Title of Invention

"A MODIFIED PARTIALLY B-DOMAINLESS PORCINE FACTOR VIII PROTEIN"

Abstract The invention relates to a modified B-domainless porcine factor VIII protein, to a DNA encoding the same, and to the use thereof for treatment of hemophilia.
Full Text The present Application is a Divisional Application out of Indian Patent Application No. IN/PCT/2002/00814/DEL dated 16.08.2002.
The present invention relates to a modified B-domainless form of porcine factor VIE protein, to a DNA encoding the same, and to the use thereof for treatment of hemophilia.
BACKGROUND OF THE INVENTION
Blood clotting begins when platelets adhere to the cut wall of an injured blood vessel at a lesion site. Subsequently, in a cascade of enzymatically regulated reactions, soluble fibrinogen molecules are converted by the enzyme thrombin to insoluble strands of fibrin that hold the platelets together in a thrombus. At each step in the cascade, a protein precursor is converted to a protease that cleaves the next protein precursor in the series. Cofactors are required at most of the steps.
Factor VIII circulates as an inactive precursor in blood, bound tightly and non-covalently to von Willebrand factor. Factor VIII is proteolytically activated by thrombin or factor Xa, which dissociates it from von Willebrand factor and activates its procoagulant function in the cascade. In its active from, the protein factor Villa is a cofactor that increases the catalytic efficiency of factor D( a towards factor X activation by several orders of magnitude.
People with deficiencies in factor VIII or antibodies against factor VIII who are not treated with factor VIII suffer uncontrolled internal bleeding that may cause a range of serious symptoms, from inflammatory reactions in joints to early death. Severe hemophiliacs, who number about 10,000 in the United States, can be treated with infusion of human factor VIII, which will restore the blood's normal clotting ability if administered with sufficient frequency and concentration. The classic definition of factor VIII, in fact, is that substance present in normal blood plasma that corrects the clotting defect in plasma derived from individuals with hemophilia A.
The development of antibodies ("inhibitors" or "inhibitory antibodies") that inhibit the activity of factor VIII is a serious complication in the management of patients with hemophilia. Autoantibodies develop in approximately 20% of patients with hemophilia A in response to therapeutic infusions of factor VIII. In previously untreated patients with hemophilia A who develop inhibitors, the inhibitor usually develops within one year of treatment. Additionally, autoantibodies that inactivate factor VIII occasionally develop in individuals with previously normal factor VIII levels. If the inhibitor titer is low enough, patients can be managed by increasing the dose of factor VIII. However, often the inhibitor titer is so high that it cannot be overwhelmed by factor VIII. An alternative strategy is to bypass the need for factor VIII during normal hemostasis using factor DC complex preparations (for example, KONYNE*, Proplex*) or recombinant human factor VIIa. Additionally, since porcine factor VIII usually has substantially less reactivity with inhibitors than human factor VIII, a partially purified, porcine factor VIII preparation (HYATE-.C) has been used. Many patients who have developed inhibitory antibodies to human factor VIII have been successfully treated with porcine factor VIII and have tolerated such treatment for long periods of time. However, administration of porcine factor "VIII is not a complete solution because inhibitors may develop to porcine factor "VIII after one or more infusions in some patients.
Several preparations of human plasma-derived factor VIII of varying degrees of purity are aVIIIlable commercially for the treatment of hemophilia A. These include a partially-purified factor VIII derived from the pooled blood of many donors that is heat- and detergent-
treated for viruses but contain a significant level of antigenic proteins; a monoclonal antibody-purified factor VIII that has lower levels of antigenic impurities and viral contamination; and recombinant human factor VIII, clinical trials for which are underway. Unfortunately, human, factor VIII is unstable at physiologic concentrations and pH, is present in blood at an extremely low concentration (0.2µg/ml plasma), and has low specific clotting activity. Public health concerns regarding the risk of viruses or other blood-borne contaminants have limited the usefulness of porcine factor VIII purified from porcine blood.
Hemophiliacs require daily replacement of factor VIII to prevent bleeding and me resulting deforming hemophilic arthropathy. However, supplies have been inadequate and problems in therapeutic use occur due to difficulty in isolation and purification, immunogenicity, and the necessity of removing the AIDS and hepatitis infectivity risk. The use of recombinant human factor VIII or partially-purified porcine factor VIII will not resolve all the problems.
The problems associated with the commonly used, commercially aVIIIlable, plasma-derived factor VIII have stimulated significant interest in the development of a better factor VIII product. There is a need for a more potent factor VIII molecule so that more units of clotting activity can be delivered per molecule; a factor VIII molecule that is stable at a selected pH and physiologic concentration; a factor VIII molecule that is less apt to cause production of inhibitory antibodies; and a factor VIII molecule that evades immune detection in patients who have already acquired antibodies to human factor VIII.
It is therefore an object of the present invention to provide a factor VIII that corrects hemophilia in a patient deficient in factor VIII or having inhibitors to human factor VIII.
It is a further object of the present invention to provide methods for treatment of hemophiliacs.
It is still another object of the present invention to provide a factor VIII that is stable at a selected pH and physiologic concentration
It is another object of the present invention to provide a factor VIII that has greater coagulant activity that human factor VIII
It is an additional object of the present invention to provide a factor VIII against which less antibody is produced
It is a further object of the invention to provide a method for making recombinant porcine factor VIII and specifically modified porcine factor VIII.
SUMMARY OF THE INVENTION
The determination of the entire DNA sequence encoding porcine factor VIII set forth herein has enabled, for the first time, the synthesis of full-length porcine factor VIII by expressing the DNA encoding porcine factor VIII in a suitable host cell, purified recombinant porcine factor VIII is therefore an aspect of the present invention. The DNA encoding each domain of porcine factor VIII as well as any specified fragment thereof, can be similarly expressed. Furthermore, porcine fVIII having all or part of the B domain deleted B-domainless porcine fVIII) is made aVIIIlable as part of the present invention, by expression DNA encoding porcine fVIII having a deletion of one or more codons of the B-domain
Accordingly the present invention relates to DNA encoding the amino acid sequence of POL 1212 as set forth m SEQ ID NO:38.
BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS
Figs 1A-1H together provide an aligned sequence comparison of the human, pig and mouse factor VIII acid sequences
DETAILED DESCRIPTION OF THE INVENTION Unless otherwise specified or. indicated, as used herein, "factor VIII denotes any functional factor VIII protein molecule from any mammal.
As used herein, "mammalian factor VIII includes factor VIII with amino acid sequence derived from any non-human mammal, unless otherwise specified. "Animal", as used herein, refers to pig and other non-human mammals.
A "fusion protein" or "fusion factor VIII or fragment thereof, as used herein, is the product of a hybrid gene in which the coding sequence for one protein is altered, for example, by joining part of it to the coding sequence for a second protein from a different gene in proper reading frame register such that uninterrupted transcription and translation of the joined segments can occur to produce a hybrid gene that encodes the fusion protein.
A "corresponding" nucleic acid or amino acid or sequence of either, as used herein, is one present at a site in a factor VIII molecule or fragment thereof that has the same structure and/or function as a site in the factor VIII molecule of another species, although the nucleic acid or amino acid number may not be identical. A DNA sequence "corresponding to" another factor VIII sequence substantially corresponds to such sequence, andhybridizes to the sequence of the designated SEQ ID NO. under stringent conditions. A DNA sequence "corresponding to" another factor VIII sequence also includes a sequence that results in the expression of a factor VIII or fragment thereof and would hybridize to the designated SEQ ID NO. but for the redundancy of the genetic code.
A "unique" amino acid residue or sequence, as used herein, refers to an amino acid sequence or residue in the factor VIII molecule of one species that is different from the homologous residue or sequence in the factor VIII molecule of another species.
"Specific activity," as used herein, refers to the activity that will correct the coagulation defect of human factor VIII deficient plasma. Specific activity is measured in units of clotting
activity per milligram total factor VIII protein in a standard assay in which the clotting time of human factor VIII deficient plasma is compared to that of normal human plasma. One unit of factor VIII activity is the activity present in one milliliter of normal human plasma. In the assay, the shorter the time for clot formation, the greater the activity of the factor VIII being assayed. Porcine factor VIII has coagulation activity in a human factor VIII assay.
"Expression" refers to the set of processes that occur whereby genetic information is utilized to yield a product. A DNA encoding the amino acid sequence of porcine factor VIII can be "expressed" within a mammalian host cell to yield porcine factor VIII protein. The materials, genetic structures, host cells and conditions which permit expression of a given DNA sequence to occur are well-known in the art and can be manipulated to affect the time and amount of expression, as well as the intra- or extra-cellular location of the expressed protein. For example, by including DNA encoding a signal peptide at the 5' end of the DNA encoding porcine factor VIII (the 5' end being, by convention, that end encoding the NH2 terminus of the protein) the expressed protein becomes exported from the interior of the host cell into the culture medium. ProVIIIing a signal peptide coding DNA in combination with the porcine factor "VIII coding DNA is advantageous because the expressed factor VIII is exported into the culture medium which simplifies the process of purification. A preferred signal peptide is a mammalian factor VIII signal peptide.
The human factor VIII cDNA nucleotide andpredicted amino acid sequences are shown in SEQ ID NOs: 1 and 2, respectively. Factor VIII is synthesized as an approximately 300 kDa single chain protein with, internal sequence homology that defines the "domain" sequence NHj-A1-A2-B-A3-C1-C2-COOH. In a factor VUI molecule, a "domain", as used herein, is a continuous sequence of amino acids that is defined by internal amino acid sequence identity and sites of proteolytic cleavage by thrombin. Unless otherwise specified, factor VUI domains include the following amino acid residues, when the sequences are aligned with the human amino acid sequence (SEQ ID NO:2): Al, residues Alal-Arg372; A2, residues Ser373-Arg740; B, residues Ser741-Argl648; A3, residues Serl690-He2032; CI, residues Aig2033-Asn2172; C2, residues Ser2173-Tyr2332. The A3-C1-C2 sequence includes residues Serl690-
Tyr2332. The remaining segment, residues Glul649-Argl689, is usually referred to as the factor VIII light chain activation peptide. Factor VIII is proteolytically activated by thrombin or factor Xa, which dissociates it from von Willebrand factor, forming factor Villa, which has procoagulant function. The biological function of factor VIIIa is to increase the catalytic efficiency of factor IXa toward factor X activation by several orders of magnitude. Thrombin-activated factor VIIIa is a 160 kDa A1/A2/A3-C1-C2 heterotrimer that forms a complex with factor IXa and factor X on the surface of platelets or monocytes. A "partial domain,, as used herein is a continuous sequence of amino acids forming part of a domain.
"Subunits" of human or animal factor VIII, as used herein, are the heavy and light chains of the protein. The heavy chain of factor VIII contains three domains, Al, A2, and B. The light chain of factor VIII also contains three domains, A3, C1, and C2.
The terms "epitope," "antigenic site," and "antigenic determinant," as used herein, are used synonymously and are defined as a portion of the human, or animal factor VIII or fragment thereof that is specifically recognized by an antibody. It can consist of any number of amino acid residues, and it can be dependent upon the primary, secondary, or tertiary structure of the protein.
The term "immunogenic site," as used herein, is defined as a region of the human or animal factor VIII, or fragment thereof, that specifically elicits the production of antibody to the factor VIII, or fragment, in a human or animal, as measured by routine protocols, such as immunoassay, e.g. EL2SA, or the Bethesda assay, described herein. It can consist of any number of amino acid residues, and it can be dependent upon the primary, secondary, or tertiary structure of the protein. In some embodiments, the hybrid or hybrid equivalent factor VIII or fragment thereof is nonimmunogenic or less immunogenic in an animal or human man human or porcine factor VIII.
"Factor VIII deficiency," as used herein, includes deficiency in clotting activity caused by production of defective factor VIII, by inadequate or no production of factor VIII, or by
partial or total inhibition of factor VIII by inhibitors. Hemophilia A is a type of factor VIII deficiency resulting from a defect in an X-linked gene and the absence or deficiency of the factor VIII protein it encodes.
As used herein, "diagnostic assays" include assays that in some manner utilize the antigen-antibody interaction to detect and/or quantify the amount of a particular antibody that is present in a test sample to assist in the selection of medical therapies. There are many such assays known to those of skill in the art. As used herein, human, porcine or modified porcine factor VIII DNA or fragment thereof and protein expressed therefrom, in whole or in part, can be substituted for the corresponding reagents in the otherwise known assays, whereby the modified assays may be used to detect and/or quantify antibodies to factor VIII. It is the use of these reagents, the factor VIII DNA or fragment thereof or protein expressed therefrom, that permits modification of known assays for detection of antibodies to human or animal factor VIII. Such assays include, but are not limited to ELISAs, iromunodifmsion assays, and immunoblots. Suitable methods for practicing any of these assays are known to those of skill in the art. As used herein, the factor VIII or fragment thereof that includes at least one epitope of the protein can be used as the diagnostic reagent. Examples of other assays in which human, porcine or modified porcine factor VIII or fragment thereof can be used include the Bethesda assay and anticoagulation assays.
The term "DNA encoding a protein, such as porcine factor VIII" means a polydeoxynucleic acid whose nucleotide sequence embodies coding information to a host cell for the amino acid sequence of the protein, e.g. porcine factor VIII, according to the known relationships of the genetic code.
The "expression product" of a DNA encoding a human or animal factor VIII or a modified factor VIII is the product obtained from expression of the referenced DNA in a suitable host cell, including such features of pre- or post-translational modification of protein encoded by the referenced DNA, including but not limited to glycosylation, proteolytic cleavage and the like. It is known in the art that such modifications can occur and can differ
somewhat depending upon host cell type and other factors, and can result in molecular isoforms of the product, with retention of procoagulant activity. See, e.g. Lind, P. etal., Eur. J. Biochem. 232:1927 (1995), incorporated herein by reference.
An "expression vector" is a DNA element, often of circular structure, having the ability to replicate autonomously in a desired host cell, or to integrate into a host cell genome and also possessing certain well-known features which permit expression of a coding DNA inserted into the vector sequence at the proper site and in proper orientation. Such features can include, but are not limited to, one or more promoter sequences to direct transcription initiation of the coding DNA and other DNA elements such as enhancers, polyadenylation sites and the like, all as well known in the art. The term "expression vector" is used to denote both a vector having a DNA coding sequence to be expressed inserted within its sequence, and a vector having the requisite expression control elements so arranged with respect to an insertion site that it can serve to express any coding DNA inserted into the site, all as well-known in the art. Thus, for example, a vector lacking a promoter can become an expression vector by the insertion of a promoter combined with a coding DNA.
GENERAL DESCRIPTION OF METHODS U.S. Patent 5,364,771 described the discovery of hybrid human/porcine factor VIII molecules having coagulant activity, m which elements of the factor VIII molecule of human or pig are substituted for corresponding elements of the factor VIII molecule of the other species. U.S. Patent 5,663,060 describes procoagulant hybrid human/ammal and hybrid equivalent factor VIII molecules, in which elements of the factor VIII molecule of one species are substituted for corresponding elements of the factor VIII molecule of the other species.
Since current information indicates that the 8 domain has no inhibitory epitope and has no known effect on factor VIII function, in some embodiments the B domain is wholly or partially deleted in the active hybrid or hybrid equivalent factor VIII molecules or fragments thereof (nB(-) factor WW) prepared by any of the methods described herein.
The human factor VIII gene was isolated and expressed in mammalian cells, as reported by Toole, J.J. et al. (1984) Nature 112:342-347 (Genetics Institute); Gitschier, J. et al.(1984) Nature 312:326-330 (Genentech); Wood, W.I. et al. (1984) Nature 312:330-337 (Genentech); Vehar, G.A. et al. (1984) Nature 112:337-342 (Genentech); WO 87/04187; WO 88/08035; WO 88/03558; U.S. Patent No. 4,757,006, and the amino acid sequence was deduced from cDNA. U.S. Patent No. 4,965,199 to Capon et al. discloses a recombinant DNA method for producing factor VIII in mammalian host cells and purification of human factor VOL Human factor VIII expression on CHO (Chinese hamster ovary) cells and BHKC (baby hamster kidney cells) has been reported. Human factor VIII has been modified to delete oart or all of the B domain (U.S. Patent No. 4,868,112), and replacement of the hurnan factor VIIIB domain with the human factor V B domain has been attempted (U.S. Patent No. 5,004,803). The cDNA sequence encoding human factor VIII and predicted amino acid sequence are shown in SEQ ID NOs:l and 2, respectively. In SEQ ID NO:l, the coding region begins at nucleotide position 208, the triplet GCC being the codon for amino acid number 1 (Ala) of the mature protein as given in SEQ ID NO:2.
Porcine factor VIII has been isolated from plasma [Fass, D.N. et al. (1982) Blood 59:594]. Partial amino acid sequence of porcine factor VIII corresponding to portions of the N-terminal light chain sequence having homology to ceruloplasmin and coagulation factor V were described by Church et al. (1984) Proc. Natl. Acad. Sci. E7&1 £1:6934. Toole, J.J. et al. (1984) Nature 312:342-347 described the partial sequencing of the N-terminal end of four amino acid fragments of porcine factor VIII but did not characterize the fragments as to their positions in the factor VIII molecule. The amino acid sequence of the B and .part of the A2 domains of porcine factor VIII were reported by Toole, J.J. et al. (1986) Proc. Natl. Acad. Sci, USA 83:5939-5942. The cDNA sequence encoding the complete A2 domain of porcine factor VIII and predicted amino acid sequence and hybrid human/porcine factor VJH having substitutions of all domains, all subunits, and specific amino acid sequences were disclosed in U.S. Patent 5,364,771 entitled "Hybrid Human/Porcine factor VIII issued on November 15, 1994, and in WO 93/20093 published October 14,1993. The cDNA sequence encoding the A2 domain of porcine factor VIII corresponding to residues 373-740 in mature human factor
VIII, as shown in SEQ ID NO: 1, and the predicted amino acid sequence are shown in SEQ ID NOs:3 and 4, respectively. More recently, the nucleotide and corresponding amino acid sequences of part of the Al domain lacking the first 198 amino acid and of the A2 domain of porcine factor VIII were reported in WO 94/11503, published May 26, 1994. The entire nucleotide sequence encoding porcine factor VIII, including the complete Al domain, activation peptide, A3, CI and C2 domains, as well as the encoded amino acid sequence, was finally obtained by LoUar, as disclosed inU.S.Patent5,859,204,issuedJanuary 12,1999, and in WO 97/49725, published December 31,1997, both incorporated herein by reference..
Both porcine and human factor VIII are isolated from plasma as a two subunit protein. The subunits, known as the heavy chain and light chain, are held together by a non-covalent bond that requires calcium or other divalent metal ions. The heavy chain of factor "VIII contains three domains, Al, A2, and B, which are linked covalently. The light chain of factor VIII also contains three domains, designated A3, C1, and C2. The B domain has no known biological function and canbe removed, or partially removed from the molecule proteolytically or by recombinant DNA technology methods without significant alteration in any measurable parameter of factor VIII. Human recombinant factor VIII has a similar structure and function to plasma-derived factor VII, though it is not glycosylated unless expressed in marnnaliag cells.
Both human and porcine activated factor VIII ("factor VIIIan) have three subunits due to cleavage of the heavy chain between the Al and A2 domains. This structure is designated A1/A2/A3-C1-C2. Human factor VIIIa is not stable under the conditions that stabilize porcine factor VIIIa, presumably because of the weaker association of the A2 subunit of human factor VIIIa. Dissociation of the A2 subunit of human and porcine factor VIIIa is associated with loss of activity in the factor VD3a molecule. Yakhyaev, A. et al. (1997) Blood 90::Suppl. 1, Abstract
#126, reported binding of A2 domain by low density lipoprotein receptor-related protein, suggesting that cellular uptake of A2 mediated by such binding acts to down-regulate factor VIII activity.
Expression of "B-domainless factor VIII is enhanced by including portions of the B-domain. The inclusion of those parts of the B domain designated "SQ" [Lind, P. et al (1995) supra] was reported to result in favorable expression. "SQ" constructs lack all of the human B domain except for 5 amino acids of the B domain N-terminus and 9 amino acids of the B domain C-teiminus.
The purified hybrid factor VIII or fragment thereof canbe assayed for immunoreactivity and coagulation activity by standard assays including, for example, the plasma-free factor VIII assay, the one-stage dotting assay, and the enzyme-linked immunosorbent assay using purified recombinant human factor VIII as a standard.
Other vectors, including both plasmid and eukaryotic viral vectors, may be used to express a recombinant gene construct in eukaryotic cells depending on the preference and judgment of the skilled practitioner (see, for example, Sambrook et al., Chapter 16). Other vectors and expression systems, including bacterial, yeast, and insect cell systems, can be used but are not preferred due to differences in, or lack of, glycosylation.
Recombinant factor VIII protein can be expressed in a variety of cells commonly used for culture and recombinant mammalian protein expression. In particular, a number of rodent cell lines have been found to be especially useful hosts for expression of large proteins. Preferred cell lines, aVIIIlable from the American Type Culture Collection, Rockville, MD, include baby hamster kidney cells, and Chinese hamster ovary (CHO) cells which are cultured using routine procedures and media.
The basis for the greater coagulant activity of porcine factor VIII appears to be the more rapid spontaneous dissociation of the human A2 subunit from human factor Villa than the porcine A2 subunit from porcine factor VIIIa. Dissociation of the A2 subunit leads to loss of activity, [Lollar, P. et al. (1990) /. Biol Chem. 265:1688-1692; Lollar, P. et al. (1992) J. Biol Chem. 262:23652-23657; Fay, P.J. et al. (1992) J. Biol Chem. 267:13246-13250],
body weight. Approximately 1 hour after each administration, the recovery of factor VIII from blood samples is measured in a one-stage coagulation assay. Samples are taken again approximately 5 hours after infusion, and recovery is measured. Total recovery and the rate of disappearance of factor VIII from the samples is predictive of the antibody titer and inhibitory activity. If the antibody titer is high, factor VIII recovery usually cannot be measured. The recovery results are compared to the recovery results in patients treated with plasma-derived human factor VIII, recombinant human factor VIII, plasma-derived porcine factor VIII, and other commonly used therapeutic forms of factor VIII or factor VIII substitutes.
After identification of clinically significant epitopes, recombinant factor VIII molecules can be expressed that have less than or equal cross-reactivity compared with plasma-derived porcine factor VIII when tested in vitro against a broad survey of inhibitor plasmas. Additional mutagenesis in epitopic regions can be done to reduce cross-reactivity. Reduced cross-reactivity, although desirable, is not necessary to produce a product that may have advantages over the existing plasma-derived porcine factor VIII concentrate, which can produce side effects due to contaminant porcine proteins or contaminant infectious agents such as viruses or prions. A recombinant porcine or modified porcine factor VIII molecule will not contain foreign porcine proteins.
Diagnostic Assays..
The factor VIII cDNA and/or protein expressed therefrom, in whole or in part, can be used in assays as diagnostic reagents for the detection of inhibitory antibodies to human or animal factor VIII or modified animal VIII in substrates, including, for example, samples of serum and body fluids of human patients with factor VIII deficiency. These antibody assays include assays such as ELISA assays, immunoblots, radioimmunoassays, immunodiffusion assays, and assay of factor VIII biological activity (e.g., by coagulation assay). Techniques for preparing these reagents and methods for use thereof are known to those skilled in the art For example, an immunoassay for detection of inhibitory antibodies in a patient serum sample can include reacting the test sample with a sufficient amount of the factor VIII to be tested that
a detectable complex can be formed with the inhibitory antibodies in the sample of the test factor VIII is indeed antigenic.
Nucleic acid and amino acid probes can be prepared based on the sequence of the hybrid factor VIII cDNA or protein molecule or fragments thereof. In some embodiments, these can be labeled using dyes or enzymatic, fluorescent, chemiluminescent, or radioactive labels that are commercially aVIIIlable. The amino acid probes can be used, for example, to screen sera or other body fluids where the presence of inhibitors to human, animal, or hybrid human/animal factor VIII is suspected. Levels of inhibitors can be quantitated in patients and compared to healthy controls, and can be used, for example, to determine whether a patient with a factor VIII deficiency can be treated with an animal or modified animal factor VIII The cDNA probes can be used, for example, for research purposes in screening DNA libraries.
Pharmaceutical Compositions.
Pharmaceutical compositions containing recombinant porcine or modified porcine factor VIII, alone or in combination with appropriate pharmaceutical stabilization compounds, delivery vehicles, and/or carrier vehicles, are prepared according to known methods, as described in Remington's Pharmaceutical Sciences by E.W. Martin.
In one preferred embodiment, the preferred carriers or delivery vehicles for intravenous infusion are physiological saline or phosphate buffered saline.
In another preferred embodiment, suitable stabilization compounds, delivery vehicles, and carrier vehicles include but are not limited to other human or animal proteins such as albumin.
Phospholipid vesicles or liposomal suspensions are also preferred as pharmaceutically acceptable carriers or delivery vehicles. These can be prepared according to methods known to those skilled in the art and can contain, for example, phosphatidylserme/-phosphatidylcholine or other compositions of phospholipids or detergents that together impart
Factor VIII molecules with reduced immunoreactivity:
Epitopes that are immunoreactive with antibodies that inhibit the coagulant activity of factor VIII ("inhibitors" or "inhibitory antibodies") have been characterized based on known structure-function relationships in factor VIII. Presumably, inhibitors could act by disrupting any of the macromolecular interactions associated with the domain structure of factor VIII or its associations with von Willebrand factor, thrombin, factor Xa, factor IXa, or factor X. However, most inhibitory antibodies to human factor VIII act by binding to epitopes located in the 40 kDa A2 domain or 20 kDa C2 domain of factor VIII, disrupting specific functions associated with these domains, as described by Fulcher et al. (1985) Proc. Natl Acad. Sci USA 82:7728-7732; and Scandella et al. (1988) Proc. Natl. Acad. Sci. USA .85:6152-6156. In addition to the A2 and C2 epitopes, there may be a third epitope in the A3 or CI domain of the light chain of factor VIII, according to Scandella et al. (1993) Blood 82:1767-1775. The significance of this putative third epitope is unknown, but it appears to account for a minor fraction of the epitope reactivity in factor VIII.
Anti-A2 antibodies block factor X activation, as shown by Lollar et al. (1994) /. Gin. Invest. 9.3:2497-2504. Previous mapping studies by deletion mutagenesis described by Ware et al. (1992) Blood Coagid. Fibrinolysis 3:703-716, located the A2 epitope to witbina 20 kDa region of the NH2-terminal end of the 40 kDa A2 domain. Competition immunoradiometric assays have indicated that A2 inhibitors recognize either a common epitope or narrowly clustered epitopes, as described by Scandella et al. (1992) Throm. Haemostas&l-M5-61l, and as demonstrated in U.S. Patent 5,859,204.
Animal or modified animal factor VIII molecules can be tested in humans for their reduced antigenicity and/or immunogenicity in clinical trials. In one type of trial, designed to determine whether the factor VIII is immunoreactive with inhibitory antibodies, factor VIII is administered, preferably by intravenous infusion, to approximately 25 patients having factor VIII deficiency who have antibodies that inhibit the coagulant activity of therapeutic human factor VIII. The dosage of the animal or modified animal factor VIII is in a range between 5 and 50 Units/kg body weight, preferably 10-50 Units/kg, and most preferably 40 Units/kg
a negative charge to the surface, since factor VIII binds to negatively charged phospholipid membranes. Liposomes may be prepared by dissolVIIIg appropriate lipid(s) (such as stearoyl phosphatidyl etnanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaVIIIg behind a thin film of dried lipid on the surface of the container. An aqueous solution of the hybrid factor VIII is then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
The recombinant porcine or modified porcine factor VIII can be combined with other suitable stabilization compounds, delivery vehicles, and/or carrier vehicles, including vitamin K dependent clotting factors, tissue factor, and von Willebrand factor (VIIIf) or a fragment of VIIIf that contains the factor VIII binding site, and polysaccharides such as sucrose.
Recombinant porcine or modified porcine factor VIII can also be delivered by gene therapy in the same way that human factor VIII can be delivered, using delivery means such as retroviral vectors. This method consists of incorporation of the desired factor VIII construct cDNA into human cells that are transplanted directly into a factor VIII deficient patient or that are placed in an implantable device, permeable to the factor VIII molecules but impermeable to cells, that is then transplanted. The preferred method will be retroviral-mediated gene transfer. In this meuiod, an exogenous gene (e.g., a factor VIII cDNA) is cloned into the genome of a modified retrovirus. The gene is inserted into the genome of the host cell by viral rnachinery where it will be expressed by the cell. The retroviral vector is modified so that it will not produce virus, preventing viral infection of the host. The general principles for this type of therapy are known to those skilled in the art and have been reVIIIwed in the literature [e.g., Kohn, D.B. etal. (1989) Transufusion 29:812-820].
Porcine or modified porcine factor VIII can be stored bound to VIIIf to increase the half-life and shelf-life of the hybrid molecule. Additionally, lyophilization of factor VIII can improve the yields of active molecules in the presence of vWf . Current methods for storage
of human and animal factor VIII used by commercial suppliers can be employed for storage of recombinant factor VIII These methods include: (1) lyophilization of factor VIII in a partially-purified state (as a factor VIII "concentrate" that is infused without further purification); (2) inimunoaffinity-purification of factor VIII by the Zimmerman method and lyophilization in the presence of albumin, which stabilizes the factor VIII; (3) lyophilization of recombinant factor VIII in the presence of albumin.
Additionally, porcine or modified porcine factor VIII has been found to be indefinitely stable at 4° C in 0.6 M NaCl, 20 mM MES, and 5 mM CaCl2 at pH 6.0 and also can be stored frozen in these buffers and thawed with minimal loss of activity.
Methods of Treatment.
Recombinant porcine or modified porcine factor VIII is used to treat uncontrolled bleeding due to factor VIII deficiency (e.g., mtraarticular, intracranial, or gastrointestinal hemorrhage) in hemophiliacs with and without inhibitory antibodies and in patients with acquired factor VTH deficiency due to the development of inhibitory antibodies. The active materials are preferably administered intravenously.
Additionally, recombinant porcine or modified porcine factor VIII can be adrninistered by transplant of cells genetically engineered to produce the protein by implantation of a device containing such cells, as described above.
In a preferred embodiment, pharmaceutical compositions of recombinant porcine or modified porcine factor VIII alone or in combination with stabilizers, delivery vehicles, and/or carriers are infused into patients intravenously according to the same procedure that is used for infusion of human or animal factor VIII.
The treatment dosages of recombinant porcine or modified porcine factor VIII composition that must be administered to a patient in need of such treatment will vary depending on the severity of the factor VIII deficiency. Generally, dosage level is adjusted in
frequency, duration, and units in keeping with the severity and duration of each patient's bleeding episode. Accordingly, the factor VIII is included in a pharmaceutically acceptable carrier, delivery vehicle, or stabilizer in an amount sufficient to deliver to a patient a therapeutically effective amount of the protein to stop bleeding, as measured by standard clotting assays.
Factor VIII is classically defined as that substance present in normal blood plasma that corrects the clotting defect in plasma derived from indiVIIIuals with hemophilia A. The coagulant activity in vitro of purified and partially-purified forms of factor VIII is used to calculate the dose of factor VIII for infusions in human patients and is a reliable indicator of activity recovered from patient plasma and of correction of the in vivo bleeding defect. There are no reported discrepancies between standard assay of novel factor VIII molecules in vitro and their behavior in the dog infusion model or in human patients, according to Lusher, J.M. et al. 328 New Engl J. Med. 328:453-459; Pittman, D.D. et al. (1992) Blood79:389-397; and Brinkhous et al. (1985) Proc. Natl. Acad. Sci. 82:8752-8755.
Usually, the desired plasma factor VIII activity level to be achieved in the patient through administration of the recombinant porcine or modified porcine factor VIII is in the range of 30-100 % of normal. In a preferred mode of administration of the therapeutic factor VIII, the composition is given intravenously at a preferred dosage in the range from about 5 to 50 units/kg body weight, more preferably in a range of 10-50 units/kg body weight, and most preferably at a dosage of 20-40 units/kg body weight; the interval frequency is in the range from about 8 to 24 hours (in severely affected hemophiliacs); and the duration of treatment in days is in the range from 1 to 10 days or until the bleeding episode is resolved. See, e.g., Roberts, H.R., and M.R. Jones, "Hemophilia and Related Conditions - Congenital Deficiencies of Prothrombin (Factor II, Factor V, and Factors VII to XII)," Ch. 153,1453-1474,1460, in Hematology. Williams, W. J., et al., ed. (1990). Patients with inhibitors may require a different amount of recombinant porcine or modified porcine factor VIII than their previous form of factor VIII. For example, patients may require less recombinant porcine or modified porcine factor VIII because of its higher specific activity than human factor VIII and
its decreased antibody reactivity. As in treatment with human or plasma-derived porcine factor VIII, the amount of therapeutic factor VIII infused is defined by the one-stage factor VIII coagulation assay and, in selected instances, in vivo recovery is determined by measuring the factor VIII in the patient's plasma after infusion. It is to be understood that for any particular subject, specific dosage regimens should be adjusted over time according to the indiVIIIual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed composition.
Treatment can take the form of a single intravenous administration of the composition or periodic or continuous adrninistration over an extended period of time, as required. Alternatively, therapeutic factor VIII can be administered subcutaneously or orally with liposomes in one or several doses at varying intervals of time.
Recombinant porcine or modified porcine factor VIII can also be used to treat uncontrolled bleeding due to factor VIII deficiency in hemophiliacs who have developed antibodies to human factor VIII. In this case, coagulant activity that is superior to mat of human or animal factor VIII alone is not necessary. Coagulant activity that is inferior to that of human factor VIII (i.e., less than 3,000 units/mg) will be useful if that activity is not neutralized by antibodies in the patient's plasma.
It has been demonstrated herein that recombinant porcine and modified porcine factor VIII's can differ in specific activity from human factor VIII. Factor VIII proteins having greater procoagulant activity from human factor VIII are useful in treatment of hemophilia because lower dosages will be required to correct a patient's factor VIII deficiency. Factor VIII's having lower procoagulant activity than human factor VIII are also suitable for therapeutic use provided they have at least 1 % of specific activity compared to normal human factor VIII. A factor VIII of the present invention having procoagulant activity is therefore defined as having at least 1 % of the specific activity of human factor VIII.
The recombinant porcine or modified porcine factor VIII molecule and the methods for isolation, characterization, making, and using it generally described above will be further understood with reference to the following non-limiting examples.
Example 1: Assay of porcine factor VIII and hybrid human/porcine factor VIII.
Porcine factor "VIII has more coagulant activity than human factor VIII, based on specific activity of the molecule. This conclusion is based on the use of appropriate standard curves that allow human porcine factor VIII to be fairly compared. Coagulation assays are based on me ability of factor VIII to shorten the clotting time of plasma derived from a patient with hemophilia A. Two types of assays were employed: the one-stage and the two stage assay.
In the one-stage assay, 0.1 ml hemophilia A plasma (George King Biomedical, Inc.) was incubated with 0.1 ml activated partial thromboplastin reagent (APTT) (Organon Teknika) and 0.01 ml sample or standard, consisting of diluted, titrated normal human plasma, for 5 min at 37°C in a water bath. Incubation was followed by addition of 0.1 ml 20 mM CaCl2, and the time for development of a fibrin clot was determined by visual inspection.
A unit of factor VIII is defined as the amount present in 1 ml of titrated normal human plasma. With human plasma as the standard, porcine and human factor VIII activity were compared directly. Dilutions of the plasma standard or purified proteins were made into 0.15 M NaCl, 0.02 M HEPES, pH 7.4. The standard curve was constructed based on 3 or 4 dilutions of plasma, the highest dilution being 1/50, and on log10 clotting time plotted against log10 plasma concentration, which results in a linear plot. The units of factor "VIII in an unknown sample were determined by interpolation from the standard curve.
The one-stage assay relies on endogenous activation of factor VIII by activators formed in the hemophilia A plasma, whereas the two-stage assay measures the procoagulant activity of preactivated factor "VIII. In the two-stage assay, samples containing factor VIII that had been reacted with thrombin were added to a mixture of activated partial thromboplastin and
human hemophilia A plasma that had been preincubated for 5 min at 37 °C. The resulting clotting times were then converted to units/ml, based on the same human standard curve described above. The relative activity in the two-stage assay was higher than in the one-stage assay because the factor VIII had been preactivated.
Example 2: Characterization of the functional difference between human andporcine factor VIII.
The isolation of porcine and human plasma-derived factor VIII and human recombinant
factor VIII have been described in the literature in Fulcher, C.A. et al. (1982) Proc. Natl
Acad. Sci. USA 79:1648-1652; Toole et al. (1984) Nature 312:342-347 (Genetics Institute);
Gitschier et al. (1984) Nature 312:326-330 (Genentech); Wood et al. (1984) Nature 312:330-
337 (Genentech); Venar et al. 312 Nature 312:337-342 (Genentech); Fass et al. (1982) Blood
59:594; Toole et al. (1986) Proc. Natl Acad. Sci. USA 83:5939-5942. This can be
accomplished in several ways. All these preparations are similar in subunit composition,
although there is a functional difference in stability between human and porcine factor VIII.
For comparison of human recombinant and porcine factor VIII, preparations of highly-purified humanrecombinant factor VIII (Cutter Laboratories, Berkeley, CA) and porcine factor VIII [immunopurified as described in Fass et al. (1982) Blood 59:594] were subjected to high-pressure liquid chromatography (HPLC) over a Mono Q™ (Pharmacia-LKB, Piscataway, NJ) anion-exchange column (Pharmacia, Inc.). The purposes of the Mono Q™ HPLC step were elimination of minor impurities of exchange of human and porcine factor VIII into a common buffer for comparative purposes. VIIIls containing 1000-2000 units of factor VIII were reconstituted with 5 ml H20. Hepes (2 M at pH 7.4) was then added to a final concentration of 0.02 M. Factor VIII was applied to a Mono Q™ HR 515 column equilibrated in 0.15 M NaCl, 0.02 M Hepes, 5mM CaCl2, at pH 7.4 (Buffer A plus 0.15 M NaCl); washed with 10 mlBufferA + 0.15MNaCl; and eluted with a 20 ml linear gradient, 0.15 M to 0.90 M NaCl in Buffer A at a flow rate of 1 mVrnin.
For comparison of human plasma-derived factor VIII (purified by Mono Q™ HPLC) and porcine factor VIII, immunoafrimty-purified, plasma-derived porcine factor VIII was
diluted 1:4 with 0.04 M Hepes, 5 mM CaCl2, 0.01% Tween-80, at pH 7.4, and subjected to Mono Q™ HPLC under the same conditions described in the previous paragraph for human factor VIII. These procedures for the isolation of human and porcine factor VIII are standard for those skilled in the art.
Column fractions were assayed for factor VIII activity by a one-stage coagulation assay. The average results of the assays, expressed in units of activity per A280 of material, are given in Table n, and indicate that porcine factor VII3I has at least six times greater activity than human factor VIII when the one-stage assay is used.
TABLE II
COMPARISON OF HUMAN AND PORCINE FACTOR VIII
COAGULANT ACTIVITY
(Table Removed)
Example 3: Comparison of the stability of human and porcine factor VUI.
The results of the one-stage assay for factor VIII reflect activation of factor VIII to lactor VIIIa in the sample and possibly loss of formed factor "VIIIa activity. A direct comparison of the stability of human and porcine factor VIII was made. Samples from Mono Q™ HPLC (Pharmacia, Inc., Piscataway, N.J.) were diluted to the same concentration and buffer composition and reacted with thrombin. At various times, samples were removed for two-stage coagulation assay. Typically, peak activity (at 2 min) was 10-fold greater for porcine than human factor VIIIa, and the actiVIIIes of both porcine and human factor VIIIa subsequently decreased, with human factor VIIIa activity decreasing more rapidly.
Generally, attempts to isolate stable human factor VIIIa are not successful even when conditions that produce stable porcine factor VIIIa are used. To demonstrate this, Mono Q™ HPLC-purified human factor VIII was activated with thrombin and subjected to Mono S™
cation-exchange (Pharmacia, Inc.) HPLC under conditions that produce stable porcine factor Villa, as described by Lollar et al. (1989) Biochemistry 28:666.
Human factor VIII, 43 /tg/ml (0.2 /zM) in 0.2 M NaCl, 0.01 M Hepes, 2.5 mM CaCl2, at pH 7.4, in 10 ml total volume, was reacted with thrombin (0.036 /iM) for 10 min, at which time FPR-CH2C1 D-phenyl-prolyl-arginyl-chloromethyl ketone was added to a concentration of 0.2 [M for irreversible inactivation of thrombin. The mixture then was diluted 1:1 with 40 mM 2-(N-morpholino) ethane sulfonic acid (MES), 5 mM CaCl2, at pH 6.0, and loaded at 2 ml/min onto a Mono S™ HR 5/5 HPLC column (Pharmacia, Inc.) equilibrated in 5 mM MES, 5 mM CaCl2, at pH 6.0 (Buffer B) plus 0.1M NaCl. Factor Villa was eluted without column washing with a 20 ml gradient from 0.1 M NaCl to 0.9 M NaCl in Buffer B at 1 ml/min.
The fraction with coagulant activity in the two-stage assay eluted as a single peak under these conditions. The specific activity of the peak fraction was approximately 7,500 U/A280. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) of the Mono S™ factor VIIIa peak, followed by silver staining of the protein, revealed two bands corresponding to a heterodimeric (A3-C1-C2/A1) derivative of factor VIII. Although the A2 fragment was not identified by silver staining under these conditions because of its low concentration, it was identified as a trace constituent by 125I-labeling.
In contrast to the results with human factor VIII, porcine factor VIIIa isolated by Mono S™ HPLC under the same conditions had a specific activity 1.6 x 10d U/A280. Analysis of porcine factor Villa by SDS-PAGE revealed 3 fragments corresponding to Al, A2, and A3C1-C2 subunits, demonstrating that porcine factor VIIIa possesses three subunits.
The results of Mono S™ HPLC of human thrombin-activated factor VIII preparations at pH 6.0 indicate that human factor VIIIa is labile under conditions that yield stable porcine factor VIIIa. However, although trace amounts of A2 fragment were identified in the peak fraction, determination of whether the coagulant activity resulted from small amounts of
heterotrimeric factor Villa or from heterodimeric factor Villa that has a low specific activity was not possible from this method alone.
A way to isolate human factor VIIIa before it loses its A2 subunit is desirable to resolve this question. To this end, isolation was accomplished in a procedure that involves reduction of the pH of the Mono S™ buffers to pH 5. Mono Q™-purified human factor VIII (0.5 mg) was diluted with H20 to give a final composition of 0.25 mg/ml (1 /an) factor VIII in 0.25 M NaCl, 0.01 M Hepes, 2.5 mM CaCl2, 0.005% Tween-80, atpH 7.4 (total volume 7.0 ml). Thrombin was added to a final concentration of 0.072 pm and allowed to react for 3 min. Thrombin was then inactivated with FPR-CH2C1 (0.2 pan). The mixture then was diluted 1:1 with 40 mM sodium acetate, 5 mM CaCl2, 0.01% Tween-80, at pH 5.0, and loaded at 2 ml/min onto a Mono S™ HR 5/5 HPLC column equilibrated in 0.01 M sodium acetate, 5 mM CaCl2, 0.01% Tween-80, at pH 5.0, plus 0.1 M NaCl. Factor Villa was eluted without column washing with a 20 ml gradient from 0.1M NaCl to 1.0 M NaCl in the same buffer at 1 ml/min. This resulted in recovery of coagulant activity in a peak that contained detectable amounts of the A2 fragment as shown by SDS-PAGE and silver staining. The specific activity of the peak fraction was tenfold greater than that recovered at pH 6.0 (75,000 U/A280 v, 7,500 U/A280). However, in contrast to porcine factor VIIIa isolated at pH 6.0, which is indefinitely stable at 4°C, human factor VIIIa activity decreased steadily over a period of several hours after elution from Mono S™. Additionally, the specific activity of factor VIIIa purified at pH 5.0 and assayed immediately is only 5 % that of porcine factor Villa, indicating that substantial dissociation occurred prior to assay.
These results demonstrate that both human and porcine factor VIIIa are composed of three subunits (Al, A2, and A3-C1-C2). Dissociation of the A2 subunit is responsible for the loss of activity of both human and porcine factor VIIIa under certain conditions, such as physiological ionic strength, pH, and concentration. The relative stability of porcine factor VIIIa under certain conditions is because of stronger association of the A2 subunit.
Example 4: Isolation and sequencing of DNA encoding theA2 domain ofporcinefactor VIII. Only the nucleotide sequence encoding the B domain and part of the A2 domain of porcine factor VIII has been sequenced previously (Toole et al. (1986) Proc. Natl Acad. Sci. USA 83:5939-5942]. The cDNA and predicted amino acid sequences (SEQ ID NOs: 3 and 4, respectively) for the entire porcine factor VIII A2 domain are disclosed herein.
The porcine factor VIII A2 domain was cloned by reverse transcription of porcine spleen total RNA and PCR amplification; degenerate primers based on the known human factor VIII cDNA sequence and an exact porcine primer based on a part of the porcine factor VIII sequence were used. A 1 kb PCR product was isolated and amplified by insertion into a Bluescript™ (Stratagene) phagemid vector.
The porcine A2 domain was completely sequenced by dideoxy sequencing. The cDNA and predicted amino acid sequences are as described in SEQ ID NOs: 3 and 4, respectively.
Example 5: Complete sequence of DNA encoding porcine factor VIII.
Klenow fragment, phosphorylated Clal linkers, NotI linkers, T4 ligase, and Taq DNA polymerase were purchased from Promega (Madison, Wisconsin). Polynucleotide kinase was purchased from Life Technologies, Inc., Gaithersburg, Maryland. y32P-ATP (Redivue, >5000Ci/mmol) was purchased from Amersham. pBluescript IIKS- and E. coli Epicurean XLl-Blue cells were purchased from Stratagene (La Jolla, California). Synthetic oligonucleotides were purchased from Life Technologies, Inc. or Cruachem, Inc. 5'-phosphorylated primers were used when PCR products were produced for cloning purposes. Nucleotide (nt) numbering of oligonucleotides used as primers for polymerase chain reaction (PCR) amplification of porcine fVIII cDNA or genomic DNA uses the human fVIII cDNA as reference (Wood et al. (1984) supra).
Porcine spleen total RNA was isolated by acid guanidinium thiocyanate-phenol-chloroform extraction [Chomc2ynski et al. (1987) Anal Biochem. 162:156-159]. Porcine cDNA was prepared from total spleen RNA using Moloney murine leukemia virus reverse
transcriptase (RT) and random hexamers to prime the reaction (First-Strand cDNA Synthesis Kit, Pharmacia Biotech) unless otherwise indicated. RT reactions contained 45 mM Tris-Cl, pH 8.3,68 mM KC1,15 mM DTT, 9 mM MgCl2l 0.08 mg/ml boVIIIe serum albumin and 1.8 mM deoxynucleotide triphosphate (dNTP). Porcine genomic DNA was isolated from spleen using a standard procedure (Strauss, W.M. (1995) In Current Protocols in Molecular BioIogy F. M. Ausubel et al, editors, John Wiley & Sons, pp. 2.2.1-2.2.3). Isolation of DNA from agarose gels was done using Geneclean H (Bio 101) or Quiex II Gel Extraction Kit (Qiagen).
PCR reactions were done using a Hybaid OmniGene thermocycler. For PCR reactions employing Taq DNA polymerase, reactions included 0.6 mM MgCl2,0.2 mM dNTPs ,0.5 µM oligonucleotide primers, 50 U/ml polymerase and 0.1 volume of first strand cDNA reaction mix. Except where indicated otherwise, PCR products were gel purified, blunt-ended with Klenow fragment, precipitated with ethanol, and either ligated to the EcoRV site of dephosphorylated pBluescript II KS- or ligated with phosphorylated Clal linkers using T4 ligase, digested with Clal, purified by Sepbacryl S400 chromatography, and ligated to Clal-cut, dephosphorylated pBluescript II KS-. Ligations were done using T4 DNA ligase (Rapid DNA ligation kit, Boehringer Mannheim) except where indicated otherwise. Insert-containing pBluescript II KS- plasmids were used to transform E. coli Epicurean XLl-BIue cells.
Sequencing of plasmid DNA was done using an Applied Biosystems 373a automated DNA sequencer and the PRISM dye terminator kit or manually using Sequenase v. 2.0 sequencing kit (Amersham Corporation). Direct sequencing of PCRproducts, including 32P-end labelling of oligonucleotides was done using a cycle sequencing protocol (dsDNA Cycle Sequencing System, Life Technologies).
Isolation of porcine fVIII cDNA clones containing 5' UTR sequence, signal peptide and Al domain codons.
The porcine fVIII cDNA 5' to the A2 domain was amplified by nested RT-PCR of female pig spleen total RNA using a 5' rapid amplification of cDNA ends (5'-RACE) protocol (Marathon cDNA Amplification, Clontech, Version PR55453). This included first strand
cDNA synthesis using a lock-docking oligo(dT) primer [Borson, N.D. et al. (1992) PCR
Methods Appl. 2:144-148], second strand cDNA synthesis using E. coli DNA polymerase I,
and ligation with a 5' extended double stranded adaptor, SEQ ID NO:5
5'-CTA ATA CGA CTC ACT ATA GGG CTC GAG CGG CCG CCC GGG CAG GT-3
3'-H2N-CCCGTCCA-P04-5' whose short strand was blocked at the 3' end with an amino group to reduce non-specific PCR
priming and which was complementary to the 8 nucleotides at the 3' end (Siebert, P.D., et al. (1995) Nucleic. Acids. Res. 221:1087-1088). The first round of PCR was done using an adaptor-specific oligonucleotide, SEQ ID NO:6 5 '-CCA TCC TAA TAC GAC TCA CTA TAG GGC-3' (designated API) as sense primer, and a porcine fVIII A2 domain specific oligonucleotide SEQ ID NO:7 5'-CCA TTG ACA TGA AGA CCG TTT CTC-3' (nt 2081-2104) as antisense primer. The second round of PCR was done using a nested, adaptor-specific oligonucleotide, SEQ ID NO:8 5"-ACT CAC TAT AGG GCT CGA GCG GC-3' (designated AP2) as sense primer, and a nested, porcine A2 domain-specific oligonucleotide SEQ ID NO:9 5 '-GGG TGC AAA GCG CTG ACA TCA GTG-3' (nt 1497-1520) as antisense primer. PCR was carried out using a commercial kit (Advantage cDNA PCR core kit) which employs an antibody-mediated hot start protocol [Kellogg, D.E. et al. (1994) BioTechniques 16:1134-1137]. PCR conditions included denaturation at 94°C for 60 sec, followed by 30 cycles (first PCR) or 25 cycles (second PCR) of denaturation for 30 sec at 94°C, annealing for 30 sec at 60°C and elongation for 4 min at 68°C using tube temperature control. This procedure yielded a prominent =1.6 kb product which was consistent with amplification of a fragment extending approximately 150 bp into the 5' UTR. The PCR product was cloned into pBluescript using ClaI linkers. The inserts of four clones were sequenced in both directions.
The sequence of these clones included regions corresponding to 137 bp of the 5' UTR, the signal peptide, the Al domain and part of the A2 domain. A consensus was reached in at least 3 of 4 sites. However, the clones contained an average of 4 apparent PCR-generated mutations, presumably due to the multiple rounds of PCR required to generate a clonable product. Therefore, we used sequence obtained from the signal peptide region to design a sense strand phosphorylated PCR primer, SEQ ID NO.10 5'-CCTCTCGAG CCA CCA TGT CGA GCC ACC ATG CAG CTA GAG CTC TCC ACC TG-3', designated RENEOPIGSP, for
synthesis of another PCR product to confirm the sequence and for cloning into an expression vector. The sequence in bold represents the start codon. The sequence 5' to this represents sequence identical to that 5' of the insertion site into the mammalian expression vector ReNeo used for expression of fVIII (Lubin et al. (1994) supra). This site includes an Xhol cleavage site (underlined). RENEOPIGSP and the nt 1497-1520 oligonucleotide were used to prime a Taq DNA polymerase-mediated PCR reaction using porcine female spleen cDNA as a template. DNA polymerases from several other manufacturers failed to yield a detectable product. PCR conditions included denaturation at 94°C for four min, followed by 35 cycles of denaturation for 1 min at 94°C, annealing for 2 min at 55°C and elongation for 2 min at 72°C, followed by a final elongation step for 5 min at 72°C. The PCR product was cloned into pBluescript using Clal linkers. The inserts of two of these clones were sequenced in both directions and matched the consensus sequence.
Isolation of porcine fVIII cDNA clones containing A3. CI and 5' half of the C2 domain codons.
Initially, two porcine spleen RT-PCR products, corresponding to a B-A3 domain fragment (nt 4519-5571) and a C1-C2 domain fragment (nt 6405-6990) were cloned. The 3' end of the C2 domain that was obtained extended into the exon 26 region, which is the terminal exon in fVIII. The B-A3 product was made using the porcine-specific B domain primer, SEQ ID NO:ll 5 CGC GCG GCC GCG CAT CTG GCA AAG CTG AGT T 3', where the underlined region conesponds to a region in porcine fVIII that aligns with nt 45194530 in human fVIII. The 5' region of the oligonucleotide includes a NotI site that was originally intended for cloning purposes. The antisense primer used in generating the B-A3 product, SEQ ID NO: 12 5'-GAA ATA AGC CCA GGC TTT GCA GTC RAA-3 was based on the reverse complement of the human fVIII cDNA sequence at nt 5545-5571. The PCR reaction contained 50 mM KC1,10 mM Tris-Cl, pH 9.0,0.1 % Triton X-100,1.5 mM MgCl2,2.5 mM dNTPs, 20 µM primers, 25 units/ml Taq DNA polymerase and 1/20 volume of RT reaction mix. PCR conditions were denaturation at 94 °C for 3 min, followed by 30 cycles of denaturation for 1 min at 94° C, annealing for 2 min at 50°C and elongation for 2 min at 72°C. The PCR products were phosphorylated using T4 DNA kinase and NotI linkers were added. After
cutting with NotI, the PCR fragments were cloned into the NotI site of BlueScript II KS- and transformed into XLl-Blue cells.
The C1-C2 product was made using the known human cDNA sequence to synthesize sense and antisense primers, SEQ ID NO:13 5'-AGG AAA TTC CAC TGG AAC CTT N-3' (nt 6405-6426) and SEQ ED NO: 14 5'-CTG GGG GTG AAT TCG AAG GTA GCG N-3' (reverse complement of nt 6966-6990), respectively. PCR conditions were identical to those used to generate the B-A2 product. The resulting fragment was ligated to the pNOT cloning vector using the Prime PCR Cloner Cloning System (5 Prime-3 Prime, Inc., Boulder, Colorado) and grown in JM109 cells.
The B-A3 and C1-C2 plasmids were partially sequenced to make the porcine-specific sense and antisense oligonucleotides, SEQ ID NO:15 5'-GAG TTC ATC GGG AAG ACC TGT TG-3' (nt 4551-4573) and SEQ ID NO:16 5'-ACA GCC CAT CAA CTC CAT GCG AAG-3 * (nt 6541-6564), respectively. These oligonucleotides were used as primers to generate a 2013 bp RT-PCR product using a Clontech Advantage cDNA PCR kit. This product, which corresponds to human nt 4551-6564, includes the region corresponding to the light chain activation peptide (nt 5002-5124), A3 domain (nt 5125-6114) and most of the C1 domain (nt 6115-6573). The sequence of the C1-C2 clone had established that human and porcine cDNAs from nt 6565 to the 3' end of the CI domain were identical. The PCR product cloned into the EcoRV site of pBluescript n KS-. Four clones were completely sequenced in both directions. A consensus was reached in at least 3 of 4 sites.
Isolation of porcine fVIII cDNA clones containing the 3' half of the C2 domain codons.
The C2 domain of human fVIII (nucleotides 6574-7053) is contained within exons 24-26 [Gitschier J. et al. (1984) Nature 312:326-330]. Human exon 26 contains 1958 bp, corresponding nucleotides 6901-8858. It includes 1478 bp of 3' untranslated sequence. Attempts to clone the exon 26 cDNA corresponding to the 3' end of the C2 domain, and the 3'UTR by 3' RACE [Siebert et al. (1995) supra], inverse PCR [Ochman, H. et al. (1990) Biotechnology (N.Y). 8:759-760], restriction site PCR [Sarkar, G. et al. (1993) PCR Meth.
Appl. 2:318-322], "unpredictably primed" PCR [Dominguez, 0. et al. (1994) Nucleic. Acids Res. 22:3247-3248] and by screening a porcine liver cDNA library failed. 3' RACE was attempted using the same adaptor-ligated double stranded cDNA library that was used to successfully used to clone the 5' end of the porcine fVIII cDNA. Thus, the failure of this method was not due to the absence of cDNA corresponding to exon 26.
A targeted gene walking PCRprocedure [Parker, J.D, etal. (1991) Nucleic. Acids. Res. 19:3055-3060] was used to clone the 3' half of the C2 domain. A porcine-specific sense primer, SEQ ID NO:175'-TCAGGGCAATCAGGACTCC-3' (nt 6904-6924) was synthesized based on the initial C2 domain sequence and was used in a PCR reaction with nonspecific "walking" primers selected from oligonucleotides aVIIIlable in the laboratory. The PCR products were then targeted by primer extension analysis [Parker et al. (1991) BioTechniques 10:94-101] using a 32P-end labelled porcine-specific internal primer, SEQ ID NO:18 5'-CCGTGGTGAACGCTCTGGACC-3' (nt 6932-6952). Interestingly, of the 40 nonspecific primers tested, only two yielded positive products on primer extension analysis and these two corresponded to an exact and a degenerate human sequence at the 3' end of the C2 domain: SEQ ID N0.19 5'-GTAGAGGTCCTGTGCCTCGCAGCC-3' (nt 7030-7053) and SEQ ID NO:205'-GTAGAGSTSCTGKGCCTCRCAKCCYAG-3', (nt 7027-7053). These primers had initially been designed to yield a product by conventional RT-PCR but failed to yield sufficient product that could be visualized by ethidium bromide dye binding. However, a PCR product could be identified by the more sensitive primer extension method. This product was gel-purified and directly sequenced. This extended the sequence of porcine fVIII 3' to nt 7026.
Additional sequence was obtained by primer extension analysis of a nested PCR product generated using the adaptor-ligated double-stranded cDNA library used in the 5'-RACE protocol described previously. The first round reaction used the porcine exact primer SEQ ID NO:215'-CTTCGCATGGAGTTGATGGGCTGT-3' (nt 6541-6564) and the API primer. The second round reaction used SEQ ID NO:22 5'-AATCAGGACTCCTCCACCCCCG-3' (nt 6913-6934) and the AP2 primer. Direct PCR sequencing extended the sequence 3' to the end of the C2 domain (nt 7053). The C2 domain sequence was unique except at nt 7045 near the
3' end of the C2 domain. Analysis of repeated PCR reactions yielded either A, G or a double read of A/G at this site.
Sequencing was extended into the 3 'UTR using two additional primers, SEQ ID NO:23 5'-GGA TCC ACC CCA CGA GCT GG-3' (nt 6977-6996) and SEQ ID NO:24 5*-CGC CCT GAG GCT CGA GGT TCT AGG-3' (nt 7008-7031). Approximately 15 bp of 3' UTR sequence were obtained, although the sequence was unclear at several sites. Several antisense primers then were synthesized based on the best estimates of the 3' untranslated sequence. These primers included the reverse complement of the TGA stop codon at their 3' termini. PCR products were obtained from both porcine spleen genomic DNA and porcine spleen cDNA that were visualized by agarose gel electrophoresis and ethidium bromide slaining using a specific sense primer SEQ ID NO:25 5'-AAT CAG GAC TCC TCC ACC CCC G-3' (nt 6913-6934) and the 3' UTR antisense primer, SEQ ID NO:26 5'-CCTTGCAGGAATTCGATTCA-3. To obtain sufficient quantities of material for cloning purposes, a second round of PCR was done using a nested sense primer, SEQ ID NO:27 5'-CCGTGGTGAACGCTCTGGACC-3' (nt 6932-6952) and the same antisense primer. The 141 bp PCR product was cloned into EcoRV-cut pBluescript II KS-. Sequence of three clones derived from genomic DNA and three clones derived from cDNA was obtained in both directions. The sequence was unambiguous except at nt 7045, where genomic DNA was always A and cDNA was always G.
Multiple DNA sequence alignments of human, porcine, and mouse fVIII (Fig. 1A-1HV
Alignments of the signal peptide, Al, A2, A3, CI, and C2 regions were done using the CLUSTALW program [Thompson, J.D. etal. (1994)Nucleic. Acids. Res. 22:4673-4680]. Gap open and gap extension penalties were 10 and 0.05 respectively. The alignments of the human, mouse, and pig B domains have been described previously [Elder et al. (1993) supra]. The human A2 sequence corresponds to amino acids 373-740 in SEQ ID NO:2. The porcine A2 amino acid sequence is given in SEQ ID NO:4, and the mouse A2 domain amino acid sequence is given in SEQ ED NO:28, amino acids 392-759.
Example 6: Expression of active, recombinant B-domainless porcine factor VIII (PF
Materials
Citrated hemophilia A and normal pooled human plasmas were purchased from George King Biomedical, Inc. Fetal boVIIIe serum, geneticin, penicillin, streptomycin, DMEM/F12 medium and AIM-V medium were purchased from Life Technologies, Inc. Taq DNA polymerase was purchased from Promega. Vent DNA polymerase was purchased from New England Biolabs. Pfu DNA polymerase and the phagemid pBlueScript H KS" were purchased from Stratagene. Synthetic oligonucleotides were purchased from Life Technologies or Cruachem, Inc. Restriction enzymes were purchased from New England Biolabs or Promega. 5 '-phosphorylated primers were used when PCR products were produced for cloning purposes. Nucleotide (nt) numbering of oligonucleotides used as primers for polymerase chain reaction (PCR) amplification of porcine fVIII cDNA or genomic DNA uses the human fVIII cDNA as reference [Wood et al. (1984) Nature 112:330-337]. A iYBI expression vector, designated HB-/ReNeo, was obtained from Biogen, Inc. HB'/ReNeo contains ampicillin and geneticin resistance genes and a human fVIII cDNA that lacks the entire B domain, defined as the Ser741-Argl648 cleavage fragment produced by thrombin. To simplify mutagenesis of fVIII C2 domain cDNA, which is at the 3' end of the fVTII insert in ReNeo, a NotI site was introduced two bases 3' to the stop codon of HB'/ReNeo by splicing-by-overlap extension (SOE) mutagenesis [Horton, R.M. et al. (1993) Methods Enzymol. 217:270-279], This construct is designated HB'ReNeo/NotJ.
Total RNA was isolated by acid guamdinium thixyanate-phenol-chloroform extraction [Chomczynski, P. et al. (1987) Anal. Biochem. 162:156-159]. cDNA was synthesized from mRNA using Moloney murine leukemia virus reverse transcriptase (RT) and random hexamers according to instructions supplied by the manufacturer (First-Strand cDNA Synthesis Kit, Pharmacia Biotech). Plasmid DNA was purified using a Qiagen Plasmid Maxi Kit (Qiagen, Inc.). PCR reactions were done using a Hybaid OmniGene thermocycler using Taq, Vent, or Pfu DNA polymerases. PCR products were gel purified, precipitated with ethanol, and ligated into plasmid DNA using T4 DNA ligase (Rapid DNA ligation kit, Boehringer Mannheim), Insert-containing plasmids were used to transform E, coli Epicurean XLl-Blue cells. All novel
fVIII DNA sequences generated by PCR were confirmed by dideoxy sequencing using an Applied Biosystems 373a automated DNA sequencer and the PRISM dye teirninator kit.
Construction of a hybrid fVIII expression vector. HP20, containing the porcine CI domain
A porcine fVIII cDNA corresponding to the 3' end of the CI domain and all of the C2 domain was cloned into pBluescript by RT-PCR from spleen total RNA using primers based on known porcine fVIII cDNA sequence [Healey, J.F. et al. (1996) Blood 88:4209-4214]. This construct and HB/ReNeo were used as templates to construct a human Cl-porcine C2 fusion product in pBlueScript by SOE mutagenesis. The C1-C2 fragment in this plasmid was removed with Apal and NotI and ligated into ApallNotl-cut HB/ReNeo/NotI to produce HP20/ReNeo/iVb//.
Construction of B-domain deleted hybrid human/porcine fVIII containing the porcicne light chain (HP18)
The human fVIII light chain consists of amino acid residues Aspl649-Tyr2332. The corresponding residues in the porcine fVIII cDNA were substituted for this region of HB" to produce a hybrid human/porcine fVIII molecule designated HP18. This was done by substituting a PCR product corresponding to porcine A2 region, the A3 domain, the CI domain, and part of the C2 domain for the corresponding region in HP20. To facilitate constructions, a synonymous AvrU site was introduced into nt 2273 at the junction of the A2 and A3 domains of HP20 by SOE mutagenesis.
Construction of B-domain deleted hybrid human/porcine fVIII containing the porcine signal peptide. Al domain and A2 domain (HP22V
The human fVIII signal peptide, Al domain and A2 domains consist of amino acid residues Met(-19)-Arg740. The corresponding residues in the porcine fVIII cDNA were substituted for this region of HB" to produce a molecule designated HP22. Additionally, a synonymous Avrll site was introduced into nt 2273 at the junction of the A2 and A3 domains of HP22 by SOE mutagenesis. HP22 was constructed by fusion of a porcine signal peptide-Al-partial A2 fragment in pBlueScript [Healy et al. (1996) supra] with a B-domainless hybrid
human/porcine fVIII containing the porcine A2 domain, designated HP1 [Lubin et al. (1994) supra].
Construction of porcine B domainless fVIII-(PB)-
A Spel/NotI fragment of HP18/BS (+ Avrll) was digested with 4vr.fi/MrtZ and ligated into AvrH/NotMgested HP22/BS (+ Avrll) to produce a construct PB7BS (+ Avrll), which consists of the porcine rVIII lacking the entire B domain. PB- was cloned into ReNeo by ligating an XbalNotI fragment of PB7BS (+ Avrll) into HP22/ReNeo/iVbr/ (+ Avrll).
Expression of recombinant fVIII molecules
PB7ReNeo/NotI (+ AvrII) and HP22/ReNeo/NotI (+AvrII) were transiently transfected into COS cells and expressed as described previously [Lubin, I.M. et al. (1994) /. Biol. Chem. 269:8639-8641]. HB/ReNeo/Notl and no DNA (mock) were transfected as a control.
The fVIII activity of PB", HP22, and HB" were measured by a chromogenic assay as follows. Samples of fVIII in COS cell culture supematants were activated by 40 nM thrombin in a 0.15 M NaCl, 20 mM HEPES, 5Mm cAC12,0.01% Tween-80, pH 7.4 in the presence of 10 nM factor IXa, 425 nM factor X, and 50 µM unilamellar phosphatidylserine-[phosphatidycholine (25/75 w/w) vesicles. After 5 min, the reaction was stopped with 0.05 M EDTA and 100 nM recombinant desulfatohirudin and the resultant factor Xa was measured by chromogenic substrate assay. In the chromogenic substrate assay, 0.4 mM Spectrozyme Xa was added and the rate of para-nitroanilide release was measured by measuring the absorbance of the solution at 405 nm.
Results of independently transfected duplicate cell culture supematants (absorbance at
405 nm per minute)
HB': 13.9 PB-: 139 HP22:100 mock: These results indicate that porcine B-domainless fVIII and a B-domainless fVIII consisting of the porcine Al and A2 subunits are active and suggest that they have superior activity to human B-domainless fVIII.
PB' was partially purified and concentrated from the growth medium by heparin-Sepharose chromatography. Heparin-Sepharose (10 ml) was equilibrated with 0.075 M NaCl, 10 mM HEPES, 2.5 mM CaCl2,0.005% Tween-80,0.02% sodium azide, pH 7.40. Medium (100-200 ml) from expressing cells was applied to the heparin-Sepharose, which then was washed with 30 ml of equilibration buffer without sodium azide. PB" was eluted with 0.65 M NaCl, 20 mM HEPES, 5 mM CaCl2, 0.01% Tween-80, pH 7.40 and was stored at -80 °C. The yield of fVIII coagulant activity was typically 50-75 %.
Stable expression of porcine B-domainless fVIII (PB')
Transfected cell lines were maintained in Dulbecco's modified Eagle's medium-F12 containing 10% fetal boVIIIe serum, 50 U/ml penicillin, 50 µg/ml streptomycin. Fetal boVIIIe serum was heat inactivated at 50°C for one hour before use. HB'/ReNeo and PB'ReNeo/NotI (+ AvrII) were stably transfected into BHK cells and selected for geneticin resistance using a general protocol that has been described previously [Lubinetal. (1994) Biol. Chem. 269:8639-8641] except that expressing cells were maintained in growth medium containing 600 µg/ml geneticin. Cells from Corning T-75 flasks grown to confluence were transferred to Nunc triple flasks in medium containing 600 pg/ml geneticin and grown to confluence. The medium was removed and replaced with serum-free, AIM-V medium (Life Technologies, Inc.) without geneticin. Factor VIII expression was monitored by one-stage factor VTH coagulant activity (VIIIe supra) and 100-150 ml of medium was collected once daily for four to five days. Maximum expression levels in medium for HB' and PB' were 102 units per ml and 10-12 units per ml of factor VIII coagulant activity, respectively.
Purification of PB"
PB" was precipitated from culture supernatant using 60% saturated ammonium sulfate and then purified by W3-3 immunoaiEnity chromatography and mono Q high pressure liquid
chromatography as described previously for the purification of plasma-derived porcine factor VIII [Lollar et al. (1993) Factor VIII/factor VIIIa. Methods Enzymol. 223:128-143]. The specific coagulant activity of PB' was measured by a one-stage coagulation assay [Lollar et al. (1993) supra] and was similar to plasma-derived porcine factor VIII.
When analyzed by SDS-polyacrylamide gel electrophoresis, the PB" preparation contained three bands of apparent molecular masses 160 kDa, 82 kDa, and 76 kDa. The 82 kDa and 76 kDa bands have been previously described as heterodimer containing the A1-A2 and ap-A3-Cl-C2 domains (where ap refers to an activation peptide) [Toole et al. (1984) Nature 312:342-3471. The 160 kDa band was transferred to a polyVIIIylidene fluoride membrane and subjected to NH2-terminal sequencing, which yielded Arg-IIe-Xx-Xx-Tyr (where Xx represents undermined) which is the NH2-terminal sequence of single chain factor VIII [Toole et al. (1984) supra]. Thus, PB" is partially processed by cleavage between the A2 and A3 domains, such that it consists of two forms, a single chain Al-A2-ap-A3-CI-C2 protein and a Al-A2/ap-A3-C1-C2 heterodimer. Similar processing of recombinant HB" has been reported [Lind et al. (1995) Eur. J. Biochem. 232:19-27].
Characterization of Porcine factor VIII
We have determined the cDNA sequence of porcine fVIII corresponding to 137 bp of the 5' UTR, the signal peptide coding region (57 bp), and the Al (1119 bp), A3 (990 bp), CI (456 bp), and C2 (483 bp) domains. Along with previously published sequence of the B domain and light chain activation peptide regions [Toole et al. (1986) supra] and the A2 domain [Lubin et al. (1994) supra], the sequence reported here completes the determination of the porcine fVIQ cDNA corresponding to the translated product. A fragment that included the 5' UTR region, signal peptide, and Al domain cDNA was cloned using a 5'-RACE RT-PCR protocol. A primer based on human C2 sequence was successful in producing an RT-PCR product that led to cloning of the A3, C1, and 5' half of the C2 domain. The cDNA corresponding to the 3' half of the C2 domain and 3' UTR cDNA proved difficult to clone. The remainder of the C2 domain ultimately was cloned by a targeted gene walking PCR procedure [Parker et al, (1991) supra].
The sequence reported herein SEQ ID NO:29 was unambiguous except at nt 7045 near the 3' end of the C2 domain, which is either A or G as described hereinabove. The corresponding codon is GAC (Asp) or AAC (Asn). The human and mouse codons are GAC and CAG (Gln), respectively. Whether this represents a polymorphism or a reproducible PCR artifact is unknown. Recombinant hybrid human/porcine B-domainless fVIII cDNAs containing porcine C2 domain substitutions corresponding to both the GAC and AAC codons have been stably expressed with no detectable difference in procoagulant activity. This indicates that there is not a functional difference between these two CI domain variants.
The alignment of the predicted amino acid sequence of full-length porcine fVIII SEQ ID NO:30 with the published human [Wood et al. (1984) supra] and murine [Elder et al. (1993) supra] sequences is shown in Fig. 1A-1H along with sites for post-translational modification, proteolytic cleavage, and recognition by other macromolecules. The degree of identity of the aligned sequences is shown in Table VII. As noted previously, the B domains of these species are more divergent than the A or C domains. This is consistent with the observation that the B domain has no known function, despite its large size [Elder et al. (1993) supra; Toole et al. (1986) supra]. The results of the present invention confirm that the B domain of porcine fVIII is not necessary for activity. Based on the sequence data presented herein, porcine fVIII having all or part of the B-domain deleted can be synthesized by expressing the porcine fVIII coding DNA having deleted therefrom all or part of codons of the porcine B domain. There is also more divergence of sequences corresponding to the Al domain APC/factor DCa cleavage peptide (residues 337-372) and the light chain activation peptide (Table VII). The thrombin cleavage site at position 336 to generate the 337-372 peptide is apparently lost in the mouse since this residue is glutamine instead of argmine [Elder et al. (1993) supra]. The relatively rapid divergence of thrombin cleavage peptides (or in mouse fVIII a possibly vestigial 337-372 activation peptide) has been previously noted for the fibrinopeptides [Creighton, T. E. (1993) In Proteins: Structures and Molecular Properties W.H. Freeman, New York, pp. 105-138]. Lack of biological function of these peptides once cleaved has been cited as a possible reason for the rapid divergence. Arg562 in human fVTII has been proposed to be the more important cleavage site for activated protein C during the
inactivation of fVIII and fVIIIa [Fay, P.J. etal. (1991)/. Biol. Chem. 266:20139-20145]. This site is conserved in human, porcine and mouse fVIII.
Potential N-linked glycosylation sites (NXS/T where X is not proline) can be seen in Fig. 1A-1H. There are eight conserved N-linked glycosylation sites: one in the Al domain, one in the A2 domain, four in the B domain, one in the A3 domain, and one in the C1 domain. The 19 A and C domain cysteines are conserved, whereas there is divergence of B domain cysteines. Six of the seven disulfide linkages in fVIII are found at homologous sites in factor V and ceruloplasmin, and both C domain disulfide linkages are found in factor V [McMullen, B. A. et al. (1995) Protein Sci. 4:740-746]. Human fVIII contains sulfated tyrosines at positions 346,718,719,723,1664, and 1680 [Pittman, D.D. et al. (1992) Biochemistry 31 :3315-3325; Michnick, D.A. et al. (1994) J. Biol Chem. 269:20095-20102]. These residues are conserved in mouse fVIII and porcine fVIII (Fig. 1), although the CLUSTALW program failed to align the mouse tyrosine corresponding to Tyr346 in human fVIII.
Mouse and pig plasma can correct the clotting defect in human hemophilia A plasma, which is consistent with the level of conservation of residues in the A and C domains of these species. The procoagulant activity of porcine fVIII is superior to that of human fVIII (Lollar, P. et al. (1992) J. Biol Chem. 267:23652-23657]. The recombinant porcine factor VIII (B domain-deleted) expressed and purified as herein described also displays greater specific coagulant activity than human fVIII, being comparable to plasma-derived porcine fVIII. This may be due to a decreased spontaneous dissociation rate of me A2 subunit from the active A1/A2/A3-C1-C2 fVIIIa heterotrimer. Whether this difference inprocoagulant activity reflects an evolutionary change in function as an example of species adaptation [Perutz, M.F. (1996) Adv. Protein Chem, 36:213-244] is unknown. Now that the porcine fVIII cDNA sequence corresponding to the translated product is complete, homolog scanning mutagenesis [Cunningham, B.C., et al. (1989) Science 243:1330-1336] may proVIIIe a way to identify structural differences between human and porcine fVTII that are responsible for the superior activity of the latter.
Porcine fVni is typically less reactive with inhibitory antibodies that arise in hemophiliacs who have been transfused with fVIII or which arise as autoantibodies in the general population. This is the basis for using porcine fVIII concentrate in the management of patients with inhibitory antibodies [Hay and Lozier (1995) supra]. Most inhibitors are directed against epitopes located in the A2 domain or CI domain [Fulcher, C.A. et al. (1985) Proc. Natl. Acad. Sci. USA 82:7728-7732; Scandelk, D. et al. (1988) Proc. Natl. Acad. Sci. USA 85:6152-6156; Scandella, D. et al. (1989) Blood 74:1618-1626]. Additionally, an epitope of unknown significance has been identified that is in either the A3 or CI domain [Scandella et al. (1989) supra] Scandella, D. et al. (1993) Blood 82:1767-1775; Nakai, H. et al. (1994) Blood 84:224a]. The A2 epitope has been mapped to residues 484-508 by homolog scanning mutagenesis [Healey et al. (1995) supra]. In this 25 residue segment, there is relatively low proportion of identical sequence (16/25 or 64%). It is interesting that this region, which appears to be functionally important based on the fact that antibodies to it are inhibitory, apparently has been subjected to relatively more rapid genetic drift. Alignment of the porcine A2 domain and A3 domains indicate that the A2 epitope shares no detectable homology with the corresponding region in the A3 domain.
The C2 inhibitor epitope of human fVIII has been proposed to be located to within residues 2248-2312 by deletion mapping [Scandella, D. et al. (1995) Blood 86:1811-1819]. Human and porcine fVIII are 83% identical in this 65 residue segment. However, homolog scanning mutagenesis of this region to characterize the C2 epitope has revealed that a major determinant of the C2 epitope was unexpectedly located in the region corresponding to human amino acids 2181-2243 (SEQ ID NO:2) and Fig. 1H.
Human-porcine hybrid factor VIII proteins were made in which various portions of the C2 domain of human factor VIII were replaced by the conespondipg portions of porcine factor VIII, using the strategy herein described. (Example 5) The synthesis of the various C2-hybrid factor VIIIs was accomplished by constructing hybrid coding DNA, using the nucleotide sequence encoding the porcine C2 region given in SEQ ID NO:30. Each hybrid DNA was expressed in transfected cells, such that the hybrid factor VIIIs could be partially purified from
amino acids, especially those which are non-identical betweenhuman/porcine or human/mouse or which are most likely to contribute to antibody binding, can yield a modified factor VIII with reduced reactivity to inhibitory antibodies.
Figs. 1A-1H taken together proVIIIe an aligned sequence comparison of the human, pig and mouse factor VIII amino acid sequences. Fig. 1A compares signal peptide regions (human, SEQ ID NO:31; porcine, SEQ ID NO.30, amino acids 1-19; murine, SEQ ID NO:28, aminc> acids 1-19). Note that the amino acids in Fig. 1A-1H are numbered at the first Alanine of the mature protein as number 1, with amino acids of the signal peptide assigned negative numbers. The Human fVIII sequence in SEQ ID NO:2 also begins with the first Alanine of the mature protein as amino acid number 1. In the amino acid sequences of mouse fVIII (SEQ ID NO:28) and porcine fVIII (SEQ ID NO:30), the first amino acid (alanine) of the mature sequence is amino acid number 20. Fig. 1A-1H shows an alignment of the corresponding sequences of human, mouse and pig fVIII, such that the regions of greatest amino acid identity are juxtaposed. The amino acid numbers in Fig. 1A-1H apply to human fVTII only. Fig.1B gives the amino acid sequences for the Al domain of human (SEQ ID NO:2, amino acids 1-372), porcine (SEQ ID NO:30, amino acids 20-391), and murine (SEQ ID NO:28, amino acids 20-391). Fig. 1C proVIIIes amino acid sequences for the Factor VIII A2 domains from human (SEQ ID NO:2, amino acids 373-740), pig (SEQ ID NO:30, amino acids 392-759) and mouse (SEQ ID NO:28, amino acids 392-759). Fig. ID proVIIIes the amino acid sequences of B domains of human factor VIII (SEQ ID NO:2, amino acids 741-1648), pig (SEQ ID NO:30, amino acids 760-1449) and mouse (SEQ ID NO:28, amino acids 760-1640). Fig. IE compares the amino acid sequences of Factor VIII light chain activation peptides of human, pig and mouse (SEQ ID NO:2, amino acids 1649-1689; SEQ ID NO:30, amino acids 1450-1490; and SEQ ID NO:28, amino acids 1641-1678, respectively). Fig. 1F proVIIIes the sequence comparison for human, pig and mouse Factor VIII A3 domains (SEQ ID NO:2, amino acids 1690-2019; SEQ ID NO:30, amino acids 1491-1820; and SEQ ID NO:28, amino acids 1679-2006, respectively. Fig. 1G proVIIIes the amino acid sequences of the Factor VIII C1 domains of human, pig and mouse (SEQ ID NO:2, amino acids 2020-2172; SEQ ID NO:30, amino acids 1821-1973; and SEQ ID NO:28, amino acids 2007-2159, respectively). Fig. 1H
the growth medium. Activity, in the absence of any inhibitor, was measured by the one-stage clotting assay.
A battery of five human inhibitors was used to test each hybrid factor VIII. The inhibitor plasmas containing anti factor VIII antibody had been previously shown to be directed against human C2 domain, based on the ability of recombinant human C2 domain to neutralize the inhibition. In all the test plasmas, the inhibitor titer was neutralized greater than 79% by C2 domain or light chain but less than 10% by recombinant human A2 domain. In addition the C2-hybrid factor VIIIs were tested against a murine monoclonal antibody, which binds the C2 domain, and like human C2 inhibitor antibodies, it inhibited the binding of factor VIII to phospholipid and to von Willebrand factor.
By comparing the antibody inhibitor titers against the C2-hybrid factor VIIIS, the major determinant of the human C2 inhibitor epitope was shown to be the region of residues 2181-2243 (SEQ ID NO:2, see also Fig. 1H). Anti-C2 antibodies directed to a region COOH-terminal to residue 2253 were not identified in four of the five patient sera. In comparing hybrids having porcine sequence corresponding to human amino acid residues numbers 2181-2199 and 2207-2243, it was apparent that both regions contribute to antibody binding. The porcine amino acid sequence corresponding to human residues 2181-2243 is numbered 1982-2044 in SEQ ID NO:30. The sequence of porcine DNA encoding porcine amino acids numbered 1982-2044 is nucleotides numbered 5944-6132 in SEQ ID NO.-29.
Referring to Fig. 1H, it can be seen that in the region 2181-2243, there are 16 amino acid differences between the human and porcine sequences. The differences are found at residues 2181,2182,2188,2195-2197,2199,2207,2216,2222,2224-2227,2234,2238 and 2243. Amino acid replacement at one or more of these numbered residues can be carried out to make a modified human factor VIII non-reactive to human anti-C2 inhibitor antibodies. Alanine scanning mutagenesis proVIIIes a convenient method for generating alanine substitutions for naturally-occurring residues, as previously described. Amino acids other than alanine can be substituted as well, as described herein. Alanine substitutions for indiVIIIual
proVIIIes sequence data for the C2 domains of the Factor VIII C2 domains of human, pig and mouse (SEQ ID NO:2, amino acids 2173-2332; SEQ ID NO:30, amino acids 1974-2133; and SEQ ID NO:28, amino acids 2160-2319, respectively).
The diamonds represent tyrosine sulfation sites, proposed binding sites for Factor Ka, phospholipid and Protein C are double-underlined, and regions involved in binding anti-A2 and anti-C2 inhibitory antibodies are italicized. Asterisks highlight amino acid sequences which are conserved. See also SEQ ID NO.29 (porcine factor VIII cDNA) and SEQ ID NO:30 (deduced amino acid sequence of porcine factor VIII). The human numbering system is used as the reference [Wood et al. (1984) supra]. The Al, A2, and B domains are defined by thrombin cleavage sites at positions 372 and 740 and an unknown protease cleavage site at 1648 as residues 1-372, 373-740, and 741-1648, respectively [Eaton, D.L. et al. (1986) Biochemistry 25:8343 8347]. The A3, C1, and C2 domains are defined as residues 1690-2019, 2020-2172, and 2173-2332, respectively [Vehar et al. (1984) supra]. Cleavage sites for thrombin (factor IIa), factor IXa, factor Xa and APC [Fay et al. (1991) supra; Eaton, D. et al. (1986) Biochemistry 25:505-512; Lamphear,B. J. etal. (1992)Blood 80:3120-3128] are shown by placing the enzyme name over the reactive arginine. An acidic peptide is cleaved from the fVIII light chain by thrombin or factor Xa at position 1689. Proposed binding sites for factor IXa [Fay, P.J. et al. (1994) /. Biol. Chem. 269:20522-20527; Lenting, P.J. et al. (1994) /. Biol Chem. 262:7150-7155), phosphohpid (Foster, P.A. et al. (1990) Blood 15:1999-2004) and protein C (Walker, F.J. et al. (1990) I Biol Chem. 265:1484-1489] are doubly underlined. Regions involved in binding anti-A2 [Lubin et al. (1994) supra; Healey et al. (1995) supra]; and previously proposed for anti-C2 inhibitory antibodies are italicized. The C2 inhibitor epitope identified as herein described (human amino acids 2181-2243) is shown by a single underline in Fig. 1H. Tyrosine sulfation sites ppittman et al. (1992) supra; Michnick et al. (1994) supra] are shown by ♦.
Example 7 Constrution of POL1212 and Expression in Baby Hamster Kidney Cells
POL1212 is a partially B-domainless porcine factor VIII, having the B-domam deleted except that 12 amino acids of the NH2 terminus of the B-domain and 12 amino acids of the COOH terminus are retained.
The cDNAs encoding for the sequences for the porcine fVIII domains Al,A2,ap-A3CI, and C2 were obtained as described in Example 5. The DNA nucleotide sequence and derived amino acid sequence of porcine factor VIII are presented as SEQ ID NO. 29 and SEQ ID NO: 30, respectively. The amplified fragments were separately cloned into the plasmid pBluescript II KS (pBS).
POL1212 refers to the cDNA encoding porcine fVIII lacking most of the B domain but containing DNA sequence encoding a 24 amino acid linker between the A2 and ap domains POL1212 was constructed in a mammalian expression vector, ReNeo, which was obtained from a commercial sources (Biogen, Cambridge M.A, U.S.A)'.ReNeo can relicate in bacteria, replicate as an episome in COS cells for transient expression of factor VIII, or be stably intergrated into a variety of mammalian cells. It consists of 1) sequences derived from plasmid pBR322 that include and origin of replication and ampicillin resistance gene, 2) a neomycin resistance gene whose expression is under control of the SV40 promoter/enhancer, SV40 small t intron, and the SV40 polyadenylation signal regulatory elements, 3) a site for insertion of fVIIIand its signal peptide, the expression of which is under control of the SV40 enhancer, adenovirus type 2 major late promoter, and adenovirus type 2 tripartite leader sequence Any vector having similar functional components can be used in place of the ReNeo vector.
P0L1212/ReNeo was prepared in several steps. First, the cDNAs encoding for porcine fVIII heavy chain (A1-A2) and the cDNAs encoding for porcine fVIII light chain (ap-A3-Cl-C2) were separately assembled in pBS. From these constructs, the DNA encoding for procine B-domainless fVIII was assembled in pBS (PB-/pBS). This form of porcine fVIII lacks the entire B domain, defined as amino acids corresponding to residues 741- 1648 in human fVIII (human
the human A2 domain in the human B-domainless fVIII expression vector ReNeo (HB-/ReNeo). The DNA encoding the remainder of the porcine heavy chain and the DNA encoding the porcine light chain was substituted for the human domains in two additional steps using the porcine heavy chain/pBS and PB-/pBS constructs made previously. A fragment of the human B domain encoding the 5 C-terminal and 9 N-terminal amino acids was inserted between the A2 and A3 domains producing a construct called PSQ/ReNeo [Healey et al. (1998) 92:3701-3709]. Residues Glu2181-Val2243 contain a major determinant of the inhibitory epitope in the C2 domain of human factor VIII). This construct was used as a template to make a fragment of the porcine B domain encoding for the 12 C-terminal and 12 N-tenninal amino acids. This fragment was inserted between the A2 and A3 domains resulting in the final construct, POL1212/ReNeo.
The POL1212 24 amino acid linker consists of the first 12 and last 12 residues of the porcine tVIII B domain. The POL1212 linker has the following sequence:
SFAQNSRPPSASAPKPPVLRRHQR. (SEQ ID NO:32)
The nucleotide sequence corresponding to the 1212 linker and surrounding amino acids is:
(Sequence Removed)
The POL1212 linker was synthesized by splicing-by-overlap extension (SOE) mutagenesis, as follows:
PCR reactions used to make SOE products were as follows:
REACTION #1
Outside primer: Rev 4, which is a porcine A2 primer, nucleotides 1742-1761. (SEQ ID NO:29) The sequence is: 5'-GAGGAAAACCAGATGATGTCA-3' (SEQ ID NO:34)
Inside primer: OL12, which is a porcine reverse primer covering the first (5') 15 amino acids of OL1212 and the last (3') 5 amino acids of porcine A2. The sequence is:
(Sequence Removed)
Template: PSQ/ReNeo
Product: porcine DNA from nucleotide 1742 in the A2 domain to 2322 in OL1212,
580 bp
REACTION #2
Outside primer: P2949 is a porcine reverse A3 primer, nucleotides 2998-3021 of SEQ ID NO:29. The sequence is: 5'-GGTCACTTGTCTACCGTGAGCAGC -3' (see SEQ ID NO:29)
" Inside primer: OL12+, a porcine primer covering the last (3') 16 amino acids of OL1212 and the first (5') 6 amino acids of the activation peptide, nucleotide 2302-2367 of SEQ ID NO:29. The sequence is:
(Sequence Removed)
Template: PSQ/ReNeo.
Product: porcine from nucleotide 2302 in OL1212 to nucleotide 3021 in the A3 domain, 719 bp
SOE REACTION
Primers Rev 4,P2849-
Templates: Fragment from rxn#l (bp) and low melt fragment from rxn # 2 (bp)
Product: porcine DNA from nucleotide 1742 in the A2 domain to nucleotide 3021 in
the A3 domain (SEQ ID NO 29) including OH 212, 1279 bp. The reaction product was
ethanol precipitated.
The 1212 linker was inserted into PSQ/ReNeo by cutting the SEO product (insert) and PSQ/ReNeo (vector) with BsaB I. The vector and insert were hgated using T4 ligase and the product was used to transform E.coli XL 1-Blue cells. Plasmid DNA was prepared from several colomes and the sequence of the 1212 linker and other PCR-generated sequence was verified by DNA sequence analysis.
CULTURE OF BABY HAMSTER KIDNEY (BHK) CRL-1632 CELLS
A BHK cell line was obtained from the ATCC, accession identification CRL-1632 and was stored frozen at - 20°C until further use. The cells were thawed at 37°C and put into 10ml of complete medium, defined as DMEM/F12,50U/ml penicillin, 50 ug/ml streptomycin plus 10% fetal boVIIIe serum (FBS) FBS was purchased from Hyclone, Logan Utah The cells were centnfuged for 2 minutes at 300 RPM. The medium was aspirated and the cells were resuspended in two ml complete medium in a T-75 flask containing 20ml of complete medium.
POL1212 has been expressed in both baby hamster kidney (BHK) and Chinese hamster ovary (CHO) cells. Two BHK lines were used, the CRL-1632 line from ATCC and another BHK line obtained form R Mcgillivray, University of British Columbia, [Funk, et al. (1990) Biochemistry 29:1654-1660]. The latter were subcultured without selection in the inventors' lab and designated BHK1632(Emory) The CHO cell line was CHO-K1, ATCC accession CCL-61. The expression of the average clone from the Emory cell line and from CHO-K1 cells was somewhat higher than from CRL-1632 cells as judged by chromogenic assay activity.
A baby hamster kidney (BHK) cell line CRL 1632 and a Chinese hamster ovary (CHO) cell line CHO-K1, were obtained from American Type Culture Collection (ATCC) Cell line BHK 1632 (Emory) was deposited at ATCC under the designated BHK-M
The cells grown in the T-75 flask formed a confluent monolayer. A 60 ml culture of E coli XLl-Blue cells in LB/ampicillin (50 mg/ml) carrying the POL1212/ReNeo plasmid was prepared.
TRANSFECTION OF CRL-1632 BHK CELLS WITH POL1212/ReNeo
DNA from the overnight culture of the POL1212/ReNeo XLl-Blue cells was prepared using a Qiagen, Valencia, CA Spin Miniprep kit. One flask of CRL-1632 cells was split into a stock flask with 0.2 ml and a flask for transfection with 0.3 ml from 2 ml total. The other flask was fed fresh medium. Medium was DMEM/F12 + 10% Hyclone FBS + 50 U/ml penicillin, 50 µg/ml streptomycin. CRL-1632 cells were split into 6 well plates airning for 50-90% confluence for transfection (0.3 ml of cells from the T-75 flask in 2 ml 1:5000 Versene [Life Technologies, Gaithersburg, MD] in each well) using fresh DMEM/F12 4-10% Hyclone FBS + 50 U/ml penicillin, 50 ug/ml streptomycin.
The following solutions were prepared in sterile 1-2 ml test tubes;
A) 48 µl (10µg) Miniprep POL1212/ReNeo DNA plus ul medium without serum (DMEM/F12) plus 10 µl Lipofectin™ (Life Technologies, Gaithersburg, MD).
B) 10 ul Lipofectin plus 190 ul medium (mock transfection) was gently mixed and the DNA and Lipofectin allowed to react for 15 minutes at room temperature. During this time, the cells were washed twice with2 ml of DMEM/F12. 1.8 ml of DMEM/F12 was then added to the cells. The DNA/Lipofectin complex was added dropwise to the cells, and swirled gently to mix. The cells remained in the incubator overnight. Removed the DNA/lipofectin and added 3 ml of medium with serum to the cells. Incubated the cells 30 - 48 hours. Geneticin was purchased from Life Technologies, Gaithersburg, MD. The cell cultures were diVIIIed 1:20, 1:50 and 1:100, 1:250, 1:500 onto 10 cm dishes in 10 ml of medium with serum containing 535 jig/ml geneticin. Over the next several days, cells that did not take up the POL1212/ReNeo plasmid were killed due to the presence of geneticin. The remaining cells continued to replicate in geneticin, forrning visible monolayer colonies on the dishes.
EXPRESSION AND ASSAY OF POL1212 from BHK CRL-1632 CELLS
Small plastic cylindrical rings were placed around the colonies. The colonies were aspirated separately using complete medium and transferred to test tubes. These colonies are referred to as ring cloned colonies. Ring cloned colonies were plated separately onto 24 well plates and grown in complete medium.
CHROMOGENIC SUBSTRATE ASSAY FOR FACTOR VIII EXPRESSION BY TRANSFECTED CRL-1632 CELLS
Samples of POL1212 from cell culture supernatants were mixed with 50 nM purified porcine factor Ka and 0,05 mM phosphtidylcholine/phosphatidylserine (PCPS) vesicles in 0.15M NaCl, 20 m HEPES, 5mM CaC12,0.01 % Tween 80, pH 7.4. As a control, cell culture medium from mock-transfected cells was used. Thrombin and factor X were added simultaneously to final concentrations of 40 and 425 nM, respectively, thrombin activates factor VIII, which then, along with PCPS, serves as a cofactor for factor Ka during the activation of factor X.
After 5 min, the activation of factor X by factor Ka/factor "VIIIa/PCPS was stopped by the addition of EDTA to a final concentration of 50 mM. At the same time the activation of factor VIII by thrombin was stopped by the addition of the thrombin inhibitor, recombinant desulfatohirudin, to a final concentration of 100 nM. A 25-µl sample of the reaction mix was transferred to a microliter well, to which was added 74 µl of Spectrozyme Xa (America Diagnostica, Greenwich, CT), which is a chromogenic substrate for factor Xa. The final concentration of Spectrozyme Xa was 0.6 mM. The absorbance at 405 nm due to the cleavage of Spectrozyme Xa by factor Xa was monitored continuously for 5 minutes with a VIIIax Kinetic Plate Reader (Molecular Devices, Inc., Menlo park, CA). The results are expressed in terms of A405/min.
Factor VIII chromogenic assay of ten ring-cloned colonies:

(Table Removed)
These results show that all ten colonies that were selected express factor VIII activity that is at least ten-fold greater than background.
The activity from medium of colony 8, which was the highest expressing colony, was further examined by one-state factor VIII clotting assay. In this assay, 50 ml of factor VIII deficient plasma (George King Biomedical Overland Park, KA), 5 ml sample or standard, and 50 ml of activated particulate thromboplastin time reagent (Organon Teknika, Durham, NC) were incubated 3 min at 37° C. Samples include colony 8 medium diluted in 0.15 M NaCl, mM hepes, pH 7.4 (HBS) or, as a control, complete medium. Clotting was initiated by addition of 50 ml of 20 mM CaC12. The clotting time was measured using an ST4 BIO Coagulation Instrument (Diagnostica Stago, Parsippany, NJ). A standard curve was obtained by making dilutions of pooled, titrated normal human plasma, lot 0641 (George King Biomedical, Overland Park, KA). The factor VIII concentration of the standard was 0.9 units per ml.
Standard curve:
Dilution U/ml Clot Time
1) Undiluted 0.96 45.2
2) 1/3 (HBS) 0.32 53.7
3) 1/11 (HBS) 0.087 62.5
4) 1/21 (HBS) 0.046 68.9
Linear regression of the clotting times versus the logarithm of the concentration of standard yielded a correlation coefficient of 0.997.
Test substances gave the following clotting times, which were converted to units per ml using the standard curve:
Sample Clot Time (sec') Units/ml
1) Colony 8 (24h), 1/10 in HBS 40.6 1.74x10 = 17.4
2) Colony 8 (24h), 1/10 in HBS 41.1 1.63x10 = 16.3
3) Colony 8 (24h), 1/20 in HBS 47.7 0.69x20 = 13.8
4) Colony 8 (24h), 1/20 in HBS 47.2 0.73x20 = 14.6
5) Complete medium 82.9 0.007
6) Complete medium 83.3 0.006
These results show that colony 8 clotting activity that is approximately 2000-fold higher than the control sample.
The DNA sequence encoding POL1212 is set forth as SEQ ID NO:37. The encoded amino acid sequence of POL1212 is set forth as SEQ ID NO:38. Further purification of POL1212 can be carried out using a variety of known methods such as immunoafrlnity chromatography and HPLC chromatography - see Examples 2 and 3.
GENERAL CONCLUDING REMARKS It will be understood that minor variations of amino acid sequence or the DNA encoding such sequence relating to POL1212 can be introduced without affecting the essential
attributes of function. For example, the length of B-domain sequence retained as a linker /between the A2 domain and the activation peptide can be increased or decreased within limits known in the art. Sequence variants can be introduced in the linker region while retaining the equivalent functional attributes of P0L1212 as taught herein and of porcine B-domainless factor VIII as taught herein and as known in the art. Based on comparisons of known factor VIII amino acid sequences having coagulant activity in human blood, sequence variants such as indiVIIIual amino acid substitutions or substitution of peptide segments with known functional variants can be made in the basic P0L1212 amino acid sequence, while retaining the equivalent functional attributes thereof. The foregoing types of variation are not intended as exhaustive, but are merely exemplary of the sequence modifications that could be made by those of ordinary skill in the art, without substantially modifying the functional attributes of the protein. All such variants and modifications are deemed to fall within the scope of the invention as claimed or as equivalents thereof.
Sequence ID listing:
SEP ID NO: Identification
1 Human factor VIII cDNA. Coding for amino acid number 1 of the mature protein begins at nucleotide number 208.
2 Human factor amino acid sequence.
3 Porcine factor VIII A2 domain cDNA
4 Porcine factor VIII A2 domain amino acid sequence
5 thru 27 Oligonucleotide primer seq. (Example 5)
28 Murine factor VIII amino acid sequence
29 Porcine factor VIII cDNA
30 Porcine factor VIII amino acid sequence
31 Human factor VIII signal peptide amino acid sequence
32 thru 36 Oligonucleotiode primer (Example 7)
37 POL1212 coding DNA
38 POL1212 amino acid sequence






We Claim:
1. A modified partially B-domainless porcine factor VIII protein, wherein said protein is expressed by DNA encoding the amino acid sequence of POL 1212 as set forth in SEQ. ID No. 38.
2. A modified partially B-domainless porcine factor VIII protein as claimed in claim-1, wherein said DNA is comprised in an expression vector.
3. A modified partially B-domainless porcine factor VIII protein as claimed in claim-1, wherein said DNA is having the nucleotide sequence of SEQ. ID No.
37.
4. A modified partially B-domainiess porcine factor VIII protein as claimed in claim-3, wherein said DNA is comprised in an expression vector.
5. A modified partially B-domainless porcine factor VIII protein as and when used for preparation of a therapeutic composition such as herein described.
6. A modified partially B-domainless porcine factor VIII protein as claimed in claim-1 obtained from a mammalian cell containing and replicating an expression vector comprising nucleic acid molecule encoding the amino acid sequence of POL 1212 as set forth in SEQ. ID No. 38.
7. A modified partially B-domainless porcine factor VIII protein as claimed in claim-6, wherein said mammalian cell is BHK CRL-1632,
8. A modified partially B-domainless porcine factor VIII protein, substantially as hereinbefore describe with reference to the accompanying drawings.

Documents:

1383-del-2004-abstract.pdf

1383-del-2004-assignment.pdf

1383-del-2004-claims cancelled.pdf

1383-del-2004-claims.pdf

1383-del-2004-complete specification (granted).pdf

1383-del-2004-correspondence-others.pdf

1383-del-2004-correspondence-po.pdf

1383-del-2004-description (complete).pdf

1383-del-2004-drawings.pdf

1383-del-2004-form-1.pdf

1383-del-2004-form-18.pdf

1383-del-2004-form-2.pdf

1383-del-2004-form-26.pdf

1383-del-2004-form-3.pdf

1383-del-2004-form-5.pdf

1383-del-2004-gpa.pdf


Patent Number 242282
Indian Patent Application Number 1383/DEL/2004
PG Journal Number 35/2010
Publication Date 27-Aug-2010
Grant Date 20-Aug-2010
Date of Filing 26-Jul-2004
Name of Patentee EMORY UNIVERSITY
Applicant Address 2009, RIDGEWOOD DRIVE, ATLANTA, GA 30322, U.S.A
Inventors:
# Inventor's Name Inventor's Address
1 LOLLAR, JOHN, S 2568 OAK CROSSING DRIVE, DECATUR, GA, 30033, U.S.A.
PCT International Classification Number C12Q 1/68
PCT International Application Number N/A
PCT International Filing date
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 09/523,656 2000-03-10 U.S.A.