutations known in this region which lead to hemophilia A. It was thus surprising that a relatively large heterologous polypeptide like albumin can be fused to the C-terminal part of FVII without preventing F1Il function by preventing platelet binding. In addition, the C2 domain also contains a binding site for VWF. This site together with the amino acid sequence 1649-1689 is responsible for the high affinity binding of FVIII to VWF. Therefore, a man skilled in the art would also not have 5 expected that a FVIII with a C-terminal albumin fusion would retain its binding to VWF. It was surprisingly found that in contrast to the prediction by Ballance et al. an albumin fusion to the N-terminus of FVIII was not secreted into the culture medium. Therefore and because of the reasons detailed above it was now even more surprisingly found that a FVIII 10 fused at its C-terminal part to albumin is secreted into the culture medium and retains its biological function including binding to membranes of activated platelets and to VWF. It was also surprising to find that the modified FVIll of the invention shows an increase of in vivo recovery by about 20% compared to the wild type FVIll 15 A man skilled in the art would also not have considered fusing human albumin to the N- or the C-terminus of VWF. In an N-terminal fusion the albumin part would be cleaved off during propeptide processing. Or if the propeptide would be omitted the multimerization would not take place. As detailed above the C-terminus of VWF is essential for the initial 20 dimerization and secretion as shown by Schneppenheim et al. (Schneppenheim R. et al. 1996. Defective dimerization of VWF subunits due to a Cys to Arg mutation in VWD type lID. Proc NatI Acad Sci USA 93:3581-3586; Schneppenheim R. et aL 2001. Expression and characterization of VWF dimerization defects in different types of VWD. Blood 97:2059 2066.), Baronciani et a. (Baronciani Let al. 2000. Molecular characterization of a 25 multiethnic group of 21 patients with VAND type 3. Thromb, Haemost 84:536-540), Enayat et al. (Enayat MS et alt 2001. Aberrant dimerization of VWF as the result of mutations in the carboxy-terminal region: identification of 3 mutations in members of 3 different families with type 2A (phenotype lD) VWD, Blood 98:674-680) and Tjemberg et alt 2006. Homozygous C2362F VWF induces intracellular retention of mutant VWF resulting in 30 autosomal recessive severe VWD. Br J Haematol. 133:409-418). Therefore the man skilled in the art would not consider fusing a large protein like human albumin to the C- or N terminus of VWF as he would expect that normal dimerization or multimerization of VWF would be impaired. As the higher multimers of VWF are the ones most active in primary hemostasis the man skilled in the art would have looked for other ways to prolong the functional half-life of VVVF. It was now surprisingly found that fusing heterologous polypeptides such as albumin to the 5 C-terminal part of VWF, not only permits expression and secretion of VWF chimeric proteins from mammalian cells but also results in modified VWF molecules that retain significant VWF activity and form high molecular weight multimers. In addition, such modified VWF molecules exhibit prolonged in vivo half-life and/or improved in vivo recovery, 10 Description of the invention It is an objective of this invention to provide a modified FVIII or a modified VWF as well as complexes of modified FVlI with non-modified VWF, complexes of non-modified FVIII with 15 modified VWF and also complexes of modified FVIII with modified VWF with enhanced in vivo half-life. The term "modified FVIIW' or "modified VWF" in the sense of the invention means FVIll or VWF polypeptides which are fused to half-life enhancing polypeptides, encompassing also 20 natural alleles, variants, deletions and insertions of FVIll or VWF. It is another objective of this invention to provide a modified FVIII or a modified VWF as well as complexes of modified FVIII with non-modified VWF, complexes of non-modified FVIll with modified VWF and also complexes of modified FVIll with modified VWF with 25 improved in vivo recovery. Another objective of the invention is that this modified FVIII or modified VWF as well as complexes of modified FVIll with non-modified VWF, non-modified FVIll with modified VWF and also complexes of modified FVIll with modified VWF can be expressed by mammalian 30 cells and retain their respective biological activities. In summary, surprisingly the modified FVIll or modified VWF as well as complexes of modified FVlIl with non-modified VWF, complexes of non-modified FVIll with modified VWF 10 and also complexes of modified FVIII with modified VWF of the invention have retained biological activity, increased in vivo half-life and in vivo recovery. An additional potential benefit of those embodiments of the present invention in which the 5 FVIII is modified and in which the A2 domain remains only non 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 as compared to FVIII variants which lead to a stabilization of the activated form of FVIII. 10 The modified FVIII or modified VWF as well as complexes of modified FVIII with non modified VWF, complexes of non-modified FVIII with modified VWF and also complexes of modified FVIII with modified VWF molecules of the invention can be generated by fusing a half-life enhancing protein (HLEP) moiety to the C-terminal part of FVIII or to the 15 C-terminal part of VWF. 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 20 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. The present invention therefore relates to a modified FVIII or modified VWF as well as 25 complexes of modified FVIII with non-modified VWF, complexes of non-modified FVIII with modified VWF and also complexes of modified FVIII with modified VWF having at the C-terminal part of the modified FVIII and/or VWF a fusion to a HLEP, characterized in that the modified FVIII or modified VWF as well as the complex of modified FVIII with non-modified VWF, the complex of non-modified FVIII with modified VWF or the complex 30 of modified FVIII with modified VWF has prolonged functional half-life compared to the functional half-life of the wild-type FVIII or wild-type VWF or the complex of wild-type VWF and wild-type FVIII. In particular, the inventor relates to a modified factor VIII (FVIII), or a complex comprising 35 modified FVIII and non-modified von Willebrand factor (VWF) or a complex comprising modified FVIII and modified VWF, wherein the modified FVIII is created by fusing the C- 10a terminus of the primary translation polypeptide of FVIII, or a variant thereof, to the N terminal part of a half life enhancing polypeptide (HLEP) and the modified VWF when present in the complex is created by fusing the C-terminus of the primary translation polypeptide of VWF or variant thereof to the N-terminal part of a HLEP wherein the HLEP 5 is albumin or variants or fragments thereof, or an immunoglobulin constant region or variants or fragments thereof.
The present invention also relates to C-terminal fusions to more than one HLEP wherein the HLEP, which is fused several times, may be the same HLEP or may be a combination of different HLEPs. 5 The present invention also relates to a modified FVIll having at the C-terminal part a fusion to a HLEP, characterized in that the modified FVII or modified VWF or the complex of modified FVIII with non-modified VWF, the complex of non-modified FVIl with modified VWF or the complex of modified FVIll with modified VWF has improved in vivo recovery compared to the in vivo recovery of the wild-type FVIll or wild-type VWF or the complex of 10 wild-type VWF and wild-type FVIll Another embodiment of the invention are modified FVII polypeptides having at the C terminal part a fusion to a HLEP, characterized in that the modified FIll is secreted into a fermentation medium at a higher yield as a wild-type FVIll. 15 Another aspect of the invention are polynucleotides or combinations of polynucleotides encoding the modified FVIlI and/or the modified VWF. The invention further relates to plasmids or vectors comprising a polynucleotide described 20 herein, to host cells comprising a polynucleotide or a plasmid or vector described herein. Another aspect of the invention is a method of producing a modified FVIII or a modified VWF or a complex of modified FVIll with non-modified VWF, a complex of non-modified FVIII with modified VWF or a complex of modified FVIII with modified VWF, comprising: 25 (a) culturing host cells of the invention under conditions such that the modified coagulation factor is expressed; and (b) optionally recovering the modified coagulation factor from the host cells or from the culture medium. 30 The invention further pertains to pharmaceutical compositions comprising a modified FVIII or a modified VWF or a complex of modified FVill with non-modified VWF or a complex of non-modified FVIII with modified VWF or a complex of modified FVIll with modified VWF, a polynucleotide, or a plasmid or vector described herein.
- IZ Yet another aspect of the invention is the use of a modified FlIll or a modified VWF or a complex of modified FVIll with non-modified VWF or a complex of non-modified FVll with modified VWF or a complex of modified FVIII with modified VWF, one or more polynucleotides, or one or more plasmids or vectors, or of host cells according to this 5 invention for the manufacture of a medicament for the treatment or prevention of a blood coagulation disorder. Detailed description of the invention 10 The invention pertains to a complex comprising FVIII and von VWF or one of its individual polypeptidic components wherein at least one polypeptidic component of said complex is fused at the C-terminal part of its primary translation product to the N-terminal part of a half-life enhancing polypeptide (HLEP) 15 The invention also pertains to a modified FVIll or a modified VWF or a complex comprising modified FVIll and non-modified VWF or a complex comprising non-modified FVIll and modified VWF or a complex comprising modified FVIlI and modified VWF wherein the modified FVIlIl is fused at a C-terminal part of the primary translation polypeptide of FVIll to the N-terminal part of a HLEP or the modified VWF is fused at a C-terminal part of the 20 primary translation polypeptide of VWF to the N-terminal part acid of a HLEP. In preferred embodiments the invention pertains to a modified FVIR or a modified VWF or a complex comprising modified FVIII and non-modified VWF or a complex comprising non modified F/Ill and modified VWF or a complex comprising modified FVIll and modified 25 VWF, wherein a. the modified FVIl has a prolonged functional half-life compared to the functional half-life of wild-type FVIII or b. the modified VWF has a prolonged functional half-life compared to the 30 functional half-life of wild-type VWF or c. the complex comprising modified FVill and non-modified VWF has a prolonged functional half-life compared to the functional half-life of the corresponding complex comprising wild-type FVIll and wild-type VWF or -is_. d. the complex comprising non-modified FVIll and modified VWF has a prolonged functional half-life compared to the functional half-life of the corresponding complex comprising wild-type FVIII and wild-type VWF or e. the complex of modified FVIII with modified VWF has a prolonged functional 5 half-life compared to the functional half-life of the corresponding complex comprising wild-type FVIII and wild-type VWF. A preferred embodiment of the invention is a modified polypeptide or a complex comprising said modified polypeptide or a complex comprising said modified polypeptides as described 10 above, wherein the modified polypeptide has a functional half-life increased by at least 25% as compared to the functional half-life of the corresponding wild-type polypeptide or the complex comprising said modified polypeptide or a complex comprising said modified polypeptides has a functional half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF, 15 Another embodiment of the invention is a modified FVlil or a modified VWF or a complex comprising modified FVIll and non-modified VWF or a complex comprising non-modified FVIII and modified VWF or a complex comprising modified FVIII and modified VWF, wherein 20 a. the modified FVIII has a prolonged antigen half-life compared to the antigen half-life of wild-type FVIII or b. the modified VWF has a prolonged antigen half-life compared to the antigen half-life of wild-type VVVF or 25 c. the complex comprising modified FVIII and non-modified VWF has a prolonged antigen half-life compared to the antigen half-life of the corresponding complex comprising wild-type FVIll and wild-type VWF or d. the complex comprising non-modified FVIII and modified VWF has a prolonged antigen half-life compared to the antigen half-life of the corresponding complex 30 of wild-type FVIll and wild-type VWF or e. the complex comprising modified FVIll and modified VWF has a prolonged antigen half-life compared to the antigen half-life of the corresponding complex of wild-type FVIII and wild-type VWF.
-14 -w A preferred embodiment of the invention is a modified polypeptide or a complex comprising said modified polypeptide or a complex comprising said modified polypeptides as described above, wherein the modified polypeptide has an antigen half life increased by at least 25% as compared to the antigen half-life of the corresponding 5 wild-type polypeptide or the complex comprising said modified polypeptide or a complex comprising said modified polypeptides has an antigen half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF, 10 Still another embodiment of the invention is a modified FVIII or a modified VWF or a complex comprising modified F/ll and non-modified VWF or a complex comprising non-modified FVIII and modified VWF or a complex comprising modified FVII and modified VVVF, wherein 15 a. the modified FVIl has an increased in vivo recovery compared to the in vivo recovery of wild-type FVII or b, the modified VWF has an increased in vivo recovery compared to the in vivo recovery of wild-type VWF or c. the complex comprising modified F/Ill and non-modified VWF has an increased in vivo recovery compared to the in vivo recovery of the corresponding complex comprising wild-type FVIll and wild-type VWF or d. the complex comprising non-modified FVIli and modified VWF has an increased in vivo recovery compared to the in vivo recovery of the corresponding complex comprising wild-type FVIll and wild-type VWF or 25 e. the complex comprising modified FVIll and modified VWF has an increased in vivo recovery compared to the in vivo recovery of the corresponding complex comprising wild-type FVIl! and wild-type VWF. Another preferred embodiment of the invention is a modified polypeptide or a complex 30 comprising said modified polypeptide or a complex comprising said modified polypeptides as described above, wherein the modified polypeptide has an in vivo recovery increased by at least 10% as compared to the in vivo recovery of the corresponding wild-type polypeptide or the complex comprising said modified polypeptide or a complex comprising said modified polypeptides has an in vivo recovery increased by at least 10% as compared to the corresponding complex of wild-type FVIII and wild-type VWF. Another preferred embodiment of the invention is 5 a) a modified polypeptide or a complex comprising said modified polypeptide or a complex comprising said modified polypeptides as described above, wherein at least one polypeptidic component of said complex is fused at the C-terminal amino acid of its primary translation product to the N-terminal part of a HLEP or 10 b) a modified polypeptide or a complex comprising said modified polypeptide or a complex comprising said modified polypeptides as described above, wherein at least one polypeptidic component of said complex is fused at the C-terminal part of its primary translation product to the N-terminal amino acid of a HLEP or c) a modified polypeptide or a complex comprising said modified polypeptide or a 15 complex comprising said modified polypeptides as described above, wherein at least one polypeptidic component of said complex is fused at the C-terminal amino acid of its primary translation product to the N-terminal amino acid of a HILEP. Another preferred embodiment of the invention is a modified polypeptide or a complex 20 comprising said modified polypeptide or a complex comprising said modified polypeptides as described above, wherein the modified polypeptide has at least 10% of the biological activity of wild-type polypeptide or the complex comprising the modified polypeptide or a complex comprising said modified polypeptides has at least 10% of the biological activity of the corresponding complex of wild-type FVIII and wild-type VWF. 25 Also comprised in the present invention is a method of preparing a modified FVII or a modified VWF having increased functional half-life, comprising fusing the N-terminal part of a half-life-enhancing polypeptide to a C-terminal part of the primary translation polypeptide of the FVIII or to a C-terminal part of the primary translation polypeptide of the VWF as well 30 as a method of preparing a complex comprising modified FVIII and non-modified VWF or a complex comprising non-modified FVIII and modified VWF or a complex comprising modified FVIll and modified VWF by mixing a modified FVIli prepared by the method described above with wild-type VWF or by mixing wild-type FVIll with a modified VWF prepared by the method described above or by mixing a modified FVIII and a modified VWF prepared by the method described above. Also encompassed in the invention is the use of 5 a. a modified FVIII as prepared by the method described above and wild-type VWF or b. a wild-type FVIII and a modified VWF prepared by the method described above or 10 c. a modified FVIII a as prepared by the method described above and a modified VWF as prepared by the method described above for the manufacture of a combined pharmaceutical preparation for simultaneous, separate or sequential use in the therapy of bleeding disorders, preferentially in the 15 therapy of hemophilia A and/or von Willebrand disease. The "functional half-life" according to the present invention is the half-life of the biological activity of the modified FVIIl or the modified VWF or a complex of modified FVIll with non modified VWF or a complex of the non-modified FVIll with modified VWF or a complex of 20 modified FVIll with modified VWF 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 modified FVIll or the modified VWF or the complex of modified FVIll with non-modified VWF or the complex of non-modified FVIll with modified VWF or the complex of modified FVIll with modified VWF has been administered. 25 The phrases "fusing" or "fused" refer to the addition of amino acids to the C-terminal part of FVIII and/or to the C-terminal part of VWF. When referring herein to a "fusion to the C terminal amino acid of FVIII" or to a "fusion to the C-terminal amino acid of VWF" this means a fusion exactly to the C-terminal amino acid of FVIll at amino acid 2332 of the 30 mature wild-type FVIll cDNA sequence or exactly to the C-terminal amino acid of VWF at amino acid 2050 of wild-type mature VWF, Mature FVIII or mature VWF meaning the respective polypeptide after cleavage of the propeptide. However the invention also encompasses a "fusion to the C-terminal part of FVIII" or a "fusion to the C-terminal part of VWF" in the sense of this invention may also include a fusion to a FVIll and/or VWF molecule respectively in which one or more amino acid position up to n amino acids from the C-terminal amino acid of FVIll and/or of VWF are deleted. The figure n is an integer that should not be greater than 5%, preferably not greater than 1% of the total number of amino acids of the FVIll and/or VWF. Usually, n is 20, preferably 15, more preferably 10, 5 still more preferably 5 or less (e.g. 1, 2, 3, 4 or 5). In one embodiment, the modified FVill has the following structure: N - FVIII - C -11- H, [formula 1] 10 wherein N is an N-tenrninal part of FVIII, LI is a chemical bond or a linker sequence H is a HLEP, and 15 C is a C-terminal part of FVIIl In another embodiment the modified VWF has the following structure: N - VWF - C -L1- H, [formula 2] 20 wherein N is an N-terminal part of VWF, LI is a chemical bond or a linker sequence H is a HLEP, and 25 C is a C-terminal part of VWF LI may be a chemical bond or a linker sequence consisting of one or more amino acids, e.g. of 1 to 20, 1 to 15, 1 to 10, 1 to 5 or I 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 30 corresponding position in the wild-type coagulation factor. Examples of suitable amino acids present in LI include Gly and Ser. Preferred HLEP sequences are described infra. Likewise encompassed by the invention are fusions to the exact "N-terminal amino acid" of the respective HLEP, or fusions to the "N-terminal part" of the respective HLEP, which includes N-terminal deletions of one or more amino acids of the HLEP. The modified FVIII or the modified VWF or the complex of the modified FVIII with the non 5 modified VWF, the complex of the non-modified F/Ill with the modified VWF or the complex of the modified FVII with modified VWF of the invention may comprise more than one HLEP sequence, e.g. two or three HLEP sequences. These multiple HLEP sequences may be fused to the C-terminal part of FVIII and/or to the C-terminal part of VWF in tandem, e.g. as successive repeats. 10 FVIl may be processed proteolytically at various stages. For example, as mentioned supra, during its secretion into plasma single chain FVI1 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. FVIII is activated via 15 proteolytic cleavage at amino acids Arg372 and Arg740 within the heavy chain and at Arg1689 in the light chain generating the activated FVIII heterotrimer consisting of the Al domain, the A2 domain, and the light chain (A3-Cl-C2), a 73 kDa fragment. Thus the active form of FVIII (FVIIla) consists of an Al-subunit associated through the divalent metal ion linkage to a thrombin-cleaved A3-CI-C2 light chain and a free A2 subunit relatively 20 loosely associated with the Al and the A3 domain. Accordingly, the present invention encompasses also modified FVIII that is not present as a single chain polypeptide but consists of several polypeptides (e.g. one or two or three) that are associated with each other via non-covalent linkages. 25 Preferably N - FVIll - C comprises the full length sequence of FVIll. Also encompassed are N-terminal, C-terminal or internal deletions of FVIII as long as the biological activity of FVIl! is retained. The biological activity is retained in the sense of the invention if the FVIll with deletions retains at least 10%, preferably at least 25%, more preferably at least 50%, 30 most preferably at least 75% of the biological activity of wild-type FVIII. The biological activity of FVIl1 can be determined by the artisan as described below. A suitable test to determine the biological activity of FVIll is for example the one stage or the two stage coagulation assay (Rizza et a. 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. The cDNA sequence and the amino acid sequence of the mature wild-type form of human 5 blood coagulation FVIll are shown in SEQ ID NO:14 and SEQ ID NO:15, 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:15 does not exclude that in 10 the modified homologue one or more amino acids at positions 1 through 2332 of SEQ ID NO:15 are missing. The terms "blood coagulation Factor Vill", "Factor Vill" and "FVill" are used interchangeably herein. "Blood coagulation Factor Vill" includes wild-type blood 15 coagulation FVIII as well as derivatives of wild-type blood coagulation FVIII having the procoagulant activity of wild-type blood coagulation FVIlI. Derivatives may have deletions, insertions and/or additions compared with the amino acid sequence of wild-type FVIIL The term FVII includes proteolytically processed forms of FVIll, e.g. the form before activation, comprising heavy chain and light chain. 20 The term "FVill" includes any FVIll variants or mutants having at least 25%, more preferably at least 50%, most preferably at least 75% of the biological activity of wild-type factor Vill. 25 As non-limiting examples, FVIFI molecules include FVIII mutants preventing or reducing APC cleavage (Amano 1998. Thromb. Hasmost. 79:557-563), FVIII mutants further stabilizing the A2 domain (WO 97/40145), FVIII mutants resulting in increased expression (Swaroop et a. 1997. JBC 272:24121-24124), FVIII mutants reducing its immunogenicity (Lollar 1999. Thromb. Haemost. 82:505-508), FVIII reconstituted from differently expressed 30 heavy and light chains (Oh et at 1999. Exp. Mol. Med. 31:95-100), FVIII mutants reducing binding to receptors leading to catabolism of FVIlI 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 - 20f 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 5 (summarized by Pipe 2004. Sem. Thromb. Hemost. 30:227-237). All of these FVIII mutants and variants are incorporated herein by reference in their entirety. VWF may be processed proteolytically at various stages. For example, as mentioned supra, the protease ADAMTS13 cleaves VWF within the A2 domain of VWF. Accordingly, 10 the present invention encompasses also modified VWF which has been cleaved proteolytically e.g. by ADAMTS1 3. Such cleavage would result in multimeric chains of VWF which comprise at their ends at least one or at most two monomers of VWF which have been cleaved by ADAMTS 13. 15 Preferably N - VWF - C comprises the full length sequence of VWF, Also encompassed are N-terminal, C-terminal or internal deletions of VWF as long as the biological activity of VWF is retained. The biological activity is retained in the sense of the invention if the VWF with deletions retains 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 20 activity of wild-type VWF can be determined by the artisan using methods for ristocetin co factor activity (Federici AB et al. 2004. Haematologica 89:77-85), binding of VWF to GP lba of the platelet glycoprotein complex lb-V-IX (Sucker et al. 2006. Clin Appi Thromb Hemost. 12:305-310), or a collagen binding assay (Kallas & Talpsep. 2001. Annals of Hematology 80:466-471). 25 "FVIII" and/or 'VWF" within the above definition also include natural allelic variations that may exist and occur from one individual to another. "FVIlIl" and/or "VWF' within the above definition further includes variants of FVII and or VWF. Such variants differ in one or more amino acid residues from the wild-type sequence. Examples of such differences may 30 include as conservative amino acid substitutions, i.e, substitutions within groups of amino acids with similar characteristics, e.g. (1) small amino acids, (2) acidic amino acids, (3) polar amino acids, (4) basic amino acids, (5) hydrophobic amino acids, and (6) aromatic amino acids. Examples of such conservative substitutions are shown in the following table.
Table 1: (1) Alanine Glycine (2) Aspartic acid Glutamic acid (3) Asparagine Glutamine Serine Threonine (4) Arginine Histidine Lysine E(6) Isoleucine Leucine Methionine Valine (6) Phenylalanine Tyrosine Tryptophane One or more HLEPs may fused to the C-terminal part of FVIll preferably as not to interfere 5 with the binding capabilities of FVIlII for example to VWF, platelets or FIX, One or more HLEPs may be fused to the C-terminal part of VWF preferably as not to interfere with the binding capabilities of VWF for example to FVIl1, platelets, heparin or collagen. 10 Once FWill is endogenously activated during coagulation in vivo, it may be no longer desirable to maintain the increased functional half-life of the now activated FVIII as this might lead to thrombotic complications what is already the case for a wild-type activated coagulation factor as FVIla (Aledort 2004. J Thromb Haemost 2:1700-1708) and what may 15 be 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 FVIII molecules, which after endogenous activation in vivo or after availability of a cofactor do have a functional half-life comparable to that of unmodified FVIll. This can by way of non-limiting example be achieved by introducing a cleavage site for example for a coagulation factor 20 between the C-terminal part of FVIll and the HLEP. With such FVIII-HLEP connecting sequences the activation of the FVIl! chimeric protein of the invention will lead to a concomitant complete separation of FVlla from the HLEP moiety. Accordingly, in one embodiment, the functional half-life of the endogenously activated modified FVIlI is substantially the same as that of the activated wild-type FVIl! (e.g. ±15%, preferably ±10%). 25 In yet another embodiment of the invention, however, one or more of the proteolytical cleavage sites, preferably the thrombin cleavage sites at Arg740 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 5 molecule. In another embodiment of the invention the FVII proteins of the invention may be expressed as two separate chains (see infra). 10 The modified FVIll 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. In another embodiment of the invention, the amino acids at or near the PACE/Furin 15 cleavage site (Arg1648) are mutated or deleted in order to prevent cleavage by PACE/Furin. This is thought to result in a one-chain FVIlIl/HLEP fusion molecule with improved half-life. In one embodiment of the invention, the modified FVIII of the invention exhibits an 20 increased functional half-life compared to the corresponding FVII1 form containing no integrated HLEP and/or to the wild-type form FVILt The functional half-life eg. can be determined in vivo in animal models of hemophilia A, like FVIll knockout mice, in which one would expect a longer lasting hemostatic effect as compared to wild-type FVIIl. The hemostatic effect could be tested for example by determining time to arrest of bleeding 25 after a tail clip. The functional half-life in one embodiment of the invention is the half-life of the biological activity of the FVIII once it has been administered to a mammal and is measured in vitro. The functional half-life of the modified FVIlI according to the invention is greater than that of 30 the FVIII lacking the modification as tested in the same species. The functional half-life is preferably increased by at least 10%, preferably 25%, more preferably by at least 50%, and even more preferably by at least 100% compared to the wild-type form of FVIll.
The functional half-life of a modified FVIII comprising a HLEP modification, can be determined by administering the respective modified FVIII (and in comparison wild-type FVIII) 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 5 modified coagulation factor in blood samples drawn at appropriate intervals after application. Suitable test methods are the activity tests described herein. The functional half-life according to another embodiment of the invention is the half-life of the biological function of the VWF once it has been administered to a mammal and is 10 measured in vitro. The functional half-life of the modified VWF according to the invention is greater than that of the VWF lacking the modification as tested in the same species. The functional half-life is increased by at least 10%, preferably increased by at least 25%, more preferably by at least 50%, and even more preferably by at least 100% compared to the VWF lacking the modification and/or to the wild-type form of VWF. 15 The functional half-life of a modified VWF comprising a HLEP modification, can be determined by administering the respective modified VWF (and in comparison that of the non-modified VWF) to rats, rabbits or other experimental animal species intravenously or subcutaneously and following the elimination of the biological activity of said modified or 20 respectively non-modified VWF in blood samples drawn at appropriate intervals after application. Suitable test methods are the activity tests described herein. As a surrogate marker for the half-life of biological activity also the levels of antigen of the modified or respectively wild-type FVIIl or the levels of antigen of the modified or 25 respectively wild-type VWF can be measured. Thus also encompassed by the invention are modified FVIII and/or VWF molecules having at the C-terminal part of FVIII and/or VWF a fusion to a HLEP, characterized in that the modified FVIII or the modified VWF or the modified VWF or the complex of modified FVIII with non-modified VWF, or the complex of the non-modified FVIll with modified VWF or the complex of modified FVIll with modified 30 VWF has a prolonged half-life of the FVII and/or VWF antigen compared to the half-life of the FVIII and/or VWF antigen lacking said insertion. The "half-life of the FVIII antigen" according to the present invention is the half-life of the antigen of the FVIll once it has been administered to a mammal and is measured in vitro. The "half-life of the \WF antigen" according to the present invention is the half-life of the antigen of the VWF 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 artisan and commercially available (e.g. Dade Behring, Instrumentation Laboratory, Abbott Laboratories, Diagnostica Stago). Functional and antigen half-lives can be calculated using 5 the time points of the beta phase of elimination according to the formula t 1
,
2 = 1n2 / k, whereas k is the slope of the regression line. In another embodiment, the functional half-life of the endogenously activated modified FVIII is prolonged compared to that of the activated wild-type FVill. The increase may be more 10 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 FVIII molecules may be beneficial in situations were only very low levels of FVIII are available that therefore are not thrombogenic. Such situations may occur e.g. upon gene therapy treatment where often 15 only low expression rates can be achieved. Therefore, such stabilized FVIII molecules might be beneficial in e.g. gene therapy despite a thrombogenic risk connected to such FVIII molecules if administered as proteins in high or physiologic doses, In another embodiment of the invention, the modified FVIII of the invention exhibits an 20 improved in vivo recovery compared to the wild-type FVIll and the modified VWF of the invention exhibits an improved in vivo recovery compared to the wild-type VWF. The in vivo recovery can be determined in vivo for example in normal animals or in animal models of hemophilia A, like FVIII knockout mice, or in models of VWD, like VWF knockout mice in which one would expect an increased percentage of the modified FVIII or VWF of the 25 invention be found by antigen or activity assays in the circulation shortly (5 to 10 mir,) after iv. administration compared to the corresponding wild-type FVIl or wild-type VWF. 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 wild-type form FVIll or to 30 wild-type VWF. 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). It is another objective of the present invention to provide long-lived FVIll molecules, which 5 after proteolytic processing in vivo do have a functional half-life comparable to that of an unmodified FVIlIl This can be achieved by maintaining certain cleavage sites in the modified FVIIl leading to a proteolytic cleavage for example when in contact with activated coagulation factors, which separates the FVIII from the HLEP. Accordingly, in one embodiment, the functional half-life of the proteolytically processed modified FVilI is 10 substantially the same as that of the non-modified VWF lacking the modification, and/or it is substantially the same as that of the wild-type VWF (e.g. ±15%, preferably ±10%). Still another embodiment of the invention are modified FVIII polypeptides which are fused to a HLEP for example albumin at the C-terminal part of the FVIII molecule which do have 15 reduced binding to VWF or do not bind VWF at all. It is another objective of the present invention to provide long-lived VWF molecules, which after proteolytic processing in vivo do have functional properties comparable to that of an unmodified VWF. This can be achieved by maintaining or inserting certain cleavage sites in 20 the modified VWF (see infra) leading to a proteolytic cleavage for example when in contact with activated coagulation factors, which separates the VWF from the HLEP. Accordingly, in one embodiment, the functional half-life of the proteolytically processed modified VWF is substantially the same as that of the non-modified VWF lacking the modification, and/or it is substantially the same as that of the wild-type VWF (e.g. ±15%, preferably ±10%). 25 Another preferred embodiment of the invention is a coexpression of wild-type VWF and a modified VWF according to the invention resulting in VWF multimers comprising non modified as well as modified VWF monomers.
Linker sequences According to this invention, the therapeutic polypeptide moiety may be coupled to the HLEP moiety by a peptide linker. The linker should be non-immunogenic and may be a 5 non-cleavable or cleavable linker. Non-cleavable linkers may be comprised of alternating glycine and serine residues as exemplified in W02007/090584. 10 In another embodiment of the invention the peptidic linker between the FVIlI and/or the VWF moiety and the albumin moiety consists of peptide sequences, which serve as natural interdomain linkers in human proteins. Preferably such peptide sequences in their natural environment are located close to the protein surface and are accessible to the immune system so that one can assume a natural tolerance against this sequence. Examples are 15 given in W020071090584. Cleavable linkers should be flexible enough to allow cleavage by proteases, In a preferred embodiment the cleavage of the linker proceeds comparably fast as the activation of FVIll within the fusion protein, if the fusion protein is a modified FVIII. 20 The cleavable linker preferably comprises a sequence derived from a) the therapeutic polypeptide to be administered itself if it contains proteolytic cleavage sites that are proteolytically cleaved during activation of the therapeutic polypeptide, 25 b) a substrate polypeptide cleaved by a protease which is activated or formed by the involvement of the therapeutic polypeptide. c) a polypeptide involved in coagulation or fibrinolysis The linker region in a more preferred embodiment comprises a sequence of FVIII and/or 30 VWF, which should result in a decreased risk of neoantigenic properties of the expressed fusion protein. Also in case the therapeutic protein is FVll which needs to be proteolytically activated, the kinetics of the peptide linker cleavage will more closely reflect the coagulation-related activation kinetics of the zymogen.
In a preferred embodiment, the therapeutic polypeptide is FVIII zymogen and the HLEP is albumin. In this case the linker sequence is either derived from the sequences of the activation regions of FVIII, from the cleavage region of any substrate of FIX like FX or FVIl or from the cleavage region of any substrate polypeptide that is cleaved by a protease in 5 whose activation FIXa is involved. In a highly preferred embodiment the linker peptide is derived from FVIl! itself and comprises of sequences encompassing the thrombin cleavage sites at amino acid positions 372, 740 and 1689 of SEQ ID NO. 15, respectively. In another preferred embodiment the 10 linker peptide is derived from FX, FIX, FVII or FXI. The linker peptides are preferably cleavable by the proteases of the coagulation system, for example FlIa, FIXa, FXa, FXIa, FXIla and FVIIa. 15 Said linker sequences can also be used in the modified VWF of the invention. Exemplary combinations of therapeutic polypeptide, cleavable linker and HLEP include the constructs listed in W02007/090584 (for example in table 2 and figure 4) and W02007/144173 (for example in table 3a and 3b), but are not limited to these. 20 Half-life enhancing polypeptides (HLEPs) 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 25 fragments thereof region 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 30 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 FVIll or wild-type VWF. The HLEP portion of the proposed coagulation factor insertion constructs of the invention 5 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 FVIII or modified VWF. 10 In particular, the proposed FVIII HLEP or VWF HLEP fusion 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, 15 Albumin as HLEP 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 20 broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof). 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., 25 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:16 herein or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof. 30 In particular, the proposed FVIII fusion and/or VWF fusion 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 -, 219 fragments of said domains, for example domains 1 (amino acids 1-194 of SEQ ID NO:16), 2 (amino acids 195-387 of SEQ ID NO: 16), 3 (amino acids 388-585 of SEQ ID NO: 16), 1 + 2 (1-387 of SEQ ID NO: 16), 2 + 3 (195-585 of SEQ ID NO: 16) or I + 3 (amino acids 1-194 of SEQ ID NO: 16 + amino acids 388-585 of SEQ ID NO: 16). Each domain is itself 5 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 Lysl06 to Glul 19, Glu292 to VaI315 and Glu492 to Ala51 1. The albumin portion of the proposed FVIII fusion and/or VWF fusion constructs of the 10 invention may comprise at least one subdomain or domain of HA or conservative modifications thereof. Afamin, alpha-fetoprotein and vitamin D binding protein as HLEPs 15 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 20051024044). 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 at. 1994. J. Biol. Chem. 269:18149-18154) and 20 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 25 HLEPs. The term "variants" includes insertions, deletions and substitutions, either conservative or non-conservative as long as the desired function is still present. 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 30 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.
lmmunoglobulins as HLEPs Immunoglobulin G (IgG) constant regions (Fe) are known in the art to increase the half-life of therapeutic proteins (Dumont JA et al. 2006. BioDrugs 20:151-160). The IgG constant 5 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 10 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 200410087778 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 15 be quickly eliminated in vivo, Fc-IFN-B 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 20031076567), glucagon-like peptide-1 (WO 2005/000892), clotting factors (WO 20 2004/101740) and interleukin-10 (US 6,403,077), all with half-life enhancing properties. Polynucleotides The invention further relates to a polynucleotide encoding a modified coagulation factor, 25 preferably a modified FVIII and/or modified VWF 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 30 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, enzymaticatiy or metabolically modified forms +31 of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including, for example, simple and complex cells. The skilled person will understand that, due to the degeneracy of the genetic code, a given 5 polypeptide can be encoded by different polynucleotides. These "variants" are encompassed by this invention. Preferably, the polynucleotide of the invention is an isolated polynucleotide. The term "isolated" polynucleotide refers to a polynucleotide that is substantially free from other 10 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. 15 The invention further relates to a group of polynucleotides which together encode the modified FVIll and/or the modified VWF of the invention. A first polynucleotide in the group may encode the N-terminal part of the modified FVIII and/or the modified VWF, and a second polynucleotide may encode the C-terminal part of the modified FVIll and/or the 20 modified VWF. 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. 25 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 Vill, the coding sequences of the signal peptide, the Al 30 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. 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. Alternatively, the coding sequence of the FVIII signal peptide, the Al and A2 domains are 5 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. Both expression vectors are cotransfected into a suitable host cell, which will lead to the expression of the light and heavy chains of the FVIlI molecule of the invention and the formation of a functional protein. 10 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. 15 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. The host cells of the invention may be employed in a method of producing a modified 20 coagulation factor, preferably a modified FVIll molecule, which is part of this invention. The method comprises: (a) culturing host cells of the invention under conditions such that the desired insertion protein is expressed; and 25 (b) optionally recovering the desired insertion protein from the host cells or from the culture medium. It is preferred to purify the modified FVII and/or the modified VWF of the present invention to ; 80% purity, more preferably 2 95% purity, and particularly preferred is a pharmaceutically pure state that is greater than 99.9% pure with respect to contaminating 30 macromolecules, particularly other proteins and nucleic acids, and free of infectious and pyrogenic agents. Preferably, an isolated or purified modified FVIII and/or the modified VWF of the invention is substantially free of other, non-related polypeptides.
The various products of the invention are useful as medicaments. Accordingly, the invention relates to a pharmaceutical composition comprising a modified FVIII and/or the modified VWF as described herein, a polynucleotide of the invention, or a plasmid or vector of the invention. 5 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 FVIII and/or the modified VWF or the modified VWF or the complex of modified FVIll with non-modified VWF, or the complex of the non 10 modified FVIll with modified VWF or the complex of modified FVIII with modified VWF 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. 15 Expression of the proposed mutants 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 20 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, 25 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 30 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.
- a4 The cDNAs are then integrated into the genome of a suitable host cell line for expression of the modified FVIII and/or VWF 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 5 secretion into the cultivation medium. Examples on other post-translational modifications are tyrosine 0-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. 10 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. 15 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 20 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 25 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. Other types of selectable marker genes which can be used together with the cDNA of the 30 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-BI 1, 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 FVIII 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. If the above cell lines are grown in the presence of the cytotoxic dhfr-inhibitor methotrexate, 5 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. 10 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 15 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 20 Purification and Formulation The recombinant modified FVIII and/or the recombinant modified VWF protein, which accumulates in the medium of secreting cells of the above types, can be concentrated and 25 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. An example of such purification is the adsorption of the recombinant mutant protein to a 30 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 modified FVIII and/or modified VWF 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, hydroxyapatite chromatography steps, 5 multimodal chromatography steps, and size exclusion chromatography steps. 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 10 detergents, radiation in the visible or UV spectrum, gamma-radiation or nanofiltration. The modified polynucleotides (e.g. DNA) of this invention may also be integrated into a transfer vector for use in the human gene therapy. 15 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. 20 The modified FVIII and/or modified VWF 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. 25 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 a!, Taylor & Francis (2000) or "Handbook of Pharmaceutical Excipients", 3d edition, Kibbe et al., Pharmaceutical Press (2000)). In particular, the pharmaceutical composition comprising the polypeptide variant of 30 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.
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 5 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 10 encompass slow release systems. 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 15 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. The pharmaceutical composition of the invention may be administered alone or in 20 conjunction with other therapeutic agents. These agents may be incorporated as part of the same pharmaceutical. One example of such an agent is the combination of modified FVII with non-modified VWF or the combination of non-modified FVIII with modified WVF or the combination of modified FVIlI with modified VWF.
Figures Figure 1: Antigen and activity levels of wild-type FVIll and FVIII-C-terminal albumin fusion polypeptides 5 Figure 2: Comparison of human FVIII:Ag pharmacokinetics in VWF ko mice following i.v. injection of 100 U (FVIIl:Ag)/kg FVIII wildtype and FVIII-FP 1656 WVF (mean; n=4itimepoint) 10 Figure 3: VWF:RCoNWF:Ag ratios of cell culture supernatants containing wt rVWF (1570/1212), rVWF-FP (1572/1212) containing C-terminally linked albumin, or a mixed expression cell culture containing a mixture of wt rVWF (1570/1212) and rVWF-FP (1572/1212) transfected in a ratio of 5:1. Values of about 0,8 were obtained in every case that are close to I which is the theoretical ratio of NHP according to the unit definitions. 15 Figure 4: SDS-Agarose gel electrophoresis of wild-type rVWF (1570/1212) expressed in HEK cells (B) and rVWF-FP (1572/1212) expressed also in HEK cells (A). Bands were detected using either antibodies to VWF or to albumin (HSA), 20 Figure 5: Comparison of human rVF wildtype and rVWF-FP pharmacokinetics following i.v. injection of 100 IU VWF:Ag in rats (mean, n=2-3 /timepoint) Examples: Example 1: Generation of expression vectors for FVIII molecules with C-terminal albumin fusion 5 An expression plasmid based on plRESpuro3 (BD Biosciences) containing the full length FVIII cDNA sequence in its multiple cloning site (pF8-FL) was first used to create a B domain deleted FVIIl For that oligonucleotides F8-1 and F8-2 (SEQ ID NO 1 and 2) were used in a site-directed mutagenesis experiment according to standard protocols 10 (QuickChange XL Site Directed Mutagenesis Kit, Stratagene, La Jolla, CA, USA) using pF8-FL as a template to delete the B domain. In a second step a sequence encoding the amino acid sequence RRGR was introduced to connect R740 of the A2 domain with R1648 of the a3 domain. This was performed in another round of site-directed mutagenesis using primers F8-3 and F8-4 (SEQ ID NO 3 and 4). The resulting plasmid was called pF8-457. 15 A FVIII albumin fusion construct was generated stepwise. First, a PinAl cleavage site was introduced at the FVIll 3'terminus. For that a PCR fragment was generated using pF8-457 as template, using PCR primers We2827 and We2828 (SEQ ID NO 5 and 6), which was subsquently gel-purified, cut by restriction endonucleases BspEl and Notl and ligated into pF8-457 previously digested with BspE1 and Noti. The resulting plasmid (pF8-1433) was 20 then cut with enzymes PinAl and Notl and a fragment obtained by PCR on a human albumin cDNA containing plasmid using primers We 2829 and We 2830 (SEQ ID NO 7 and 8) and subsequently digested with enzymes PinAl and Notl was inserted. The resulting expression plasmid (pF8-1434) contained the coding sequences for a B domain deleted FVIII followed by a PinAl site to insert linkers (encoding the amino acid sequence ThrGly) 25 and the coding sequence for human albumin. The amino acid sequence encoded by pF8 1434 is depicted as SEQ ID NO 9. Linker sequences separating the FVIII and albumin moieties could then easily be inserted into the newly created PinAl site described above. The insertion of two linker sequences is 30 described in the following. In addition, based on pF8-1434, the TG linker might be deleted in completion and even deletions into the C-terminus of FVIll or the N-terminus of albumin can be performed using site directed mutagenesis. Insertion of a cleavable linker, derived from the FVl thrombin cleavage site: First a PCR fragment containing the sequence encoding the thrombin cleavage site at position 372 was 40 generated by PCR using primers We2979 and We298O (SEQ ID NO 10 and 11) and pFB 457 as template. This fragment was purified, digested with PinAl and ligated into PinAl digested pF8-1434. Sequencing verified insertion of correct orientation of the fragment, the resulting plasmid was called pFB-1563. 5 Insertion of a flexible glycine/serine linker: A PCR fragment containing the coding sequence for a 31 amino acid glycine/serine linker was amplified by PCR from pFVIl-937 described in W02007/090584 using primers We2991 and We2992 (SEQ ID NO 12 and 13). This fragment was then purified, digested by restriction endonuclease FinAl and ligated into 10 PinAl digested pF8-1434. Sequencing verified insertion of correct orientation of the fragment, the resulting plasmid was called pFB-1568. 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 15 Molecular Biology, Ausubel FM et aL (eds.) John Wiley & Sons, Inc.; http://wvw.currentprotocols.com/WileyCDAI) other constructs can be made by the artisan to replace albumin by another HLEP or insert any other linker into the described PinAl site. Transfer of the FVIll/albumin cDNA into suitable vectors like pIRESneo3 (Invitrogen) and pEE12.4 (Lonza) permitted expression and selection of clones expressing the respective 20 FVIll albumin fusion protein in CHO cells. Example 2: Transfection and expression of FVIII and VWF proteins Expression plasmids were grown up in E.coli TOPIO (Invitrogen, Carlsbad, CA, USA) and 25 purified using standard protocols (Qiagen, Hilden, Germany). HEK-293 (Invitrogen) cells were transfected using the Lipofectamine 2000 reagent (Invitrogen) and grown up in serum-free medium (Invitrogen 293 Express) in the presence of 4 pg/ml Puromycin and optionally 0.5 lU/mI VWF. CHO cells (CHO-S, Invitrogen; CHOK1SV, Lonza) were transfected using the Lipofectamine 2000 reagent (Invitrogen) and grown up in serum-free 30 medium (Invitrogen CD CHO, 6 mM glutamine for CHO-S and CD-CHO for CHOK1SV) in the presence of 500-1000 pg/mI Geneticin (CHO-S only). For FVIll expression optionally 0.5 lU/mi VWF were added. For vWF expression an expression plasmid encoding PACE/furin (pFu-797) as described in W02007/144173 was cotransfected. In another experiment two plasmids encoding VWF wild-type and VWF fused at the C-terminus to albumin were cotransfected with pFu-797 resulting in VWF multimeres with wild-type VWF monomers and albumin-fused VWF monomers (see figure 3). Transfected cell populations were spread through T-flasks into roller bottles or small scale fermenters from which supernatants were harvested for purification. 5 Table 2 lists HEK-293 expression data of the constructs described in example 1. Table 2: Construct Activity [U/mL] pF8-457 1.54 pF8-457 + 0.5 U/ml VWF 1.66 pF8-1434 1.59 pFS-1434 + 0.5 U/mI VWF 1 82 pF8-1563 + 0.5 U/mi VWF 204 pFS-1 568 + 0.5 U/mI VWF 1.21 10 Example 3: Increased expression rate of FVIII albumin fusion protein Figure 1 summarizes the results of an expression study of a FVIll albumin fusion protein in serum-free cell culture. HEK-293 cells were transfected in triplicate with pF8-1434 (FVIll C terminal albumin fusion) and pF8-457 (FVII wild-type), respectively, seeded into T80 flasks 15 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. The results demonstrated an expression enhancing effect of the albumin moiety when present as an integral part of the FVIlI molecule in cell culture. Consequently, the 20 productivity was clearly improved in the case of the fusion protein compared to wild-type FVIII (Figure 1), -42-, Example 4: Purification of FVIII proteins To the expression supernatant containing the FVIll molecule a sufficient amount of an immune affinity resin was added to bind the FVIll activity almost completely. The immune 5 affinity resin had been prepared by binding an appropriate anti-FIll 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 CaCl 2 and 50% ethylene glycol. The immune affinity chromatography (IAC) fractions containing FVIII:C activity were 10 pooled, dialyzed against formulation buffer (excipients: sodium chloride, sucrose, histidine, calcium chloride, and Tween 80), and concentrated. Samples were either stored frozen or freeze-dried using an appropriate freeze-drying cycle. Alternatively, the FVIll containing cell culture supernatant is concentrated/purified by a first ion exchange chromatography followed by further purification using immune affinity 15 chromatography (lAC). In this case the eluate of the ion exchange chromatography is loaded onto an lAC column using the above mentioned resin. Example 5: Analysis of FVIll activity and antigen 20 For activity determination of FVIIl:C in vitro either a clotting assay (e.g. Pathromtin SL reagent and FVIll 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. 25 FVIll antigen (FVIII:Ag) was determined by an ELISA whose performance is known to those skilled in the art. Briefly, microplates were incubated with 100 pL per well of the capture antibody (sheep anti-human FVIII 1gG, 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 30 buffer (Cedarlane) as well as serial dilutions of a FVIII preparation (CSL Behring; 200 - 2 mU/mL) in sample diluent buffer (volumes per well: 100 pL) were incubated for two hours at ambient temperature. After three wash steps with buffer B, 100 pL 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 pL 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 pl stop solution (Dade Behring, OSFA) prepared the samples for reading in a suitable microplate reader at 450 5 nrm wavelength. Concentrations of test samples were then calculated using the standard curve with the FVIII preparation as reference. Example 6: Assessment of Pharnacokinetics of FVIII-FP in VWF ko mice following a single Lv. injection 10 In order to compare the pharmacokinetics of FVI wildtype (DNA 457) and a C-terminal FVIII-FP (DNA 1656), both FVIII variants were administered intravenously to mice. A VWF ko mouse strain (Denis C. et al, Proc. Nati. Acad. Sci. USA, 1998, Vol 95, 9524-9529) was chosen because, amongst other functions, VWF serves as a carrier and stabilizing protein 15 for FVIII, thereby protecting FVIII from premature degradation, e.g. by proteases, and from premature elimination from circulation. For unmodified FVIII an undisturbed interaction with VWF is essential as exemplified by hemophilia A cases, caused by mutation in the C terminal region resulting in decreasing binding to VWF. In the case of modified FVIII such binding may, however, be even unwanted, in order to examine or achieve improved 20 pharmacokinetics. Accordingly both products were injected Lv. at a dose of 100 U (FVII:Ag)/kg as bolus to two groups of mice (Tab. 3). Blood was sampled retroorbitally at appropriate intervals starting at 5 minutes after application of the test substances and up to 24 hours. One blood sample / mouse was taken, processed to plasma and stored frozen at -20'C until analysis. Human FVIII:Ag concentration was quantified using an ELISA assay 25 specific for human FVIll or by a mixed ELISA specific for human albumin and FVIlI, respectively. The mean plasma concentration of the, for each timepoint pooled, samples was used for calculation of pharmacokinetic parameters. Half-live was calculated using the time points of the beta phase of elimination according to the formula t, 1 = 1n2 / k, whereas k is the slope of the regression line. The result is depicted in Figure 2. Surprisingly, FVIll-FP 30 1656 (t 92 = 3,06 h, between 5 and 960 min) had an about 3-4 times longer terminal half-life as compared to FVIII wildtype (ti = 0,8 h, between 5 and 240 min). In addition, the recovery of FVIII-FP 1656 was increased by about 20% as compared to wildtype FVIll (Tab. 4).
Table 3: Treatment groups for comparison of pharmacokinetics FV11! in VWF ko mice Treatment oose (FVIII:C) / volume / schedule I route N [00 U (FV1l1:Ag)/kg FVIlIl wildtype 10U(VI.9kg24 w0.2 mLJ20g b.w. / t=0 h /i.v.. 100 U(FVill:Ag)/kg FVIll-FP 1656 2 4 r0.2 mL/20g b.w. / t=0 h /i.v Table 4: Bioavailability (%) of FVIll wildtype and modified FVIlI, FVIII-FP 1656, upon iv. injection into VWF ko mice Treatment Bioavailability (%) FVIII wildtype 100 FVIll-FP 1656 120,4 5 Example 7: Generation of expression vectors for VWF wild-type and VWF albumin fusion proteins 10 An expression plasmid containing the full length VWF cDNA sequence in its multiple cloning site was generated first. For that the coding sequence of VWF was amplified by polymerase chain reaction (PCR) using primer set VWF+ and VWF- (SEQ ID NO. 17 and 18) under standard conditions known to those skilled in the art (and as described e.g. in Current Protocols in Molecular Biology, Ausubel FM et al. (eds.) John Wiley & Sons, Inc.; 15 htto://www.currentomis.com/WileyCDen from a plasmid containing VWF cDNA (as obtainable commercially, e.g. pMT2-VWF from ATCC, No. 67122). The resulting PCR fragment was digested by restriction endonuclease EcoRI and ligated into expression vector plRESpuro3 (BD Biosciences, Franklin Lakes, NJ, USA) which had been linearized by EcoRI. The resulting expression plasmid containing the wild-type cDNA of VWF 20 downstream of the CMV promoter was called pVWF-1570, A PCR fragment containing the coding sequence for a 31 amino acid glycine/serine linker and the human albumin cDNA was amplified from pFVIl-937 described in W02007/090584 using primers We2994 and Wel 335 (SEQ ID NO. 19 and 20). This PCR fragment was then digested by restriction endonuclease Noti and ligated into Noti digested pVWF-1570, The resulting plasmid containing the coding sequences of VWF wt, the linker sequence and human albumin was called pVWF-1574. 5 In order to achieve expression of a fusion protein several bases had to be deleted between VWF and the linker sequence. This was peformed by site directed mutagenesis according to standard protocols (QuickChange XL Site Directed Mutagenesis Kit, Stratagene, La Jolla, CA, USA) using oligonucleotides We2995 and We2996 (SEQ ID NO 21 and 22). The 10 resulting expression plasmid called pVWF-1 572 contained the coding sequences of VWF in frame with that of a 31 amino acid glycin/serine linker and human albumin. The amino acid sequence of the expressed rVWF-FP is outlined as SEQ ID No. 25, The amino acid sequence of the human VWF preproprotein is outlined as SEQ ID NO. 24. 15 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, ibid) other constructs can be made by the artisan for replacement of the albumin sequence by another HLEP sequence or the linker sequence by another linker sequence. 20 Example 8: Purification of VWF and VWF albumin fusion proteins Cell culture supernatants containing VWF wild-type (rVWF wt) or VWF albumin fusion protein (rVWF-FP) were sterile-filtered through a 0,2pm filter and dialysed against 25 equilibration buffer (EB; 10mM Tris-HCI, 10MM CaC 2 , pH 7.0). This material was then applied to a Heparin Fractogel column equilibrated with EB. The column was washed with EB and VWF proteins were equated with 500mM NaCl in EB. The elution peak was concentrated and dialysed against FB buffer (3g/L sodium chloride, 20 g/L glycine, 5.5 g/L trisodium citrate dihydrate, pH 7.0). Finally the material was sterile filtrated and frozen in 30 aliquots. If needed, further purification steps were applied comprising anion and/or cation exchange chromatography, HIC and SEC.
Example 9: Analysis of VWF activity and antigen Samples were analysed by immunoturbidimetric determination of VWF:Ag (OPABO3, Siemens Healthcare Diagnostics, Marburg, Germany) and for collagen binding (Technozym 5 VWF:CBA ELISA, Ref. 5450301 with calibrator set 5450310 and control set 5450312, Technoclone, Vienna, Austria) as described by the manufacturer. VWF:RCo testing was done using the BC VWF reagent of Siemens Healthcare Diagnostics, Marburg, Germany according to the manufacturers description. The 10 International Concentrate Standard was used as a primary standard preparation to calibrate an in-house standard preparation for day to day use. The ratios of VWF:RCo and VWF:Ag assays are calculated in order to compare this parameter for different constructs tested. As is shown in figure 3 the VWF:RCo/VWF:Ag 15 ratio was comparable for wt rVWF and the C-terminal rVWF-albumin fusion protein. For pharmacokinetic analyses VWF antigen was determined by an ELISA whose performance is known to those skilled in the art. Briefly, microplates were incubated with 100 pL per well of the capture antibody (rabbit anti human VWF-IgG, Dako A0082 [Dako, 20 Hamburg, Germany], diluted 1:2000 in buffer A [Sigma C3041, Sigma-Aldrich, Munich, Germany]) overnight at ambient temperature. After washing plates three times with buffer B (Sigma P3563), each well was incubated with 200 pL buffer C (Sigma P3688) for 1.5 hours at ambient temperature (blocking). After another three wash steps with buffer B, serial dilutions of the test sample in buffer B as well as serial dilutions of standard human plasma 25 (ORKL21; 20 - 0.2 mU/mL; Siemens Healthcare Diagnostics, Marburg, Germany) in buffer B (volumes per well: 100 pL) were incubated for 1.5 hours at ambient temperature. After three wash steps with buffer B, 100 pL of a 1:16000 dilution in buffer B of the detection antibody (rabbit anti human VWF-IgG, Dako P0226, peroxidase labelled) were added to each well and incubated for 1 hour at ambient temperature. After three wash steps with 30 buffer B, 100 pL of substrate solution (OUVF, Siemens Healthcare Diagnostics) were added per well and incubated for 30 minutes at ambient temperature in the dark. Addition of 100 pL undiluted stop dilution (OSFA, Siemens Healthcare Diagnostics) prepared the samples for reading in a suitable microplate reader at 450 nm wavelength. Concentrations -47: A of the test samples were then calculated using the standard curve with standard human plasma as reference. Example 10: Multimer analysis of VWF and VWF albumin fusion proteins 5 VWF Multimer analysis was performed by SDS-agarose gel electrophoresis as recently described (Tatewaki et al.,. Thromb. Res. 52: 23-32 (1988), and Metzner et al., Haemophilia 4 (Suppl. 3): 25-32 (1998)) with minor modifications. Briefly, after equilibration in running buffer ready to use 1 % agarose mini gels (BioRad) were used to standardize the 10 method as far as possible. Comparable amounts of VWF antigen were subjected to electrophoresis on the SDS-agarose gels. After Western blotting the VWF protein bands were detected using anti-VWF (DAKO, prod. No, 0854) or anti-albumin antibodies followed by alkaline phosphatase labelled anti-IgG antibodies (SIGMA, prod. No. 1305) and colour reaction quantified by densitometry. 15 Using wild-type rVWF (15701797) and rVWF-FP (15721797) it could be demonstrated by Western blotting and detection using anti-albumin or anti VWF antibodies that rVWF-FP forms a regular multimer distribution detected both by anti-albumin and anti-WVF antibodies (Figure 4). This confirms that although every subunit of the multimeric VVVF 20 contains albumin, a regular VWF multimer pattern is formed. The albumin moiety obviously does neither inhibit the N-terminal dimerization nor the C-terminal multimerization of the VWF molecules. Example 11: Assessment of pharmacokinetics of VWF and VWF albumin fusion 25 protein in rats following a single iv. injection rVWF-FP and rVWF wt were administered intravenously to a total of 4 CD rats each. The dose was 100 U (VWF:Ag)/kg body weight, at an injection volume of 4 mLlkg. Blood samples were drawn retroorbitally at appropriate intervals starting at 5 minutes after 30 application of the test substances, using an alternating sampling scheme, resulting in samples from 2 animals / timepoint (t=0, 5, 30, 90 min, 4h, Id for subset Nr. I and 0, 15 min, 1, 2, 8 h and 2 d for subset Nr. 2). The scheme was designed to minimize potential effects of blood sampling on the plasma concentration to be quantified. Blood was processed to plasma and stored deep frozen until analysis. The VWF:Ag level in plasma

Claims (28)

1. A modified factor VIII (FVIII), or a complex comprising modified FVIII and non modified von Willebrand factor (VWF) or a complex comprising modified FVIII and modified VWF, wherein the modified FVIII is created by fusing the C-terminus of the 5 primary translation polypeptide of FVIII, or a variant thereof, to the N-terminal part of a half life enhancing polypeptide (HLEP) and the modified VWF when present in the complex is created by fusing the C-terminus of the primary translation polypeptide of VWF or variant thereof to the N-terminal part of a HLEP wherein the HLEP is albumin or variants or fragments thereof, or an immunoglobulin constant region or variants or 10 fragments thereof.
2. A modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to claim 1 wherein the variant of the primary translation polypeptide of FVIII is a deletant and when present in the complex, the variant of the primary translation polypeptide of VWF is a deletant. 15
3. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to claim 2, wherein said deletant of the primary translation polypeptide of FVIII comprises N-terminal, C terminal and/or internal deletions, and when modified VWF is present in the complex, said deletant of the primary translation polypeptide of VWF comprises N-terminal, C 20 terminal and/or internal deletions.
4. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to any one of the previous claims, wherein the HLEP is fused to an amino acid of FVIII located at a distance from the C-terminal amino acid of up to 5% of the total length of the FVIII 25 primary translation polypeptide, based on the total number of amino acids in the FVIII primary translation polypeptide and when modified VWF is present in the complex, the HLEP is fused to an amino acid of VWF located at a distance from the C-terminal amino acid of up to 5% of the total length of the VWF primary translation polypeptide, based on the total number of amino acids in the VWF primary translation polypeptide. 30
5. The modified polypeptide or complex according to any one of the previous claims, wherein 1 to 20 amino acids at the C-terminus of the FVIII primary translation polypeptide have been deleted, and wherein the resulting C-terminal amino acid of the FVIII polypeptide is fused to the N-terminal amino acid of the HLEP and when modified VWF is 49 present in the complex 1 to 20 amino acids at the C-terminus of the VWF primary translation polypeptide have been deleted, and wherein the resulting C-terminal amino acid of the VWF polypeptide is fused to the N-terminal amino acid of the HLEP.
6. The modified polypeptide or complex according to any one of the previous claims, 5 wherein the C-terminal amino acid of the FVIII primary translation polypeptide has been deleted, and wherein the resulting C-terminal amino acid of the FVIII polypeptide is fused to the N-terminal amino acid of the HLEP and when modified VWF is present in the complex the C-terminal amino acid of the VWF primary translation polypeptide has been deleted, and wherein the resulting C-terminal amino acid of the VWF polypeptide is fused 10 to the N-terminal amino acid of the HLEP.
7. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to any one of the previous claims, wherein a. the modified FVIII has a prolonged functional half-life compared to the functional 15 half-life of wild-type FVIII, or b. the complex comprising modified FVIII and non-modified VWF has a prolonged functional half-life compared to the functional half-life of the corresponding complex comprising wild-type FVIII and wild-type VWF, or c. the complex comprising modified FVIII and modified VWF has a prolonged 20 functional half-life compared to the functional half-life of the corresponding complex comprising wild-type FVIII and wild-type VWF.
8. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to any one of the previous claims, wherein 25 a. the modified FVIII has a functional half-life increased by at least 25% as compared to the functional half-life of the corresponding wild-type polypeptide, or b. the complex comprising modified FVIII and non-modified VWF has a functional half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF, or 50 c. the complex comprising modified FViII and modified VWF has a functional half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF.
9. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF 5 or a complex comprising modified FVIII and modified VWF according to according to any one of the previous claims, wherein a. the modified FVIII has a prolonged antigen half-life compared to the antigen half life of wild-type FVIII, or b. the complex comprising modified FVIII and non-modified VWF has a prolonged 10 antigen half-life compared to the antigen half-life of the corresponding complex of wild-type FVIII and wild-type VWF, or c. the complex comprising modified FVIII and modified VWF has a prolonged antigen half-life compared to the antigen half-life of the corresponding complex of wild-type FVIII and wild-type VWF. 15
10. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to claim 9, wherein a. the modified FVIII has an antigen half-life increased by at least 25% as compared to the antigen half-life of the corresponding wild-type FVIII, or b. the complex comprising modified FVIII and non-modified VWF has an antigen 20 half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF, or c. the complex comprising modified FVIII and modified VWF has an antigen half-life increased by at least 25% as compared to the corresponding complex of wild-type FVIII and wild-type VWF. 25
11. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF according to any one of the previous claims, wherein a. the modified FVIII has an increased in vivo recovery compared to the in vivo recovery of wild-type FVIII, or 51 b. the complex comprising modified FVIII and non-modified VWF has an increased in vivo recovery compared to the in vivo recovery of the corresponding complex comprising wild-type FVIII and wild-type VWF, or c. the complex comprising modified FVIII and modified VWF has an increased in 5 vivo recovery compared to the in vivo recovery of the corresponding complex comprising wild-type FVIII and wild-type VWF.
12. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to claim 11, wherein 10 a. the modified FVIII has an in vivo recovery increased by at least 10% as compared to the in vivo recovery of the corresponding wild-type FVIII, or b. the complex comprising modified FVIII and non-modified VWF has an in vivo recovery increased by at least 10% as compared to the corresponding complex of wild-type FVIII and wild-type VWF, or 15 c. the complex comprising modified FVIII and modified VWF has an in vivo recovery increased by at least 10% as compared to the corresponding complex of wild-type FVIII and wild-type VWF.
13. The modified FVIII, or a complex comprising modified FVIII and non-modified VWF or a complex comprising modified FVIII and modified VWF, according to any one of the 20 previous claims, wherein a. the modified FVIII has at least 10% of the biological activity of wild-type FVIII, or b. the complex comprising modified FVIII and non-modified VWF has at least 10% of the biological activity of the corresponding complex of wild-type FVIII and wild type VWF, or 25 c. the complex comprising modified FVIII and modified VWF has at least 10% of the biological activity of the corresponding complex of wild-type FVIII and wild-type VWF.
14. An isolated polynucleotide, or a group of isolated polynucleotides, encoding a modified FVIII or a complex comprising said modified FVIII, according to any one of 30 claims 1 to 13. 52
15. A plasmid or vector comprising an isolated polynucleotide according to claim 14, or a group of plasmids or vectors, said group comprising the group of isolated polynucleotides according to claim 14.
16. A host cell comprising an isolated polynucleotide or a group of polynucleotides 5 according to claim 14, or a plasmid or vector or a group of plasmids or vectors according to claim 15.
17. A method of producing a modified FVIII, comprising: (a) culturing host cells according to claim 16 under conditions such that the modified FVIII is expressed; and 10 (b) optionally recovering the modified FVIII from the host cells or from the culture medium.
18. A pharmaceutical composition comprising a modified FVIII, or a complex comprising said modified FVIII according to any one of claims 1 to 13, a polynucleotide or group of polynucleotides according to claim 14, or a plasmid or vector or a group of 15 plasmids or vectors according to claim 15.
19. The use of a modified FVIII, or a complex comprising said modified FVIII, according to any one of claims 1 to 13, a polynucleotide or group of polynucleotides according to claim 14, or a plasmid or vector or group of plasmids or vectors according to claim 15, or a host cell according to claim 16 in the manufacture of a medicament for the treatment or 20 prevention of a blood coagulation disorder.
20. A method of treating or preventing a blood coagulation disorder, wherein a subject in need is administered an effective amount of a modified FVIII or a complex comprising said modified FVIII, according to any one of claims 1 to 13, a polynucleotide or group of polynucleotides according to claim 14, or a plasmid or vector or group of plasmids or 25 vectors according to claim 15, or a host cell according to claim 16, or medicament made according to claim 19.
21. The use according to claim 19 or method of claim 20, wherein the blood coagulation disorder is hemophilia A.
22. The use according to claim 19 or method of claim 20, wherein the blood 30 coagulation disorder is von Willebrand disease. 53
23. The use according to any one of claims 19, 21 or 22, or method of any one of claims 20 to 22, wherein the treatment comprises human gene therapy.
24. A method of preparing a modified FVIII having increased functional half-life, comprising fusing the N-terminal part of a half-life-enhancing polypeptide to a C-terminus 5 of the primary translation polypeptide of VWF, or a variant thereof.
25. The method of claim 24, wherein the variant of the primary translation polypeptide of FVIII is a deletant.
26. A method of preparing a complex comprising modified FVIII and non-modified VWF by mixing modified FVIII with a wild-type VWF prepared by the method of claim 24 or 25. 10
27. A method of preparing a complex comprising modified FVIII and modified VWF by mixing modified FVIII with a modified VWF, said modified FVIII being prepared by the method of claim 24 or 25.
28. A modified FVIII, or a complex comprising modified FVIII and non-modified VWF, or a complex comprising modified FVIII and modified VWF according to any one of claims 1 15 to 13, an isolated polynucleotide, or a group of isolated polynucleotides according to claim 14, a plasmid or vector according to claim 15, a host cell according to claim 16, a method according to any one of claims 17, or 20 to 27, a pharmaceutical composition according to claim 18, a use according to any one of claims 19 or 21 to 23, substantially as hereinbefore described. 20 CSL BEHRING GMBH WATERMARK PATENT AND TRADE MARKS ATTORNEYS P33964AU01
AU2013202564A 2008-06-24 2013-04-05 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life Ceased AU2013202564B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202564A AU2013202564B2 (en) 2008-06-24 2013-04-05 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP08011429.1 2008-06-24
AU2009262476A AU2009262476C1 (en) 2008-06-24 2009-06-24 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
AU2013202564A AU2013202564B2 (en) 2008-06-24 2013-04-05 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2009262476A Division AU2009262476C1 (en) 2008-06-24 2009-06-24 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life

Publications (2)

Publication Number Publication Date
AU2013202564A1 AU2013202564A1 (en) 2013-05-02
AU2013202564B2 true AU2013202564B2 (en) 2015-09-17

Family

ID=48407950

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013202564A Ceased AU2013202564B2 (en) 2008-06-24 2013-04-05 Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life

Country Status (1)

Country Link
AU (1) AU2013202564B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016025764A2 (en) 2014-08-13 2016-02-18 The Children's Hospital Of Philadelphia An improved expression cassette for packaging and expression of variant factor viii for the treatment of hemostasis disorders

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001079271A1 (en) * 2000-04-12 2001-10-25 Principia Pharmaceutical Corporation Albumin fusion proteins
WO2005024044A2 (en) * 2003-09-05 2005-03-17 Gtc Biotherapeutics, Inc. Method for the production of fusion proteins in transgenic mammal milk

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001079271A1 (en) * 2000-04-12 2001-10-25 Principia Pharmaceutical Corporation Albumin fusion proteins
WO2005024044A2 (en) * 2003-09-05 2005-03-17 Gtc Biotherapeutics, Inc. Method for the production of fusion proteins in transgenic mammal milk

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
WEIMER, T., et al., Thrombosis and Haemostasis, published online 12 March 2008, vol. 99, pages 659-667 *

Also Published As

Publication number Publication date
AU2013202564A1 (en) 2013-05-02

Similar Documents

Publication Publication Date Title
AU2009262476B2 (en) Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
AU2007338298B2 (en) Modified coagulation factors with prolonged in vivo half-life
US20180092966A1 (en) Covalent complex of von willebrand factor and factor viii, compositions, and uses relating thereto
US9458223B2 (en) Von willebrand factor variants having improved factor VIII binding affinity
EP1935430A1 (en) Modified coagulation factors with prolonged in vivo half-life
AU2013202564B2 (en) Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired