Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
VECTOR CO-EXPRESSING TRUNCATED VON WILLEBRAND FACTOR AND FACTOR VIII
Document Type and Number:
WIPO Patent Application WO/2020/205630
Kind Code:
A1
Abstract:
The present invention relates to a co-expression vector that comprises both truncated von Williebrand Factor (vWF)-Fc DNA construct and B-domain deleted FVIII DNA construct in the same vector. By co-expressing FVIII and truncated vWF-Fc with one expression vector in cells, the present invention controls the ratio of gene templates for FVIII and vWF proteins, provides a higher protein ratio of FVIII to vWF during cell expression, that results in a better occupancy of FVIII in vWF, and creates a higher yield (>1000 IU/ml) and more stable expression of FVIII protein molecules. In one preferred embodiment, the truncated vWF contains mutations of cysteines in the D'D3 domain of vWF to reduce multimer assembly of recombinant vWF during expression, and thus increasing recovery and quality of FVIII.

Inventors:
LI QI (US)
ZHANG HAILONG (US)
Application Number:
PCT/US2020/025475
Publication Date:
October 08, 2020
Filing Date:
March 27, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
BEIJING NEOLETIX BIOLOGICAL TECH CO LTD (CN)
International Classes:
C07K14/755
Domestic Patent References:
WO2014147386A12014-09-25
Foreign References:
US8759293B22014-06-24
US20150023959A12015-01-22
EP0253870B11993-03-31
Other References:
See also references of EP 3947439A4
Attorney, Agent or Firm:
KUNG, Viola T. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A vector comprising:

(a) a first polynucleotide sequence encoding a B-domain-deleted factor VIII protein (FVIII), operably linked to a first promoter, and

(b) a second polynucleotide sequence encoding a fusion protein comprising a truncated von Willebrand factor (vWF) and an immunoglobulin Fc fused to the C-terminal of the truncated vWF, operably linked to a second promoter,

wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 1, SEQ ID NO: 2, SEQ ID NO: 3, or SEQ ID NO: 4, or having 95% amino acid sequence identity thereof.

2. The vector according to Claim 1, wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 1, or having 95% amino acid sequence identity thereof, provided that the amino acid residues 336 and 379 are not cysteine.

3. The vector according to Claim 2, wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 1.

4. The vector according to Claim 1, wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 2, or having 95% amino acid sequence identity thereof, provided that the amino acid residues 1077 and 1020 are not cysteine.

5. The vector according to Claim 4, wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 2.

6. The vector according to Claim 1, wherein the B-domain deleted FVIII has the amino acid sequence of SEQ ID NO: 6, 7, 8, 9, or 10, or having 95% amino acid sequence identity thereof.

7. The vector according to Claim 6, wherein the B-domain - deleted FVIII has the amino acid sequence of SEQ ID NO: 6, or having 95% amino acid sequence identity thereof.

8. The vector according to Claim 1, wherein the first polynucleotide sequence further comprises a first signal sequence and the second polynucleotide sequence further comprises a second signal sequence. 9. An isolated host cell comprising the vector of Claim 1.

10. A method for preparing FVIII, comprising the steps of:

(a) transfecting cells with a plasmid comprising the vector of Claim 1,

(b) selecting the cells secreting FVIII,

(c) collecting the supernatant of the selected cells, and

(d) purifying FVIII from the selected supernatant.

Description:
VECTOR CO-EXPRESSING TRUNCATED VON WILLEBRAND

FACTOR AND FACTOR VIII

FIELD OF THE INVENTION

The present invention relates to a co-expression vector that comprises both truncated von Willebrand Factor (vWF)-Fc DNA construct and B-domain deleted FVIII DNA construct in the same vector.

REFERENCE TO SEQUENCE LISTING, TABLE OR COMPUTER PROGRAM

The Sequence Listing is concurrently submitted herewith with the specification as an ASCII formatted text file via EFS-Web with a file name of Sequence Listing.txt with a creation date of March 28, 2019, and a size of 129 kilobytes. The Sequence Listing filed via EFS-Web is part of the specification and is hereby incorporated in its entirety by reference herein.

BACKGROUND

Blood clotting proceeds through a complex and dynamic biological pathway of interdependent biochemical reactions, referred to as the coagulation cascade, in which three pathways, the extrinsic pathway, the intrinsic pathway and the common pathway are included. Factor VIII (FVIII) is a glycoprotein, circulating as an inactive cofactor with full- length von Willebrand factor (vWF) in plasma in 1 :50 ratio, that plays a critical role in the intrinsic pathway for maintaining normal hemostasis. Full-length FVIII is a non-covalently bound heterodimer comprised of a heavy chain (A1-A2-B domains) and light chain (A3-C1- C2 domains). In response to injury, the activated form of factor VIII separates from vWF and forms a complex with factor IXa and Factor X (so-called Xase complex) on the charged phospholipid membranes provided by activated platelets. The Xase complex further activates Factor V (FV) to generate FVa, which in turn activates prothrombin to thrombin together with Factor Xa and other components in the coagulation cascade to generate a stable clot.

Hemophilia A is a congenital X chromosome-linked bleeding disorder characterized by a deficiency in FVIII activity. Diminished FVIII activity inhibits the positive feedback loop in the coagulation cascade, ultimately leading to bleeding episodes with increased duration, extensive bruising, spontaneous oral and nasal bleeding, joint stiffness and chronic pain, and possibly internal bleeding and anemia, in severe cases. The most common treatment for Hemophilia A is replacement therapy with either human plasma-derived or recombinant FVIII through intravenous administration.

High production of recombinant FVIII (rFVIII) in mammalian cells has been found to be difficult. The large molecular weight of this protein (~ 300 kDa), the complexity of the post-translational modifications required (e.g., numerous glycosylation and tyrosine sulfation sites), and the limits of expression elements (like mRNA instability) make the high production of rFVIII a challenge. Removal of the central B-domain has greatly improved rFVIII production, but the commercial production level of rFVIII is still generally in the range of 20 - 50 IU/ml. Additional possible factors contributing to the low expression level of the recombinant FVIII in mammalian cells may include, specifically or non-specifically, binding to expression cell membranes and FVIII instability (Kaufman, 1989, Mol. Cell. Biol., vol. 3, pp. 1233-1242: US Patent No. 8,759,293).

US Patent No. 8,759,293 discloses a method for preparing FVIII. FIG. 6 of the‘293 Patent illustrates a first plasmid expression vector that expresses (i) mature vWF or truncated vWF domain-Fc fusion polypeptides, and (ii) propeptide sequences from independent promoters, and a second and a different plasmid expression vector the expresses (iii) human FVIII using a different selectable marker. The first and the second plasmids are co transfected and taken up into mammalian cells under selection to create a stable cell line that expresses (i) vWF or vWF-Fc, (ii) vWF propeptide, and (iii) FVIII. The problem for this method is that each selected cell transfectant is obtained by independent and separate entry of FVIII and vWF-Fc plasmids into a mammalian cell. Selected cells can have different ratios of each plasmid. It is difficult to obtain cells reliably and reproducibly with consistent expression of proteins if the ratio of the transfected plasmids is unknown, and thus the absolute levels of the expressed proteins will change over time. For example, a primary cell transfectant produced by sequential transfection may have 10 copies of FVIII and 30 copies of vWF-Fc. Over-expression of vWF-Fc over FVIII, would result in excessive quantities of vWF-Fc relative to FVIII, and thus make downstream purification of FVIII difficult. Further, if the ratio of the two plasmids changes over time, a cell may become undesirable due to reduction in FVIII plasmids, while maintaining a constant number of vWF plasmids. The loss of one plasmid relative to another may upset the balance in expression levels, which results in difficult purification scenarios.

There is a need for improvement in the quality and quantity of recombinant FVIII available to patients. There is a need for improving the expression of FVIII and improving the yield of expressed FVIII after purification. BRIEF DESCRIPTION OF THE DRAWINGS

FIGs. 1 A-1C illustrate the construction of a dual expression vector, in which both human FVIII and truncated, and in some cases, mutated, vWF-Fc cDNAs are subcloned juxtaposed into the same expression vector. (1A) A general representation of the dual expression vector containing FVIII and vWF-Fc variant gene sequences under control of cytomegalovirus (CMV) promoters. (IB) A representation of the co-expression of FVIII and truncated vWF (with or without the vWF propeptide domain) and fused to an

immunoglobulin Fc domain (Pro-D’D3-Fc contains the propeptide domain, while Del-D’D3- Fc does not). (1C) A representation of the co-expression of FVIII and truncated and mutated vWF (at amino acids Cl 099 and Cl 142 of complete wild-type vWF sequence, according to the HGVS numbering convention, and with or without the vWF propeptide domain) and fused to an immunoglobulin Fc domain from human IgGi. Other plasmid elements are also shown.

FIGs. 2A-2D show a schematic representation of the derivation of a truncated human vWF-Fc protein created by fusion of a D’D3 domain to the Fc hinge domain of human IgGi. (2A) Domain structure of a secreted von Willebrand factor polypeptide (signal sequence not shown). D1 and D2 represent the vWF propeptide sequence; D’D3 includes the nominal Factor VIII binding site; CK is the terminal dimerization domain of wild-type vWF. (2B1) Pro-D’D3-Fc is a truncated vWF protein consisting of the propeptide (D1D2 domains) and D’D3 domains fused to the Fc domain. (2B2) In this alternate form, the D1D2 domains are physically removed from vWF and fused directly to the signal sequence at the N-terminus of the D’D3-Fc sequence, creating Del-D’D3-Fc. (2C) With or without the presence of D1D2, a mature truncated product, namely, D’D3-Fc. (2D) Multimers of wild-type D’D3 domain are formed through disulfide bridges, while dimerization of D’D3 is mediated by interaction of the Fc fusion domain at the C-terminus.

FIGs. 3A-3D show a schematic representation of truncated human vWF-Fc protein, with mutations at amino acid position C1099 and Cl 142. (3 A) Domain structure of a mutated and secreted von Willebrand factor polypeptide (signal sequence not shown). D1D2 are propeptide domains; D’D3 includes the nominal Factor VIII binding site, but includes mutations at amino acids C1099 and Cl 142 (vertical lines in D3 domain). (3B1) Pro- D’D3mut-Fc is as described above for Pro-D’D3-Fc, but contains mutations at position C1099 and Cl 142. (3B2) In this alternate form, the D1D2 domains are physically removed from vWF and fused directly to the signal sequence at the N-terminus of the D’D3-Fc sequence that has mutations at C1099 and Cl 142, resulting in Del-D’D3mut-Fc. (3C) With or without the presence of D1D2, a mature truncated product, D’D3mut-Fc. (3D) Formation of multimers of wild-type D’D3 domain is blocked by the C1099 and Cl 142 mutations

(asterisks), while dimerization of D’D3 is mediated by interaction of the Fc fusion domain at the C-terminus.

FIGs. 4A-4B show a representation of the assembly of D’D3-Fc and D’D3mut-Fc proteins, with FVIII. (4A) D’D3-Fc (no introduced mutations) results in multimer assembly and dimerization as described in FIG. 3. As an example, binding to FVIII is shown at two sites on multimerized D’D3-Fc molecule. (4B) D’D3mut-Fc fusion protein is expected to result in assembly of dimers instead of multimers, due to introduced mutations Cl 099 and Cl 142 (shown as an asterisk).

FIGs. 5A-5B illustrate differences in the likely capture and recovery of

FVIII/truncated vWF-Fc complexes due to differences between non-mutated and mutated D’D3 variants, and consequences of purification of resulting Factor VIII though a Factor VIII affinity matrix. (5 A) Multimers D’D3-Fc that bind to FVIII are shown. FVIII binds to a purification matrix through binding interactions to an affinity ligand and is retained along with D’D3-Fc on the column. FVIII eluted from such a column is probably heavily contaminated with D’D3-Fc, so that [FVIII] < [D’D3-Fc] (5B) Dimeric forms of D’D3mut- Fc are shown to bind Factor VIII. Due to the lack of multimerization, individual FVIII/D’D3- Fc complexes are expected to bind to anti-FVIII affinity matrix efficiently, yielding a significantly purer population of FVIII, where [FVIII] » [D’D3mut-Fc].

FIGs. 6A-6B show differences in the capture and recovery of FVIII- vWF-Fc complexes and the consequences of purification of resulting Factor VIII though a vWF-Fc capture matrix, similar to FIG 5. (6A) Multimers of D’D3-Fc that bind to FVIII are shown. This could result in decreased recovery of FVIII, if D’D3-Fc is abundantly co-expressed. In addition, due to increased concentration and proximity of D’D3-Fc, FVIII may be poorly eluted from an affinity column, so that [FVIII] « [D’D3-Fc] (6B) Dimeric forms of D’D3mut-Fc are shown to bind FVIII are represented to bind Factor VIII. Due to the lack of multimerization, FVIII/D’D3mut-Fc complexes are expected to bind to the anti-Fc affinity matrix without aggregate formation, yielding an enriched population of FVIII binding to vWF-Fc at 1 : 1 ratio.

FIGs. 7A-7B demonstrate the purity of FVIII expressed with Del-D’D3mut-Fc and purified by affinity chromatography. (7 A) A gel electropherogram of recombinantly- expressed FVIII obtained by purification, and (7B) a corresponding Western Blot for the protein lanes in (7 A). An anti-human FVIII antibody was used to detect the protein in the Western blot. Lane 1, Molecular weight marker (MW); Lane 2, starting material from cell supernatants; Lane 3, flow-through material after Protein A chromatography; Lane 4, 0.3 M CaCh elution fraction from a Protein A affinity column; Lane 5, 50% ethylene glycol elution of material from FVIIISelect affinity resin column; Lane 6, commercial protein BDD- FVIII (Xyntha ® ). Arrows indicate the three expected BDD-FVIII protein bands corresponding to approximately 170, 90 and 80 kilodaltons (kD).

FIG. 8 shows the expression of truncated vWF-Fc dimers and multimers. Protein A- purified, truncated vWF-Fc variants including Pro-D’D3-Fc (Lane 2); Del-D’D3-Fc (Lane 3), Pro-D’D3mut-Fc (Lane 4) and Del-D’D3mut-Fc (Lane 5) were compared with plasma- derived full-length vWF protein (Lane 6) by non-reducing PAGE. The high molecular weight bands (>180 kD, arrows) in lanes 2 and 3 represent truncated vWF-Fc multimers. Since the mutations at C1099 and Cl 142 interrupt multimer formation, the Pro-D’D3mut-Fc (Lane 4) and Del-D’D3mut-Fc (Lane 5) only formed dimers (-160 kD; arrows).

FIG. 9 demonstrates the protective effects of truncated vWF-Fc fusion proteins on recombinant FVIII protein expression. Truncated vWF-Fc fusion proteins, including wild- type Pro-D’D3-Fc, Del-D’D3-Fc, as well as Pro-D’D3mut-Fc and Del-D’D3mut-Fc proteins and plasma-derived vWF (negative control protein, human serum albumin (HSA), were tested by addition of equimolar amounts of variant (or HSA) with cells expressing only FVIII. FVIII activity was determined using a one-stage clotting assay, with samples removed for testing at timed intervals, between 0 and 8 hours.

FIG. 10 illustrates that co-expression of truncated vWF-Fc with FVIII considerably improved the in vitro stability of FVIII in cell culture. Samples of cell culture media containing either FVIII, or FVIII/Del-D’D3mut-Fc complexes, were maintained at room temperature over a 24-hour period, with sampling at 0, 4, 8, and 24 hours. Stability was assessed as a function of FVIII activity in a one-stage clotting assay.

FIG. 11 demonstrates that co-expression of truncated vWF-Fc with FVIII improved recombinant FVIII stability during a solvent-detergent viral inactivation procedure. Cell cultures expressing either recombinant BDD-FVIII only, or BDD-FVIII co-expressed with Del-D’D3mut-Fc vWF variant, were treated with viral inactivation reagents (i.e, TNBP and Triton-X-100) for 24 hours, and the FVIII coagulation activity were monitored by a one-stage clotting assay at 0, 4, 8, and 24 hours timepoints. DETAILED DESCRIPTION OF THE INVENTION

The present invention provides the solution to many of the current issues in human recombinant coagulation Factor VIII purification and production, namely, to have both FVIII and truncated vWF-Fc expression cassettes co-expressed from two independent promotors on the same expression vector with a single selectable marker gene. While the absolute expression levels of the expressed proteins might change, a desirable 1 : 1 ratio of FVIIFvWF- Fc expression is established, thereby minimizing the likelihood that vWF significantly overwhelms FVIII during purification, which would make recovery of FVIII problematic.

In addition to controlling the relative ratios of expression genes, and their proteins, a method has been developed to improve recovery of FVIII: vWF complexes. This is done by fusing an immunoglobulin Fc region onto the C-terminus of truncated regions of human rec- vWF, specifically, the D’D3 region that binds FVIII (ca. aa 776-1241). In addition, however, the vWF-Fc domains are created by direct fusion to a signal sequence to promote efficient processing, and further, has been modified with specific mutations to promote higher ratios of FVIIFvWF complexes. The fusion with Fc provides a straightforward method to select FVIII- vWF complexes from the bulk of the cell supernatant, by virtue of the ability of Fc regions to bind to staphylococcal-derived protein A, anti-Fc antibody, etc., that are immobilized on solid matrices. Naturally, other methods can used to achieve a similar result, but a preferred C-terminal fusion partner is immunoglobulin Fc as it also facilitates truncated vWF fragment dimerization, that is important for FVIII binding (Chiu et ak, 2015, Blood, vol. 126, pp. 935-938 and Yee et ak, 2015, Blood, vok 126, pp. 939-942). The potential issue of expressing wild-type truncated vWF-Fc is that the truncated vWF-Fc proteins engage in normal head-to-head and tail-to-tail multimerization through one or two specific cysteine residues in the D’D3 domains, as well as through the dimerization of Fc domains.

Recombinant vWF-Fc polymers can thus yield long molecules as long as 50 polypeptide units in length, and covalently-linked; typically, they are in the size range of about 20-25 polypeptide units. Such size ranges represent molecular weights of 5-10 million daltons, or more, that become problematic for large-scale column purification typically employed for preparing proteins, especially on a commercial scale. The large size of such complexes can create tangles or aggregates that often block column flow, create excessive back-pressure, and make recovery of the desired proteins difficult.

The present invention solves this problem by including mutations at certain cysteine amino acids in the D’D3 domain, specifically, at amino acids C1099 and Cl 142 of vWF polypeptide. These amino acids appear to be important in driving inter-molecular disulfide bridges between D’D3 dimers, but do not appear to be critical for FVIII binding. As such, using mutations introduced into the D’D3 domain of vWF-Fc should prevent multimer assembly, and limit the size of the D’D3 domains to monomers or dimers, especially at high concentrations of expressed protein. This is ideal for large-scale column purification and preventing clogging of depth-filtration columns, affinity columns, etc., and will yield better recovery and purity of FVIII.

An additional modification disclosed in the present invention can further enhance both expression and purification - elimination of the gene sequence encompassing the vWF propeptide domain from the expression cassette encoding truncated vWF-Fc. This propeptide normally remains associated with, and helps to fold, vWF after cleavage in, and transit from, the Golgi apparatus - it is included in expression of mammalian vWF proteins in cell-based systems. However, large amounts of vWF that are expressed in mammalian systems are inefficiently processed by the furin/Kex2-like proteases that are present (or insufficiently present) in mammalian cells; as a consequence, unprocessed propeptide still attached to “mature” vWF protein, or truncated fragments, can further complicate the large size of vWF multimers that may be formed, and render purification difficult.

The present invention removes the propeptide sequence from the vWF expression cassette and solves this problem by eliminating the need for the furin/Kex2-like proteins: mature domains are expressed directly by cleavage of the pre-protein from its signal peptide. Despite the absence of the propeptide in the folding of vWF domains, the fused Fc domain provides sufficient folding to drive dimerization of the D’D3 sequence that binds to FVIII. Elimination of the propeptide sequence further reduces the cassette size in expression vectors and allows for better expression in mammalian cells.

The present invention uses a single DNA expression vector comprising DNA constructs of recombinant FVIII and its modified binding partner protein, i.e., truncated, recombinant von Willebrand factor (vWF), to co-express the two proteins in mammalian cells, at very high expression levels of FVIII.

Four different truncated vWF-Fc DNA cassettes, each having a nucleotide sequence that encodes a truncated vWF-Fc fusion protein, are inserted into the expression plasmid vector; truncated vWF-Fc constructs are defined as follows: (a) Pro-D’D3-Fc represents a construct that contains vWF propeptide domain (D1D2, comprising amino acids +1-741 of the wild-type vWF molecule) and domains D’D3 (comprising amino acids 742-1247 of the wild-type vWF molecule), fused at its C-terminus with an immunoglobulin Fc domain from human IgGi (e.g., UniProtKB 01857, comprising amino acids 104-330); (b) Del-D’D3-Fc represents a construct that contains vWF domains D’D3 (comprising amino acids 742-1247 of the wild-type vWF molecule), fused at its C-terminus with an immunoglobulin Fc domain from human IgGi (e.g., UniProtKB 01857, comprising amino acids 104-330); in contrast to construct (a) above, the propeptide domain (D1D2) has been deleted in this construct; (c)

comprising amino acids +1-741 of the wild-type vWF molecule) and domains D’D3, amino acids of the wild-type vWF molecule, but with mutations at Cl 099 and Cl 142 of the complete vWF molecule (including its signal sequence; numbering is according to the Human Genome Variant Society (HGVS) numbering convention), fused at its C-terminus with an immunoglobulin Fc domain from human IgGi (e.g., UniProtKB 01857, comprising amino acids 104-330); and (d) Del-D’D3mut-Fc represents a construct that contains vWF domains D’D3 (comprising amino acids 742-1247 of the wild-type vWF molecule) but with mutations at Cl 099 and Cl 142 of the complete vWF molecule (including its signal sequence;

numbering is according to the HGVS numbering convention), fused at its C-terminus with an immunoglobulin Fc domain from human IgGi (e.g., UniProtKB 01857, comprising amino acids 104-330), but in contrast to construct (c), the propeptide domain (D1D2) has been deleted in this construct.

vWF and FVIII polypeptides of human and non-human (e.g., primates, dogs, cats, horses, pigs, mice, rats, guinea pigs, rabbits, cows, other vertebrates) origin are contemplated by the present invention, which include natural, synthetic, and recombinant proteins. Also within the scope of the present invention are vWF and FVIII polypeptides corresponding to wild-type proteins, or mutants, variants, and/or truncations thereof.

Human wild-type vWF amino acid sequence and nucleic acid sequence encoding vWF are disclosed by, e.g., GenBank Accession Nos.: NP_000543, NM_000552. Human vWF is a polypeptide of 2813 amino acids including a signal peptide of 22 amino acids and repetitive functional domains, A, B, C, D and CK, which are distributed from the amino terminal in the order Dl, D2, D’, D3, Al, A2, A3, D4, Bl, B2, B3, Cl, C2, and CK (FIG.

2A). The "mature" vWF subunit is composed of, from the N- to the C-terminus in the order, the domains D'-D3-A1-A2-A3-D4-B1-B2-B3-C1-C2-CK. Domains D1D2 (propeptide) are amino acid +1-741 of the wild-type vWF protein, and domains D’D3 are amino acids +742- 1247 of the wild-type vWF protein. (+ refers to signal sequence removal at the N-terminus, i.e., +1 is the first amino acid of the wild-type vWF protein without the signal sequence). D’D3 domains are recognized as the binding site for FVIII. Human FVIII is synthesized as a single-chain molecule of approximately 300 kDa and consists of the structural domains A1-A2-B-A3-C1-C2. It is often cleaved within the B domain during secretion and circulates in the blood as a heterodimer bound to von

Willebrand factor (vWF). The smallest active FVIII, with a molecular weight of 170 kD, consists of one 90 kD heavy chain which includes the A1-A2 domains, lacking the major part of the heavily glycosylated B-domain (Ser741 to Argl648), and one 80 kD light chain including A3 -Cl and C2 domains. It can be activated with thrombin to the same extent as the forms of higher molecular mass. The B-domain of FVIII is not necessary for the clotting activity of FVIII, but it was found that by deleting the B-domain, the FVIII has substantially higher expression levels from heterologous systems without impairing it’s in vitro or in vivo functionality. The present invention utilizes B-domain deleted (BDD)-FVIII, wherein the two chains of FVIII are joined by a linker (SFSQNPPVLKRHQR). A human BDD FVIII sequence is shown in GenBank Protein Acc. ABV90867.

The inventors have designed A DNA vector containing dual expression cassettes to have both FVIII and truncated vWF-Fc genes co-expressed under two independent promotors. While the absolute expression levels of the expressed proteins might change, the DNA vector of the present invention may achieve a constant and desirable 1 : 1 ratio of FVIII to truncated vWF-Fc expression, which ensures that the level of truncated vWF-Fc protein does not significantly overwhelm the level of FVIII protein during purification, and therefore, the recovery of FVIII is under control. The expression level of FVIII produced by co expression of FVIII and truncated vWF-Fc in one expression vector is significantly higher than those produced by other known methods.

The present invention is directed to a vector comprising: (a) a first polynucleotide sequence encoding a B-domain deleted FVIII protein (FVIII), operably linked to a first promoter, and (b) a second polynucleotide sequence encoding a fusion protein comprising a truncated von Willebrand factor (vWF) and an immunoglobulin Fc fused to the C-terminal of the truncated vWF, operably linked to a second promoter, wherein the truncated vWF comprises the amino acid sequence of SEQ ID NO: 1 [D’/D3 + mutation, or Del-D’D3mut], or SEQ ID NO: 2 [Dl/D2/D’/D3 + mutation, or Pro-DD3mut], or SEQ ID NO: 3 [D’/D3, or Del-D’D3], or SEQ ID NO: 4 [Dl/D2/D’/D3, or Pro-D’D3], or having 95% amino acid sequence identity thereof. The first promoter and the second promoter can be the same or different. In one embodiment, the first promoter and the second promoter are the same, e.g., cytomegalovirus promoter. In the vector of the present invention, a first polynucleotide sequence encodes a B- domain-deleted FVIII (BDD-FVIII), or an amino acid having at least 95%, or 96%, or 97%, or 98%, or 99% sequence identify thereof. The amino acid changes are preferably of a minor change such as a conservative amino acid substitution that does not significantly affect FVIII folding or binding capacity.

In the vector of the present invention, the second polynucleotide sequence encodes a fusion protein comprising a truncated vWF and an immunoglobulin Fc fused to the C- terminal of the truncated vWF, or an amino acid having at least 95%, or 96%, or 97%, or 98%, or 99% sequence identify thereof. The amino acid changes are preferably of a minor change such as a conservative amino acid substitution that does not significantly affect the binding activity of vWF to FVIII or cause multimerization of vWF. When the truncated vWF is D’D3 + mutation, the amino acid substitution does not include the change of amino acid residue 1099 or 1142 (numbered according to the HGVS numbering convention) or, equivalently, 336 or 379 (counted from the N-terminus of D’) back to cysteine, which negates the mutation. When the truncated vWF is Dl/D2/D’/D3 + mutation, the amino acid substitution does not include the change of amino acid residue 1099 or 1042 or, equivalently, 336 or 379 (counted from the N-terminus of D’) back to cysteine, which negates the mutation.

To improve recovery of FVIII- vWF complexes during purification, the present invention fuses an immunoglobulin Fc region onto the C-terminus of truncated regions of human recombinant vWF, specifically, onto the D’D3 region (amino acids +742-1247 of the wild-type vWF protein). Suitable immunoglobulin Fc for the present invention includes those that can bind with high affinity to protein A or protein G or other similar Fc-binding matrices, and include the Fc regions of human IgG, murine IgG, or fragments thereof comprising at least the hinge region to facilitate the formation of interchain disulfide bonds. A preferred Fc is human IgGl or human IgG4. For example, a preferred Fc sequence is shown in SEQ ID NO: 5, which is amino acids 104-330 of human IgGi.

The fusion with Fc provides a straightforward method to select FVIII-vWF complexes from the bulk of the cell supernatant, by the ability of Fc regions to bind to its binding partner, such as staphylococcal-derived protein A, anti-Fc antibody, etc., immobilized on solid matrices. Since the truncated vWF lacks the CK domain for dimerization, the Fc portion of the vWF-Fc fusion protein also provides a mean for self-dimerization of truncated vWF-Fc through cysteines in the Fc domains. In one embodiment, mutations are implemented into the truncated human vWF to create smaller binding complexes compared to wild-type vWF, which provide overall enhanced purification and greater in vitro stability of recombinant FVIII-vWF-Fc complexes. During secretion, vWF multimerization forms via disulfide bonds in D3 domain (Cl 099 and Cl 142), that leads to the formation of large, to extremely large, vWF molecules, typically multimers of 20-25 vWF molecules (or 20-25 - mers), up to 50 - mers. These size ranges represent molecular weights of at least 5-10 million daltons, which becomes problematic for large-scale column purification of F VIII. This is because the large size of such complexes can create tangles or aggregates that often block column flow and make recovery of the desired proteins difficult or impossible, while affecting the overall purity. Amino acids Cl 099 and Cl 142 (HGVS numbering) are two important residues for driving inter-molecular disulfide bridges between D’D3 domains, but they are not critical for binding to FVIII. By converting C1099 and Cl 142 of the D’D3 domain of vWF-Fc to A1099 and A1142, multimerization is prevented, which dramatically improves the purification and protein quality of FVIII from FVIII-vWF complexes. The cysteine mutation in domain D’D3 can be changed to alanine as shown in SEQ ID NO: 1, or other amino acids such as leucine, glycine, valine, isoleucine, phenylalaninse, serine, threonine, histidine, methionine, and possibly arginine, lysine, glutamine, asparagine, glutamic acid, aspartic acid, and tryptophan and tyrosine, but not proline, as long as the substitution for cysteine does not significantly affect the ability of the resulting vWF to bind to FVIII.

In one embodiment, the propeptide domain of vWF is deleted by elimination of the gene sequence encompassing the propeptide from the expression cassette encoding vWF-Fc. One of the functions of the propeptide is to aid in the N-terminal multimerization of vWF; the propeptide is then cleaved by furin when released into blood. However, furin/Kex-2-like proteases are often limiting in cells, so that when large amounts of vWF that are expressed in mammalian systems, they may be inefficiently processed by the furin/Kex2-like proteases, leaving uncleaved propeptide attached to vWF. This, in turn, significantly increases the molecular weight of the resulting vWF molecule and limits proper domain association. By removing of the propeptide sequence from the vWF expression cassette, the mature domains are expressed directly from its signal peptide, leading to a significant increase in vWF expression, and resulting FVIII-vWF complex formation. Despite the absence of the propeptide, the fused Fc domain provides sufficient folding to drive dimerization of the D’D3 that has higher affinity for FVIII than monomeric D’D3. Elimination of the propeptide domain further reduces the gene size in expression vectors and results in better protein expression.

Preferred B-domain deleted FVIII polypeptides for this invention include human BDD-FVIII (GenBank Protein Accession No. ABV90867, SEQ ID NO: 6), porcine BDD- FVIII, including synthetic porcine BDD-FVIII (GenBank Protein Accession No.

AGV79859.1, OBIZUR ® , SEQ ID NO: 7); a variant of porcine BDD-FVIII, sequence derived from GenBank Nucleotide Accession No. NM_214167.2, but with an artificial 14 amino acid linker derived from the human FVIII B-domain designed to replace the porcine FVIII full- length B-domain (SEQ ID NO: 8); porcine BDD-FVIII, sequence derived from GenBank Nucleotide Accession No. U49517, but with an artificial 14 amino acid linker derived from the human FVIII B-domain designed to replace the porcine FVIII full-length B-domain (SEQ ID NO: 9), and canine BDD-FVIII sequence derived from GenBank Nucleotide Accession No. AF049489.1, but with an artificial 14 amino acid linker derived from the human B- domain, and designed to replace the canine FVIII full-length B-domain (SEQ ID NO: 10).

The use of porcine and canine (and even other mammalian FVIIIs in addition to human) are contemplated here. Porcine FVIII is currently used as a treatment for patients with acquired hemophilia A. Porcine and canine FVIII molecules, or hybrids thereof, have been evaluated and considered as alternative treatments in patients with congenital hemophilia, or hemophilia with inhibitors. By creating expression cassettes encoding porcine or canine FVIII, with a shortened B-domain linker in the presence of wild-type, truncated and/or mutated vWF, can yield large amounts of FVIII product according to the present invention; in addition, such porcine and canine FVIIIs, or hybrids thereof, may have higher catalytic activity and thus smaller amounts of FVIII may be needed in treatment scenarios.

The present invention provides recombinant expression vectors for expression FVIII and truncated vWF-Fc, and host cells transformed with the expression vectors. Any suitable expression system may be employed. The vector comprises a first and a second DNA sequence encoding FVIII and truncated vWF-Fc proteins, respectively, operably linked to suitable transcriptional or translational regulatory nucleotide sequences, such as those derived from a mammalian, microbial, viral, or insect gene. Examples of regulatory sequences include transcriptional promoters, operators, or enhancers, an mRNA ribosomal binding site, and appropriate sequences which control transcription and translation initiation and termination. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the encoding DNA sequence. Thus, a promoter nucleotide sequence is operably linked to the encoding DNA sequence if the promoter nucleotide sequence controls the transcription of the encoding DNA sequence. The ability to replicate in the desired host cells, usually conferred by an origin of replication, and a selection gene by which

transformants are identified, may additionally be incorporated into the expression vector.

In one embodiment, DNA sequences encoding appropriate signal peptides that may or may not be native to the FVIII or vWF can be incorporated into expression vectors. A signal peptide (also known as a leader peptide or a signal sequence) is a short peptide (typically 16- 30 amino acids long) present at the N-terminus of the newly synthesized proteins that are destined towards the secretory pathway. For example, a DNA sequence for a signal peptide (secretory leader) may be provided in frame to the first sequence so that the expressed polypeptide is initially translated as a fusion protein comprising the signal peptide. A signal peptide that is functional in the intended host cells enhances extracellular secretion of the polypeptide. In some embodiments, the signal peptide is cleaved from the polypeptide upon secretion of the polypeptide from the cell. Appropriate signal peptides that are native or not native to the amino acid sequence can also be used in the present invention.

To improve the secretion of human FVIII protein, the first polynucleotide preferably includes a signal sequence before the FVIII sequence. Any signal sequence that facilitates the secretion of FVIII is suitable for the present invention. For example, the signal sequence is a wild-type human FVIII signal sequence MQIELSTCFFLCLLRFCFS (SEQ ID NO: 11).

To improve the secretion of porcine FVIII proteins, the first polynucleotide preferably includes a signal sequence before the FVIII sequence. Any signal sequence that facilitates the secretion of FVIII is suitable for the present invention. For example, the signal sequence is a wild-type Porcine VFIII signal sequence MQLEL S T C VFLCLLPLGF S (SEQ ID NO:

12).

To improve the secretion of canine FVIII protein, the first polynucleotide preferably includes a signal sequence before the FVIII sequence. Any signal sequence that facilitates the secretion of FVIII is suitable for the present invention. For example, the signal sequence is a wild-type canine VFIII signal sequence MQVELYTCCFLCLLPFSLS (SEQ ID NO: 13).

To improve the secretion of truncated vWF-Fc protein, the second polynucleotide preferably includes a signal sequence before the vWF sequence. Any signal sequence that facilitates the secretion of truncated vWF-Fc is suitable for the present invention. For example, the signal sequence is a wild-type human vWF signal sequence

MIP ARF AGVLL AL ALILPGTLC (SEQ ID NO: 14).

Suitable host cells for co-expression of the polypeptides of the present invention include prokaryotes, yeast, filamentous fungi, or higher eukaryotic cells. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are well-known, for example, described in Pouwels et al. in Cloning Vectors: A Laboratory Manual, Elsevier, New York, (1985).

Suitable eukaryotic promoters for mammalian expression include the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus (RSV), and metallothionein promoters, such as the mouse metallothionein-I promoter.

In some embodiments, a vector construct can be introduced into the cultured host cell by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid- mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals.

Transcriptional and translational control sequences for mammalian host cell expression vectors may derive from viral genomes. Suitable promoter sequences and enhancer sequences for the present invention can be derived from cytomegalovirus (CMV), Polyoma virus, Adenovirus 2, Simian Virus 40 (SV40). DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide other genetic elements for expression of a structural gene sequence in a mammalian host cell. Viral early and late promoters are particularly useful because both are easily obtained from a viral genome as a fragment which may also contain a viral origin of replication.

Sequences encoding selectable markers can be included in the DNA construct.

Selectable markers for mammalian cells are known in the art, which include, glutamine synthetase, thymidine kinase, dihydrofolate reductase, aminoglycoside phosphotransferase, hygromycin B phosphotransferase, asparagine synthetase, adenosine deaminase,

metallothionien, and antibiotic resistant genes such as neomycin.

The present invention comprises four major improvements related to high expression and purification of recombinant human FVIII, and its modified binding partner protein, in this case, truncated, recombinant human von Willebrand factor (vWF), in mammalian cells, namely, 1) co-expression of rec-FVIII and truncated vWF-Fc from independent promoters on a single mammalian expression vector to yield high levels of expression of both proteins through the control of optimal expression ratio of these two molecules; 2) implementation of mutations into variants of truncated human vWF to provide enhanced purification of recombinant FVIII-vWF-Fc complexes by creating smaller (i.e., monomer, dimer, or short oligomer) binding complexes compared to wild-type forms (multimers); 3) introduction of a substantial modification of the mutated von Willebrand molecule, namely, deletion of the propeptide region, that will significantly increase its expression, by eliminating the need for cleavage by certain, limiting, endogenous proteases; and 4) stabilization of the FVIII molecule in cell culture, storage in vitro , and during methods to achieve inactivation of mammalian viruses.

The combination of the elements described above results in great improvements in expression of FVIII, in greater purification of expressed protein with less loss of material, and reduced host cell protein in the final product, and also during solvent/detergent steps during inactivation of potential viruses present. These combined elements greatly improve the quality and quantity of recombinant FVIII available to patients.

The overall method of synthesis, assembly, or process of the present invention is outlined below. FVIII and truncated vWF-Fc mutants are synthesized and cloned in tandem into a single expression vector that comprises two independent expression cassettes driven by mammalian promoters and terminated with mammalian polyadenylation signals (FIG. 1). Transfected cells are selected using antibiotic (e.g. Geneticin) or metabolic (e.g., glutamine synthetase) gene products, and then further selected and analyzed for optimal FVIII and vWF expression.

Creation of truncated vWF-Fc variants such as Pro-D’D3-Fc, Pro-D’D3mut-Fc, as well as the equivalent forms that specifically lack the propeptide domain with or without mutations, namely, Del-D’D3-Fc and Del-D’D3mut-Fc, are performed by gene synthesis of the desired DNA segments.

The FVIII-vWF complex may be removed from media by conventional

chromatographic methods including absorption onto charge matrices, and/or by affinity or pseudo-affinity chromatography, for example protein A chromatography. FVIII can then be further purified away from the FVIII-vWF complex by using a FVIIISelect chromatography resin, and selective washing steps, to yield an enriched population of FVIII molecules, minimally contaminated by vWF.

The overall method for preparing FVIII is summarized as follows:

1) Transfection of cells with the co-expression plasmid containing BDD-FVIII and mutated/modified D’D3 vWF-Fc gene cassettes.

2) Identification of cells optimally expressing FVIII.

3) Recovery of secreted cell supernatant by separation of cell mass from culture medium. 4) Preparation of recovered cell medium into desired buffer for application to a column matrix that selects FVIII-vWF-Fc complexes (for example, binding to immobilized Protein A column (preferred), anti-vWF affinity matrix, or an ion exchange column).

5) The retained or eluted FVIII is further purified with a second column (for example, a camelid anti-FVIII antibody column, affinity ligand, or other affinity matrix) to remove any residual vWF-Fc and cell proteins which were not removed in step 4; it is clear that the order of removal may be reversed (steps 4 and 5), as needed, utilizing either direct (via anti-FVIII) or indirect (via anti-vWF or vWF-Fc) binding matrices.

6) FVIII can be further purified and polished by additional chromatographic steps, as necessary, including ion exchange column and nanofiltration.

The sequence identifiers for the corresponding encoded amino acid sequences are shown in Table 1.

Table 1.

The following examples further illustrate the present invention. These examples are intended merely to be illustrative of the present invention and are not to be construed as being limiting.

EXAMPLES

Example 1. Construction of co-expression vectors for truncated vWF-Fc and FVIII polypeptides

Four truncated vWF-Fc DNA cassettes, each having a nucleotide sequence that encodes a truncated vWF-Fc fusion protein, were commercially synthesized (GenScript, Piscataway, NJ). The four truncated vWF-Fc DNA cassettes are defined as follows.

(a) Pro-D’D3-Fc (D 1/D2/D7D3-Fc) represents a construct that contains vWF propeptide domain (D1D2, amino acids +1-741 of the wild-type vWF molecule, GenBank NM_000552) and domains D’D3 (amino acids 742-1247 of the wild-type vWF molecule,), fused at its C-terminus with an immunoglobulin Fc domain derived from human IgGi (e.g., UniProtKB 01857, amino acids 104-330).

(b) Del-D’D3-Fc (D’/D3-Fc) represents a construct that contains vWF domains D’D3, fused at its C-terminus with an immunoglobulin Fc domain; in contrast to construct (a), the propeptide domain (D1D2) has been deleted.

(c) Pro-D’D3mut-Fc (Dl/D2/D’/D3 + mutation-Fc) represents a construct similar to (a), except having mutations at Cl 099 and Cl 142 of the complete vWF molecule (including its signal sequence). C1099 and Cl 142 are from an old and recognized naming convention from the Human Genome Variation Society, where the amino acid counting starts from the signal sequence of the complete vWF molecule; when counting starts from the D’ domain, the equivalent mutations are C336 and C379.

(d) Del-D’D3mut-Fc (D’/D3 + mutation) represents a construct similar to (b) except having mutations at Cl 099 and Cl 142 of the complete vWF molecule (including its signal sequence, with nomenclature according to HGVS numbering convention).

DNA encoding the human FVIII B-domain deleted protein (BDD FVIII) was also commercially synthesized (Genewiz, South Plainfield, NJ).

Nucleotide sequences encoding for each protein listed in Table 1 were codon- optimized using algorithms that account for enhanced heterologous expression Chinese Hamster Ovary (CHO) cells. The DNA sequence that encodes the signal peptide for each protein was also codon-optimized. The truncated vWF-Fc and BDD FVIII cDNAs were each subcloned into a DNA plasmid expression vector through cloning sites BsiWI and Fsp I on the plasmid (Fig.1), where‘CMV’ is a cytomegalovirus promoter sequence, represented by a white squiggle line for FVIII and black squiggle line for vWF-Fc;‘FVIIF is a FVIII gene sequence (BDD FVIII); ‘vWF-Fc’ is a generic name for any vWF-Fc variant described above, including Pro-D’D3- Fc, Del-D’D3-Fc, Pro-D’D3mut-Fc and Del-D’D3mut-Fc;‘arnpR’ is an ampicillin-resistance gene;‘selectable mkri is a selectable marker for specifically isolating cells that carry the desired plasmid (e.g. glutamine synthetase, G418);‘SV40 ori’ is the sequence of mammalian simian virus that contains an origin of replication (for replication in mammalian cells);‘fl ori’ is the sequence of bacteriophage fl that contains an origin of replication (for replication in bacteria cells).

Since dimerization of vWF fragment is important for its interaction with FVIII, the DNA sequence encoding the Fc fragment was introduced to the C-terminus of truncated vWF fragment to direct truncated vWF dimer formation. In addition, since highly-expressed, wild- type vWF, despite the fact that it is truncated in the present invention, may form aggregates due to its extensive multimerization, and thus complicate FVIII and purification, two mutations were introduced at amino acid positions Cl 099 and Cl 142 (including signal sequence; HGVS numbering convention), or C336 and C379 (counting starting from D’ in the vWF D’D3 domain). These two sites are involved in vWF multimer formation and thus introducing mutations at these sites block vWF multimer formation, simplify purification, and create a better quality of FVIII than current methods.

Example 2. Co-expression of human BDD FVIII and truncated vWF-Fc in CHO mammalian cell lines

CHO cells were cultured in CD-CHO medium (Thermo Fisher Scientific, Waltham, MA) and prepared for electroporation. Cells were seeded at 0.3 x lO 6 cells/mL in E125 (125 ml) shaker flasks (SF) and incubated in CO2 shaker incubator. Cells were sub-cultured when the cell density reached 1.5-3. Ox 10 6 cells/ mL; at cell passage 3 (P3), electroporation was performed. On the day of the electroporation, 100 mΐ transfection solution (Nucleofector Kit V Solution, Lonza, Walkersville, MD) containing 5 pg of linearized expression vector of Example 1 was used to suspend 3X10 6 cells (early log phase). The cell /DNA mixture was then transferred to the cuvette of a Nucleofector 2b electroporation device, and program U-24 (Lonza) was used to electroporate the DNA into the cells. After electroporation, the cells were immediately transferred into the 24-well plate containing pre-warmed CD-CHO medium (without drug selection) and incubated in a humidified environment at 37°C with 5% CO2. After 24-hour incubation, the post-electroporation cells were plated into 96-well microplates at a density of 3000 cells/well in CD-CHO medium containing 25 mM L- methionine sulfoximine (MSX) and incubated for 2-3 weeks at 37°C with 95% humidity and 5% CO2. The successfully-transfected cell colonies were seen after 2-3 weeks incubation and the FVIII-expressing cells were selected using a one-stage clotting assay of the supernatant. Selected cell pools were then gradually expanded from 96-well plates to 6-well plates, and eventually to shaker flasks, for suspension culture. The selected cell pools were then subjected to limited dilution cloning at a cell density of 0.3 cell/well. FVIII expression levels in excess of 1000 IU/ml in a 1 L shake flask were determined with a one-stage clotting assay.

Example 3. FVIII Protein expression and purification

Once FVIII-expressing cell pools were obtained, the FVIII expression level was tested in fed-batch culture. Cells expressing both FVIII and truncate vWF-Fc variants were first adapted into ActiPro medium (Hyclone, Salt Lake City, Utah) and seeded at a cell density of 1 x 10 6 /ml. When the cell density reached 6 x 10 6 cells/ml, Cell Boost 7a/7b Supplement (Hyclone) was added every other day according to the manufacture’s recommendation. The cell density and the FVIII activity were checked regularly to monitor the cell growth and FVIII expression. After 14-days, the fed-batch culture was terminated, and the cell culture supernatant was collected by centrifuging the cell culture medium at 100 x g, and stored in - 80°C freezer.

For FVIII purification, the clarified supernatant was further processed with high speed centrifugation, then filtered through 0.45 pm filter. The supernatant was mixed with an equal volume of dilution buffer (40 mM Tris-HCl, pH 7.0, 150 mM NaCl). Using the AKTA Pure chromatography system, the diluted supernatant was loaded onto a HiTrap MabSelect Prism A column, and the column was then washed with 10 column volumes (CV) of wash buffer (20 mM Tris-HCl, pH 7.0, 150 mM NaCl). A FVIIISelect affinity ligand column (GE Healthcare) was connected in series with the Protein A column outlet. The elution buffer (20 mM Tris-HCl, pH 7.0, 0.3 M CaCh) was applied to the protein A column to separate the FVIII from bound vWF-Fc fusion complex. After washing, the connected column series were separated and FVIII elution buffer (20 mM histidine, 20 mM CaCh, 1.5 M NaCl and 0.02% Tween-80 in 50% ethylene glycol, pH 6.5) was used to elute the FVIII from the FVIIISelect column. The purified BDD-FVIII was then buffer exchanged into 0.3% (w/v) sucrose, 2.2% (w/v) glycine, 20 mM histidine, 220 mM NaCl, 25 mM CaCh and 0.008% Tween-80, pH 6.9 for further protein characterization.

The FVIII purification intermediate and final product are shown on lanes 4-6 of an SDS- PAGE gel (FIG. 7A) and corresponding Western Blot (FIG. 7B).

Example 4. Truncated vWF-Fc dimer and multimer form analysis

Four recombinant truncated vWF-Fc polypeptide constructs were evaluated for their ability to form dimers or high molecular weight complexes (i.e., multimers) by

electrophoretic analysis on a non-reducing 4% SDS-PAGE gel. After electrophoresis, the gel was stained with Coomassie Brilliant Blue (FIG. 8). The results indicated that the vWF-Fc variants, Pro-D’D3-Fc (Lane 2) and Del-DD3-Fc (Lane 3), both formed dimers of about 160 kD) and multimers (>180 kD), while the vWF-Fc variants that contain mutations, Pro- DD3mut-Fc (Lane 4) and Del-D’D3mut-Fc (Lane 5), only formed dimers of ca. 160 kD, due to the mutations that block multimer formation.

Example 5. Characterization of truncated vWF-Fc polypeptide binding affinity to B- domain-deleted (BDD)-FVIII

Recombinant protein constructs, corresponding to each of the truncated vWF-Fc variants, were expressed in CHO-K1 cells, and proteins were purified from supernatant using Protein A affinity chromatography. To characterize their respective FVIII binding affinities, equimolar amounts of the vWF-Fc variants were prepared in TBST buffer (20mM Tris, pH 7.6, 150 mM NaCl, 0.1% Tween-20) and directly coated onto 96-well microplates overnight at 4°C. After coating, plates were blocked with TBST-3% bovine serum albumin (BSA) buffer for 2 hours at room temperature (RT) and then incubated with increasing

concentrations (0-4000 pM) of recombinant FVIII (Prospec Bio, East Brunswick, NJ) for 1 hour at room temperature (RT). After equilibration and washing, bound FVIII was detected with a sheep anti-FVIII polyclonal antibody, followed by a secondary antibody against sheep IgG, that itself was conjugated to horseradish peroxidase. Blank values were subtracted from the absorbance measurements. The assay was performed in triplicates. The analysis of the binding assay showed that all four vWF-Fc fragments bound to recombinant BDD-FVIII with similar apparent affinity constants (about 500 pM) that is comparable to the binding affinity of full-length plasma-derived vWF to BDD-FVIII (Shiltagh et al., Blood, vol. 123, p. 4143- 4151, 2014).

Example 6. vWF variant D’D3mut-Fc improves the FVTTT purification recovery rate

As discussed above, mature, wild-type Pro-D’D3-Fc fusion protein results in multimer formation, and possibly protein aggregation, through the N-terminal disulfide bonds mediated by D3 domains, and C-terminal dimerization mediated by Fc domains. Such aggregation interferes with vWF-Fc binding to an Fc affinity matrix, like Protein A; in addition, FVIII elution from the FVIII/vWF-Fc complex is compromised by aggregate formation. By introducing mutations at amino acids C1099 and Cl 142, multimer assembly is interrupted, while dimer formation is promoted, thus leading to uniform vWF-Fc dimer formation. Therefore, both FVIII capture (in the form of a FVTTT/vWF-Fc complex) on Protein A matrix, and FVIII elution (from the captured FVIII/vWF-Fc complex), are improved.

The cell-culture medium from transfected cells co-expressing FVIII and either Pro- D’D3-Fc or Del-D’D3mut-Fc were purified using Protein A chromatography; the FVIII-vWF purification recovery rates are summarized in Table 2. The results show that the FVIII-vWF purification recovery rate is markedly higher when FVIII is co-expressed with Del-D’D3mut- Fc than FVIII is co-expressed with Pro-D’D3-Fc fusion (87% vs 64%). In addition, high column back-pressure was consistently observed during the FVIII purification using the Pro- D’D3-Fc variant, possibly due to the presence of high numbers of high-molecular weight multimers.

Table 2.

Example 7. Recombinant truncated vWF-Fc variants enhance the soluble FVIII expression

It has been suggested that 90% of the BDD-FVIII is bound to cell membrane when it is expressed alone in cell system; the reason for this is unknown (Kolind et al, J. Biotech., 2011, 151, pp. 357-362). This phenomenon may be explained as naturally the FVIII molecule is present in a non-covalent complex with, and circulates with, vWF in plasma, which blocked FVIII interaction with the cell membrane

To test whether co-expression of the truncated vWF-Fc fusion protein could increase recombinant FVIII expression, four purified vWF-Fc variants (Pro-D’D3-Fc, Pro-D’D3muts- Fc, Del-D’D3mut-Fc and Del-D’D3-Fc proteins), plasma vWF, and a negative control protein (HSA) were included in the assay.

The four truncated recombinant vWF-Fc variants were expressed in CHO cells and purified with Protein A chromatography. To demonstrate that truncated vWF-Fc variants, co expressed with BDD-FVIII, would increase the BDD-FVIII yield in cell culture by shielding BDD-FVIII from cell binding, equal masses of each of the four-recombinant truncated vWF- Fc variants, as well as full-length plasma-derived vWF (FL-vWF), were added to the supernatant of the cell line that expresses only BDD-FVIII; human serum albumin (HSA) was added as a negative control. FVIII activities of the cell culture supernatants were tested every 2 hours using a one-stage clotting assay for each sample. The results demonstrated that all four truncated vWF-Fc variants, as well as the plasma-derived, full-length vWF, can substantially increase FVIII yield in cell culture. Unexpectedly, the Del-D’D3mut-Fc fusion demonstrated the highest effect on FVIII expression (FIG. 9). Cells expressing only FVIII, or cell cultures to which HSA was added, did not show detectable increase in FVIII activity.

Example 8. Co-expression of truncated vWF-Fc fragments (Del-D’D3mut-Fc) with human FVIII completely blocked the FVIII binding to cell membrane

It has been recognized that B-domain-deleted FVIII significantly interacts with cell membranes during recombinant protein production, thereby lowering its apparent expression in cell culture.

To confirm this observation, cell culture medium from cells expressing only human BDD-FVIII were treated with 0.5 M NaCl (salt ions provide charge shielding/electrical double layer on the charged protein to modulate the nonspecific protein-surface interaction) for 5 min at room temperature and then centrifuged to harvest the supernatant. FVIII coagulation activity was measured using a one-stage clotting assay for both NaCl-treated and non-treated supernatant. The results showed that as much as 90% of secreted BDD-FVIII was bound to the cells. By contrast, when FVIII is co-expressed with vWF-Fc variant, Del- D’D3mut-Fc, addition of NaCl did not increase the cell culture supernatant FVIII activity, suggesting that the Del-D’D3mut-Fc completely blocked FVIII binding to the cell membrane, resulting in almost 100% secreted recombinant BDD-FVIII being released into the cell culture supernatant.

Example 9. Recombinant truncated vWF-Fc variant (Del-D’D3mut-Fc) enhanced the in vitro stability of FVIII in cell culture

Literature indicates that recombinant BDD-FVIII has poor in vitro stability in cell culture medium at room temperature (RT).

To determine if co-expression with vWF could increase the FVIII stability in cell culture, the stability of FVIII expressed either alone in cell culture, or co-expressed with Del- D’D3mut-Fc variant, was evaluated. Samples of cell culture media containing either FVIII or FVIII-Del-D’D3mut-Fc complex were tested at room temperature, and the FVIII activity were measured using a one-stage clotting assay at timed intervals (0, 4, 8, and 24 hours) for each sample. The results showed that, unexpectedly, FVIII, co-expressed with the Del- D’D3mut-Fc vWF variant, could maintain its activity unchanged for at least 24 hours at room temperature (FIG.10). Without the co-expression with this vWF-Fc variant, FVIII was not stable, and nearly 90% of its activity was lost at 24 hours (FIG.10).

Example 10. Co-expression of truncated vWF-Fc variant (Del-D’D3mut-Fc) stabilized the recombinant FVIII during the virus inactivation procedure

Cell cultures expressing only recombinant BDD-FVIII and cell cultures containing the BDD-FVIII co-expressed with Del-D’D3mut-Fc vWF variant were treated with the virus inactivation reagents (1% Triton X-100 and 0.3% Tri (n-butyl) phosphate) for 24 hours. The FVIII coagulation activity were monitored with one-stage clotting assay at timed intervals (0, 4, 8, and 24 hours after treating with inactivation reagent) for each sample. The results indicated that cell cultures expressing FVIII alone lost almost 90% of its activity after 8 hours of virus inactivation treatment. In contrast, the FVIII activity of the FVIII- vWF-Fc - expressing cell culture only lost about 40% during the same time period. The results indicated that the co-expressed vWF-Fc variant substantially stabilized the FVIII activation during the virus inactivation process (FIG. 11). This indicates that the Del-D’D3mut-Fc variant is remarkably stable under these conditions.

The invention, and the manner and process of making and using it, are now described in such full, clear, concise and exact terms as to enable any person skilled in the art to which it pertains, to make and use the same. It is to be understood that the foregoing describes preferred embodiments of the present invention and that modifications may be made therein without departing from the scope of the present invention as set forth in the claims. To particularly point out and distinctly claim the subject matter regarded as invention, the following claims conclude the specification.