COMPOSITIONS AND METHODS FOR THE TREATMENT OF HEMOPHILIA A

Improved materials and methods for the treatment of Hemophilia A are provided.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims priority under 35 U.S.C. §119(e) to U.S. Provisional Patent Application No. 60/382,486 filed on May 22, 2002, the entire disclosure of which is incorporated by reference herein.

GOVERNMENT RIGHTS

Pursuant to 35 U.S.C. §202(c), it is acknowledged that the U.S. Government has certain rights in the invention described herein, which was made in part with funds from the National Institutes of Health, Grant Numbers: HL48322 and T32HL07439.

FIELD OF THE INVENTION

This invention relates to the fields of medicine and gene therapy. More specifically, the invention provides materials and methods for the restoring factor VIII activity in patients in need thereof.

BACKGROUND OF THE INVENTION

Several publications and patent documents are referenced in this application in order to more fully describe the state of the art to which this invention pertains. Full citations for these references are found at the end of the specification. The disclosure of each of these publications is incorporated by reference herein.

Hemophilia is a genetic disease characterized by a blood clotting deficiency. In hemophilia A (classic hemophilia, Factor VIII deficiency), an X-chromosome-linked genetic defect disrupts the gene encoding Factor VIII, a plasma glycoprotein, which is a key component in the blood clotting cascade. The cDNA sequence encoding human Factor VIII is available at GenBank with Accession No. K01740 (SEQ ID NO: 1). Within SEQ ID NO: 1, nucleic acids 208-7206 encode the full-length wild-type FVIII polypeptide (2332 amino acids, SEQ ID NO: 2) and nucleic acids 151-207 encode a preceding 19-residue signal sequence peptide (SEQ ID NO: 3). Human Factor VIII may be synthesized as a single chain polypeptide, with a predicted molecular weight of 265 kDa. The Factor VIII protein (SEQ ID NO: 2) has six domains, designated from the amino to the carboxy terminus as A1-A2-B-A3-C1-C2 (Wood et al., Nature 312:330 [1984]; Vehar et al., Nature 312:337 [1984]; and Toole et al., Nature 312:342 [1984]). Human Factor VIII is processed within the cell to yield a heterodimer primarily comprised of a heavy chain of 200 kDa containing the A1, A2, and B domains and an 80 kDa light chain containing the A3, C1, and C2 domains (Kaufman et al., J. Biol. Chem., 263:6352-6362 [1988]). Both the single chain polypeptide and the heterodimer circulate in the plasma as inactive precursors (Ganz et al., Eur. J. Biochem., 170:521-528 [1988]). Activation of Factor VIII in plasma is initiated by thrombin cleavage between the A2 and B domains, which releases the B domain and results in a heavy chain consisting of the A1 and A2 domains. The 980 amino acid B domain is deleted in the activated procoagulant form of the protein. Additionally, in the native protein, two polypeptide chains (“a” and “b”), flanking the B domain, are bound to a divalent calcium cation. Hemophilia may result from point mutations, deletions, or mutations resulting in a stop codon (See, Antonarakis et al., Mol. Biol. Med., 4:81 [1987]).

The disease is relatively rare, afflicting approximately one in 10,000 males. Hemophilia in females is extremely rare, although it may occur in female children of an affected father and carrier mother, as well as in females with X-chromosomal abnormalities (e.g., Turner syndrome, X mosaicism, etc.). The severity of each patient's disease is broadly characterized into three groups--“mild,” “moderate,” and “severe,” depending on the severity of the patient's symptoms and circulating Factor VIII levels. While normal levels of Factor VIII range between 50 and 200 ng/mL plasma, mildly affected patients have 6-60% of this value, and moderately affected patients have 1-5% of this value. Severely affected hemophiliacs have less than 1% of normal Factor VIII levels.

While hemophiliacs clearly require clotting factor after surgery or severe trauma, on a daily basis, spontaneous internal bleeding is a greater concern. Hemophiliacs experience spontaneous hemorrhages from early infancy, as well as frequent spontaneous hemarthroses and other hemorrhages requiring clotting factor replacement.

Without effective treatment, chronic hemophilic arthropathy occurs by young adulthood. Severely affected patients are prone to serious hemorrhages that may dissect through tissue planes, ultimately resulting in death due to compromised vital organs.

Clearly a need exists for improved compositions and methods for the treatment of this genetic disorder.

SUMMARY OF THE INVENTION

In accordance with the present invention, variant Factor VIII (FVIII) molecules having higher specific activity than native molecules are provided.

In one aspect, nucleic acid molecules encoding variant FVIII polypeptides are provided. These nucleic acids can be used to advantage in methods of gene therapy for the treatment of Hemophilia A.

In another aspect of the invention, the variant polypeptides described in Table II are expressed in a recombinant system, isolated and purified. The variant FVIII molecules are then formulated into a pharmaceutical composition for administration to patients in need thereof.

In yet further aspect of the invention, methods are provided for the treatment of Hemophilia A using the variant FVIII molecules disclosed herein.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 is a schematic diagrams showing the proteolytic processing of human Factor VIII.

FIG. 2 is a schematic diagram of recombinant Factor VIII-SQ.

FIG. 3 depicts full length FVIII, rFVIII-SQ, rFVIII-RS, and recombinant FVIII constructs of the present invention, rFVIII-R4 and rFVIII-RKR2. Numbers in parentheses indicate the base pairs of the entire FVIII gene construct and total amino acids, including the signal sequence.

FIG. 4 depicts full length FVIII, rFVIII-SQ, and the recombinant FVIII constructs of the present invention, FVIII(Δ721-1689) and FVIII(Δ731-1689). Numbers in parentheses indicate the base pairs of the entire FVIII gene construct and total amino acids, including the signal sequence.

FIG. 5 is a graph showing FVIII levels expressed by the various constructs shown in FIG. 3 in transiently transfected COS-1 cells.

FIG. 6 shows the results obtained following stable transfection of BHK cells with constructs encoding recombinant Factor VIII. The data show that the constructs of the present invention produce FVIII with greater activity than prior art recombinant constructs.

FIG. 7 is a graph showing Factor VIII levels expressed by the various constructs shown in FIG. 4 in transiently transfected COS-1 cells.

DETAILED DESCRIPTION OF THE INVENTION

The invention described herein constitutes a novel form of the FVIII gene product for the treatment of Hemophilia A in a gene therapy or protein-based setting. An exemplary minigene of the invention consists of DNA sequences which encode for amino acids 1-740 of the heavy chain and 1690-2332 of the light chain (lacking the acidic region comprising amino acids 1648-1689 in the N-terminus of the light chain) which is interconnected by a linker DNA segment coding for a linker peptide which is recognized by the furin or furin-like enzymes (i.e. basic amino acids, RRRR (SEQ ID NO: 4) or RKRRKR (SEQ ID NO: 5) or any combination of basic residues which allows for efficient intracellular processing). Additionally, further deletion of the C-terminus of the heavy chain and the N-terminus of the light chain (with protease cleavage site in the middle, and combinations thereof) to further shorten the gene are also disclosed. This shortened form of the FVIII gene is more suitable for delivery via gene therapy than the full-length, native FVIII-encoding sequence and also creates a FVIII gene product with enhanced activity in the secreted form. While not wishing to be bound to any particular molecular theory, it appears that the protein is either activated more efficiently by thrombin or other activating protease to yield the active cofactor (FVIIIa) or, it may be secreted from the cell as a “partially” active pro-cofactor.

I. Definitions

The following definitions are provided to aid in understanding the subject matter regarded as the invention.

“Gene transfer” and “gene delivery” refer to methods or systems for reliably inserting a particular nucleic acid sequence into targeted cells.

As used herein, “Factor VIII (FVIII)” refers to a protein which functions as an essential co-factor in the activation of Factor X in the intrinsic blood coagulation system. FVIII minigene refers to a nucleic acid encoding modified FVIII proteins of the invention.

“Nucleic acid” or a “nucleic acid molecule” as used herein refers to any DNA or RNA molecule, either single or double stranded and, if single stranded, the molecule of its complementary sequence in either linear or circular form. In discussing nucleic acid molecules, a sequence or structure of a particular nucleic acid molecule may be described herein according to the normal convention of providing the sequence in the 5′ to 3′ direction. With reference to nucleic acids of the invention, the term “isolated nucleic acid” is sometimes used. This term, when applied to DNA, refers to a DNA molecule that is separated from sequences with which it is immediately contiguous in the naturally occurring genome of the organism in which it originated. For example, an “isolated nucleic acid” may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a procaryotic or eucaryotic cell or host organism.

When applied to RNA, the term “isolated nucleic acid” refers primarily to an RNA molecule encoded by an isolated DNA molecule as defined above. Alternatively, the term may refer to an RNA molecule that has been sufficiently separated from other nucleic acids with which it would be associated in its natural state (i.e., in cells or tissues). An isolated nucleic acid (either DNA or RNA) may further represent a molecule produced directly by biological or synthetic means and separated from other components present during its production.

“Natural allelic variants”, “mutants” and “derivatives” of particular sequences of nucleic acids refer to nucleic acid sequences that are closely related to a particular sequence but which may possess, either naturally or by design, changes in sequence or structure. By closely related, it is meant that at least about 75%, but often, more than 90%, of the-nucleotides of the sequence match over the defined length of the nucleic acid sequence referred to using a specific SEQ ID NO. Changes or differences in nucleotide sequence between closely related nucleic acid sequences may represent nucleotide changes in the sequence that arise during the course of normal replication or duplication in nature of the particular nucleic acid sequence. Other changes may be specifically designed and introduced into the sequence for specific purposes, such as to change an amino acid codon or sequence in a regulatory region of the nucleic acid. Such specific changes may be made in vitro using a variety of mutagenesis techniques or produced in a host organism placed under particular selection conditions that induce or select for the changes. Such sequence variants generated specifically may be referred to as “mutants” or “derivatives” of the original sequence.

The terms “percent similarity”, “percent identity” and “percent homology” when referring to a particular sequence are used as set forth in the University of Wisconsin GCG software program.

A “fragment” or “portion” of the FVIII recombinant polypeptide means a stretch of amino acid residues of at least about five to seven contiguous amino acids, often at least about seven to nine contiguous amino acids, typically at least about nine to thirteen contiguous amino acids and, most preferably, at least about twenty to thirty or more contiguous amino acids.

A “derivative” of the FVIII recombinant polypeptide or a fragment thereof means a polypeptide modified by varying the amino acid sequence of the protein, e.g. by manipulation of the nucleic acid encoding the protein or by altering the protein itself. Such derivatives of the natural amino acid sequence may involve insertion, addition, deletion or substitution of one or more amino acids, and may or may not alter the essential activity of original the FVIII polypeptide.

As mentioned above, the FVIII recombinant polypeptide or protein of the invention includes any analogue, fragment, derivative or mutant which is derived from a FVIII recombinant polypeptide and which retains at least one property or other characteristic of the FVIII recombinant polypeptide. Different “variants” of the FVIII recombinant polypeptide may be generated. These variants may be alleles characterized by differences in the nucleotide sequences of the gene coding for the protein, or may involve different RNA processing or post-translational modifications. A skilled person can produce variants having single or multiple amino acid substitutions, deletions, additions or replacements. These variants may include inter alia: (a) variants in which one or more amino acids residues are substituted with conservative or non-conservative amino acids, (b) variants in which one or more amino acids are added to the FVIII recombinant polypeptide, (c) variants in which one or more amino acids include a substituent group, and (d) variants in which the FVIII recombinant polypeptide is fused with another peptide or polypeptide such as a fusion partner, a protein tag or other chemical moiety, that may confer useful properties to the FVIII recombinant polypeptide, such as, for example, an epitope for an antibody, a polyhistidine sequence, a biotin moiety and the like.

To the extent such allelic variations, analogues, fragments, derivatives, mutants, and modifications, including alternative nucleic acid processing forms and alternative post-translational modification forms result in derivatives of the FVIII recombinant polypeptide that retain any of the biological properties of the FVIII polypeptide, they are included within the scope of this invention.

The term “functional” as used herein implies that the nucleic or amino acid sequence is functional for the recited assay or purpose.

The phrase “consisting essentially of” when referring to a particular nucleotide or amino acid means a sequence having the properties of a given SEQ ID No:. For example, when used in reference to an amino acid sequence, the phrase includes the sequence per se and molecular modifications that would not affect the functional and novel characteristics of the sequence.

A “replicon” is any genetic element, for example, a plasmid, cosmid, bacmid, phage or virus, that is capable of replication largely under its own control. A replicon may be either RNA or DNA and may be single or double stranded.

A “vector” is a replicon, such as a plasmid, cosmid, bacmid, phage or virus, to which another genetic sequence or element (either DNA or RNA) may be attached so as to bring about the replication of the attached sequence or element. Exemplary vectors of the invention include without limitation, adenoviral-based vectors, adeno-associated viral vectors, retroviral vectors, and transposon-transposase vector systems (Cell 91:501-10, 1991; Nature Genetics 25:35-41, 2000).

An “expression operon” refers to a nucleic acid segment that may possess transcriptional and translational control sequences, such as promoters, enhancers, translational start signals (e.g., ATG or AUG codons), polyadenylation signals, terminators, and the like, and which facilitate the expression of a polypeptide coding sequence in a host cell or organism.

The term “oligonucleotide,” as used herein refers to primers and probes of the present invention, and is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide.

The term “probe” as used herein refers to an oligonucleotide, polynucleotide or nucleic acid, either RNA or DNA, whether occurring naturally as in a purified restriction enzyme digest or produced synthetically, which is capable of annealing with or specifically hybridizing to a nucleic acid with sequences complementary to the probe. A probe may be either single-stranded or double-stranded. The exact length of the probe will depend upon many factors, including temperature, source of probe and use of the method. For example, for diagnostic applications, depending on the complexity of the target sequence, the oligonucleotide probe typically contains 15-25 or more nucleotides, although it may contain fewer nucleotides. The probes herein are selected to be “substantially” complementary to different strands of a particular target nucleic acid sequence. This means that the probes must be sufficiently complementary so as to be able to “specifically hybridize” or anneal with their respective target strands under a set of pre-determined conditions. Therefore, the probe sequence need not reflect the exact complementary sequence of the target. For example, a non-complementary nucleotide fragment may be attached to the 5′ or 3′ end of the probe, with the remainder of the probe sequence being complementary to the target strand. Alternatively, non-complementary bases or longer sequences can be interspersed into the probe, provided that the probe sequence has sufficient complementarity with the sequence of the target nucleic acid to anneal therewith specfically.

The term “specifically hybridize” refers to the association between two single-stranded nucleic acid molecules of sufficiently complementary sequence to permit such hybridization under pre-determined conditions generally used in the art (sometimes termed “substantially complementary”). In particular, the term refers to hybridization of an oligonucleotide with a substantially complementary sequence contained within a single-stranded DNA or RNA molecule of the invention, to the substantial exclusion of hybridization of the oligonucleotide with single-stranded nucleic acids of non-complementary sequence.

The term “primer” as used herein refers to an oligonucleotide, either RNA or DNA, either single-stranded or double-stranded, either derived from a biological system, generated by restriction enzyme digestion, or produced synthetically which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis. When presented with an appropriate nucleic acid template, suitable nucleoside triphosphate precursors of nucleic acids, a polymerase enzyme, suitable cofactors and conditions such as a suitable temperature and pH, the primer may be extended at its 3′ terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield an primer extension product. The primer may vary in length depending on the particular conditions and requirement of the application. For example, in diagnostic applications, the oligonucleotide primer is typically 15-25 or more nucleotides in length. The primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able to anneal with the desired template strand in a manner sufficient to provide the 3′ hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template. For example, a non-complementary nucleotide sequence may be attached to the 5′ end of an otherwise complementary primer. Alternatively, non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product. Amino acid residues described herein are preferred to be in the “L” isomeric form. However, residues in the “D” isomeric form may be substituted for any L-amino acid residue, provided the desired properties of the polypeptide are retained.

All amino-acid residue sequences represented herein conform to the conventional left-to-right amino-terminus to carboxy-terminus orientation.

The term “isolated protein” or “isolated and purified protein” is sometimes used herein. This term refers primarily to a protein produced by expression of an isolated nucleic acid molecule of the invention. Alternatively, this term may refer to a protein that has been sufficiently separated from other proteins with which it would naturally be associated, so as to exist in “substantially pure” form. “Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, addition of stabilizers, or compounding into, for example, immunogenic preparations or pharmaceutically acceptable preparations.

The term “substantially pure” refers to a preparation comprising at least 50-60% by weight of a given material (e.g., nucleic acid, oligonucleotide, protein, etc.). More preferably, the preparation comprises at least 75% by weight, and most preferably 90-95% by weight of the given compound. Purity is measured by methods appropriate for the given compound (e.g. chromatographic methods, agarose or polyacrylamide gel electrophoresis, HPLC analysis, and the like). “Mature protein” or “mature polypeptide” shall mean a polypeptide possessing the sequence of the polypeptide after any processing events that normally occur to the polypeptide during the course of its genesis, such as proteolytic processing from a polyprotein precursor. In designating the sequence or boundaries of a mature protein, the first amino of the mature protein sequence is designated as amino acid residue 1.

The term “tag,” “tag sequence” or “protein tag” refers to a chemical moiety, either a nucleotide, oligonucleotide, polynucleotide or an amino acid, peptide or protein or other chemical, that when added to another sequence, provides additional utility or confers useful properties, particularly in the detection or isolation, to that sequence. Thus, for example, a homopolymer nucleic acid sequence or a nucleic acid sequence complementary to a capture oligonucleotide may be added to a primer or probe sequence to facilitate the subsequent isolation of an extension product or hybridized product. In the case of protein tags, histidine residues (e.g., 4 to 8 consecutive histidine residues) may be added to either the amino- or carboxy-terminus of a protein to facilitate protein isolation by chelating metal chromatography. Alternatively, amino acid sequences, peptides, proteins or fusion partners representing epitopes or binding determinants reactive with specific antibody molecules or other molecules (e.g., flag epitope, c-myc epitope, transmembrane epitope of the influenza A virus hemaglutinin protein, protein A, cellulose binding domain, calmodulin binding protein, maltose binding protein, chitin binding domain, glutathione S-transferase, and the like) may be added to proteins to facilitate protein isolation by procedures such as affinity or immunoaffinity chromatography. Chemical tag moieties include such molecules as biotin, which may be added to either nucleic acids or proteins and facilitates isolation or detection by interaction with avidin reagents, and the like. Numerous other tag moieties are known to, and can be envisioned by, the trained artisan, and are contemplated to be within the scope of this definition.

As used herein, the terms “reporter,” “reporter system”, “reporter gene,” or “reporter gene product” shall mean an operative genetic system in which a nucleic acid comprises a gene that encodes a product that when expressed produces a reporter signal that is a readily measurable, e.g., by biological assay, immunoassay, radioimmunoassay, or by calorimetric, fluorogenic, chemiluminescent or other methods. The nucleic acid may be either RNA or DNA, linear or circular, single or double stranded, antisense or sense polarity, and is operatively linked to the necessary control elements for the expression of the reporter gene product. The required control elements will vary according to the nature of the reporter system and whether the reporter gene is in the form of DNA or RNA, but may include, but not be limited to, such elements as promoters, enhancers, translational control sequences, poly A addition signals, transcriptional termination signals and the like.

The terms “transform”, “transfect”, “transduce”, shall refer to any method or means by which a nucleic acid is introduced into a cell or host organism and may be used interchangeably to convey the same meaning. Such methods include, but are not limited to, transfection, electroporation, microinjection, PEG-fusion and the like.

The introduced nucleic acid may or may not be integrated (covalently linked) into nucleic acid of the recipient cell or organism. In bacterial, yeast, plant and mammalian cells, for example, the introduced nucleic acid may be maintained as an episomal element or independent replicon such as a plasmid. Alternatively, the introduced nucleic acid may become integrated into the nucleic acid of the recipient cell or organism and be stably maintained in that cell or organism and further passed on or inherited to progeny cells or organisms of the recipient cell or organism. In other manners, the introduced nucleic acid may exist in the recipient cell or host organism only transiently.

A “clone” or “clonal cell population” is a population of cells derived from a single cell or common ancestor by mitosis.

A “cell line” is a clone of a primary cell or cell population that is capable of stable growth in vitro for many generations.

II. Preparation of FVIII-Encoding Nucleic Acid Molecules, FVIII Proteins, and Antibodies Thereto

A. Nucleic Acid Molecules

Nucleic acid molecules encoding the recombinant FVIII of the invention may be prepared by two general methods: (1) Synthesis from appropriate nucleotide triphosphates, or (2) recombinant cloning of nucleic acid sequences encoding modified FVIII. Both methods utilize protocols well known in the art.

The availability of nucleotide sequence information, such as cDNA having the sequence of SEQ ID NO: 1 or segments thereof, enables preparation of an isolated nucleic acid molecule of the invention by oligonucleotide synthesis. Synthetic oligonucleotides may be prepared by the phosphoramidite method employed in the Applied Biosystems 38A DNA Synthesizer or similar devices. The resultant construct may be purified according to methods known in the art, such as high performance liquid chromatography (HPLC). Long, double-stranded polynucleotides, such as a DNA molecule of the present invention, must be synthesized in stages, due to the size limitations inherent in current oligonucleotide synthetic methods. Thus, for example, a 2.4 kb double-stranded molecule may be synthesized as several smaller segments of appropriate complementarity. Complementary segments thus produced may be annealed such that each segment possesses appropriate cohesive termini for attachment of an adjacent segment. Adjacent segments may be ligated by annealing cohesive termini in the presence of DNA ligase to construct an entire 2.4 kb double-stranded molecule. A synthetic DNA molecule so constructed may then be cloned and amplified in an appropriate vector.

Nucleic acid sequences encoding FVIII may be isolated from appropriate biological sources using methods known in the art and then modified in accordance with the teachings in the present specification. In a preferred embodiment, a cDNA clone is isolated from a cDNA expression library of human origin.

FVIII-encoding nucleic acid molecules of the invention include cDNA, genomic DNA, RNA, and fragments thereof which may be single- or double-stranded. Thus, this invention provides oligonucleotides (sense or antisense strands of DNA or RNA) having sequences capable of hybridizing with at least one sequence of a nucleic acid molecule of the present invention, such as selected segments of the cDNA having SEQ ID NO: 1. Such oligonucleotides are useful as probes for detecting or isolating FVIII genes.

Nucleic acid according to the present invention may be used in methods of gene therapy, for instance in treatment of individuals with the aim of preventing or curing (wholly or partially) Hemophilia A. This too is discussed below.

B. Proteins

FVIII protein functions as a co-factor in the blood coagulation system. A full-length FVIII protein of the present invention may be prepared in a variety of ways, according to known methods. The protein may be purified from appropriate sources, e.g., transformed bacterial or animal cultured cells or tissues, by immunoaffinity purification.

The availability of nucleic acid molecules encoding FVIII enables production of the protein using in vitro expression methods known in the art. For example, a cDNA or gene may be cloned into an appropriate in vitro transcription vector, such as pSP64 or pSP65 for in vitro transcription, followed by cell-free translation in a suitable cell-free translation system, such as wheat germ or rabbit reticulocyte lysates. In vitro transcription and translation systems are commercially available, e.g., from Promega Biotech, Madison, Wis. or BRL, Rockville, Md.

Alternatively, according to a preferred embodiment, larger quantities of FVIII may be produced by expression in a suitable procaryotic or eucaryotic system. For example, part or all of a DNA molecule, such as the cDNA having SEQ ID NO: 1 or segments thereof, may be inserted into a plasmid vector adapted for expression in a bacterial cell, such as E. coli. Such vectors comprise the regulatory elements necessary for expression of the DNA in the host cell (e.g. E. coli) positioned in such a manner as to permit expression of the DNA in the host cell. Such regulatory elements required for expression include promoter sequences, transcription initiation sequences and, optionally, enhancer sequences.

The FVIII produced by gene expression in a recombinant procaryotic or eucaryotic system may be purified according to methods known in the art. In a preferred embodiment, a commercially available expression/secretion system can be used, whereby the recombinant protein is expressed and thereafter secreted from the host cell, to be easily purified from the surrounding medium. If expression/secretion vectors are not used, an alternative approach involves purifying the recombinant protein by affinity separation, such as by immunological interaction with antibodies that bind specifically to the recombinant protein or nickel columns for isolation of recombinant proteins tagged with 6-8 histidine residues at their N-terminus or C-terminus. Alternative tags may comprise the FLAG epitope or the hemagglutinin epitope. Such methods are commonly used by skilled practitioners.

As discussed above, a convenient way of producing a polypeptide according to the present invention is to express nucleic acid encoding it, by use of the nucleic acid in an expression system. The use of expression systems has reached an advanced degree of sophistication today.

Accordingly, the present invention also encompasses a method of making a polypeptide (as disclosed), the method including expression from nucleic acid encoding the polypeptide (generally nucleic acid according to the invention). This may conveniently be achieved by growing a host cell in culture, containing such a vector, under appropriate conditions which cause or allow production of the polypeptide. Polypeptides may also be produced in in vitro systems, such as reticulocyte lysate.

Polypeptides which are amino acid sequence variants, alleles, derivatives or mutants are also provided by the present invention. A polypeptide which is a variant, allele, derivative, or mutant may have an amino acid sequence that differs from that given in SEQ ID NO: 2 by one or more of addition, substitution, deletion and insertion of one or more amino acids. Preferred such polypeptides have FVIII function.

A polypeptide which is an amino acid sequence variant, allele, derivative or mutant of the amino acid sequence shown in SEQ ID NO: 2 may comprise an amino acid sequence which shares greater than about 35% sequence identity with the sequence shown, greater than about 40%, greater than about 50%, greater than about 60%, greater than about 70%, greater than about 80%, greater than about 90% or greater than about 95%. Particular amino acid sequence variants may differ from that shown in SEQ ID NO: 2 by insertion, addition, substitution or deletion of 1 amino acid, 2, 3, 4, 5-10, 10-20, 20-30, 30-40, 40-50, 50-100, 100-150, or more than 150 amino acids.

A polypeptide according to the present invention may be used in screening for molecules which affect or modulate its activity or function. Such molecules may be useful in a therapeutic (possibly including prophylactic) context.

III. Uses of Recombinant FVIII-Encoding Nucleic Acids, and Modified FVIII Proteins

The nucleic acids encoding the recombinant FVIII of the invention may be used in gene therapy protocols for the treatment of Hemophilia A. The improved construct encoding FVIII can be inserted into the appropriate gene therapy vector and administered to a patient to correct FVIII deficiency.

Vectors, such as viral vectors have been used in the prior art to introduce genes into a wide variety of different target cells. Typically the vectors are exposed to the target cells so that transformation can take place in a sufficient proportion of the cells to provide a useful therapeutic or prophylactic effect from the expression of the desired polypeptide (e.g., FVIII). The transfected nucleic acid may be permanently incorporated into the genome of each of the targeted cells, providing long lasting effect, or alternatively the treatment may have to be repeated periodically.

A variety of vectors, both viral vectors and plasmid vectors are known in the art, see U.S. Pat. No. 5,252,479 and WO 93/07282. In particular, a number of viruses have been used as gene transfer vectors, including papovaviruses, such as SV40, vaccinia virus, herpes viruses including HSV and EBV, and retroviruses. Many gene therapy protocols in the prior art have employed disabled murine retroviruses.

Several recently issued patents are directed to methods and compositions for performing gene therapy. See U.S. Pat. Nos. 6,168,916; 6,135,976; 5,965,541 and 6,129,705. Each of the foregoing patents is incorporated by reference herein.

In another aspect of the invention, the variant FVIII polypeptide may be administered to patients. Pharmaceutical compositions containing variant FVIII, 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,

phosphatidylserine/phosphatidylcholine or other compositions of phospholipids or detergents that together impart a negative charge to the surface, since FVIII binds to negatively charged phospholipid membranes. Liposomes may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the variant FVIII 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 variant FVIII 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 (vWf) or a fragment of vWf that contains the FVIII binding site, and polysaccharides such as sucrose.

IV. Therapeutics

As mentioned previously, the FVIII-encoding nucleic acids or polypeptides/proteins, of the invention can be formulated in pharmaceutical compositions. These compositions may comprise, in addition to one of the above substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material may depend on the route of administration, e.g. oral, intravenous, cutaneous or subcutaneous, nasal, intramuscular, intraperitoneal routes.

Whether it is a polypeptide, or nucleic acid molecule, according to the present invention that is to be given to an individual, administration is preferably in a “prophylactically effective amount” or a “therapeutically effective amount” (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual.

The treatment dosage of variant FVIII composition that must be administered to a patient in need of such treatment will vary depending on the severity of the FVIII 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 variant FVIII is included in the pharmaceutically acceptable carrier, delivery vehicle, or stabilizer in an amount sufficient to deliver to patient a therapeutically effective amount of the variant protein to stop bleeding, as measured by standard clotting assays.

FVIII is classically defined as the substance present in normal blood plasma that corrects the clotting defect in plasma derived from individuals with hemophilia A. The coagulant activity in vitro of purified and partially-purified forms of FVIII is used to calculate the dose of FVIII 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 FVIII 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. 328:453459; Pittman, D. D. et al. (1992) Blood 79:389-397; and Brinkhous et al. (1985) Proc. Natl. Acad. Sci. 82:8752-8755.

Usually, the described plasma FVIII level to be achieved in the patient through administration of the variant FVIII is in the range of 30-100% of normal. In a preferred mode of administration of the variant FVIII, 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 (Facor II, Factor V, an Factors VII to XII),” ch. 153, 1453-1474, 1460, in Hematology, Williams, W. J., et al., ed. (1990). As in treatment with human or porcine FVIII, the amount of variant FVIII infused is defined by the one-stage FVIII coagulation assay and, in selected instances, in vivo recovery is determined by measuring the FVIII 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 individual 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 of practice of the claimed composition.

Treatment can take the form of a single intravenous administration of the composition or periodic or continuous administration over an extended period of time, as required. Alternatively, variant FVIII can be administered subcutaneously or orally with liposomes in one or several doses at varying intervals of time.

The following example is provided to illustrate certain embodiments of the invention. It is not intended to limit the invention in any way.

EXAMPLE I Novel Minigene Encoding Improved Factor VIII

In accordance with the present invention, new recombinant Factor VIII minigenes are provided which stimulate production of Factor VIII in cells comprising the minigene with higher activity than prior art recombinant constructs.

Native FVIII is synthesized as a single chain polypeptide (2332 amino acids) preceded by a 19-residue signal sequence and has a molecular weight of Mr=330,000. The signal sequence is removed upon translocation of FVIII into the ER and the native FVIII is then cleaved in the B-domain in connection with its secretion. This results in the release of a heterodimer comprised of a Mr=200,000 heavy chain and a Mr=80,000 light chain, the association of which is metal-ion dependent. See FIG. 1.

Creation of the FVIII Minigenes:

The FVIII cDNA in the expression plasmid pSP64 was purchased from the ATCC. Following a Sal I restriction digest, the FVIII cDNA was subcloned into the mammalian expression plasmid, pED. To fuse the heavy chain to the light chain with a furin-like cleavage site in the middle, the following specific oligonucleotide primers were constructed for rFVIII-RKR2 (a similar set of primers were used for rFVIII-R4): forward primer A, 5′-CCACTTTGCCTTTCTCTCCACAGG-3′, (SEQ ID NO: 6), which corresponds to a region in the pED plasmid which is just 5′-prime to the FVIII cDNA; reverse primer B, 5′-GCTTTCTACGCTTTCTTCTTGGTTCAATGGCATT-3′ (SEQ ID NO: 7), in which the first 15 bp correspond to a RKRRKR sequence (SEQ ID NO: 5) and the remaining bp correspond to FVIII gene sequence coding for residues 740-735 of FVIII polypeptide (SEQ ID NO: 2); forward primer C, 5′-AGAAAGCGTAGAAAGCGCAGCTTTCAAAAGAAAACA-3′ (SEQ ID NO: 8), in which the first 18 bp correspond to a RKRRKR sequence (SEQ ID NO: 5) and the remaining bp correspond to FVIII gene sequence coding for residues 1690-1695 of FVIII polypeptide (SEQ ID NO: 2); and reverse primer D, 5′-CTCTTTTTTTCGTACGGTGAACAC-3′ (SEQ ID NO: 9), in which the underlined portion is a BsiWI restriction site and the 24 bp correspond to FVIII gene sequence coding for residues 1969-1962 of FVIII polypeptide (SEQ ID NO: 2). The DNA sequence encoding the heavy and light chains along with the furin cleavage sites were ligated together by the technique of splicing by overlap extension or “geneSOEing” where primers B and C are the SOEing primers and primers A and D are the outside primers. The resulting DNA fragment was digested with SpeI and BsiWI, gel purified, and subcloned into pED-FVIII, cut with the same enzymes to generated PEDFVIII-RKR2 (or pEDFVIII-R4). To confirm the presence of the desired construct and to ensure the absence of polymerase induced errors, DNA sequencing of the entire insert was performed. The plasmid was introduced into bacterial cells (DH5α strain of E. coli) for propagation and large amounts of the plasmid DNA was purified by standard techniques. The final constructs are shown schematically in FIG. 3. Thus for rFVIII-R4 and rFVIII-RKR2, we have: 1) amino acids 1-740 of SEQ ID NO: 2; 2) protease cleavage site, i.e., RRRR (SEQ ID NO: 4) or RKRRKR (SEQ ID NO: 5); and 3) amino acids 1690-2332 of SEQ ID NO: 2.

Similarly, minigenes coding for recombinant FVIII variants FVIII(Δ721-1689) and FVIII(Δ731-1689) were constructed. Recombinant FVIII (Δ721-1689) and FVIII (Δ731-1689) comprises 1) amino acids 1-720 or 1-730 of SEQ ID NO: 2; 2) protease cleavage site, i.e., RKRRKR (SEQ ID NO: 5); and 3) amino acids 1690-2332 of SEQ ID NO: 2.

FVIII is activated by thrombin through specific proteolytic cleavages in both the heavy and light chains. In the heavy chain, one cleavage occurs at amino acid Arg372 to generate separate A1 and A2 domains and another at Arg740 at the junction between the A2 domain and the B-domain resulting in the release of the B-domain. In the light chain there is a cleavage at Arg1689 in the acidic region at the amino-terminal of the A3 domain, whereby a new N-terminus is created. Cleavages at 372 and 1689 are necessary for full activation of FVIII. The 200+80 heterodimer is thus converted to a heterotrimer as shown in FIG. 1.

A problem in the packaging of the rFVIII cDNA or various B-domainless derivatives (i.e. rFVIII-SQ; ReFacto) in various AAV serotypes and other gene delivery vectors is the size of the insert, which when combined with required regulatory elements, often exceeds the packaging capacity of these vectors. Thus, development of rFVIII gene constructs with minimal lengths is a necessary step in successful gene transfer methods for the treatment of Hemophilia A.

The rFVIII-SQ construct, currently in use by several groups, is the shortest version of a FVIII gene currently reported. See FIG. 2. rFVIII-SQ is a deletion derivative of FVIII, lacking the major part of the central B-domain. The N- and C-terminal parts of the B-domain are retained and fused at ser743-Gln-1638. The molecular mass of rFVIII-SQ is 170 kDa, comprising a 90 kDa heavy chain and a 80 kDa light chain (in the cleaved, secreted form), which are non-covalently associated. rFVIII-SQ has structural and functional properties which are similar to those of plasma-derived FVIII (similar specific activity; theoretical specific activity of purified plasma-derived FVIII is 5000-6000 IU/mg assuming 1U/mL FVIII activity is equal to 1500-200 ng/mL). The product has been developed by Pharmacia AB, Biopharmaceuticals, Stockholm, Sweden and Genetics Institute under the ReFacto Trade name. Information on the product can be found in the following references: Lind, et al., (1995) Eur J. Biochem 232, 19-27; Berntorp, E., (1997) Thrombosis and Haemostasis 78, 256-260; and Sandberg, H., et al. (2001) Seminars in Hematology 38, 4-12. rFVIII-SQ is disclosed in U.S. Pat. No. 5,661,008 issued in Aug. 26, 1997 and assigned to Kabi Pharmacia AB (Upsala, SE).

rFVIII-SQ encodes a Factor VIII molecule which is efficiently processed in mammalian cells. The single chain molecule, following post-translational modification is cleaved at Arg1648, by an unknown PACE-furin-like enzyme and FVIII-SQ is secreted in the surrounding milieu as a heterodimer (A1-A2-B(partial)) and (B(partial)-A3-C1-C2), and this molecule is efficiently cleaved by thrombin (at Arg372 Arg740, and Arg1689) to yield biologically active rFVIIIa (heterotrimers; A1, A2, A3-C1-C2).

The present invention differs from the current technology in the following ways: 1) the constructs described herein encode FVIII variants that are ˜55-˜75 amino acids shorter, lacking the so-called acidic region, all of the B-domain, and in some cases part of the C-terminal of the A2 domain; 2) based on our findings the current invention has greater activity (˜5-13-fold) compared to rFVIII-SQ in a one-stage APTT clotting assay. See FIGS. 5-7 and Table I.

The invention represents a significant improvement over rFVIII-SQ. First, the constructs are ˜150-˜200 bp shorter than the cDNA sequence for rFVIII-SQ, thus allowing for more efficient packaging into, for example, AAV vectors, which may allow for the incorporation of enhancer elements or more efficient promoters into any gene delivery construct. Second, since the new constructs appear to have a greater specific activity in a one-stage APTT assay, potentially less protein may be needed to correct a prolonged EPTT.

The recombinant constructs are efficiently synthesized as a single chain molecule initially. The PACE-furin-like enzyme should efficiently remove the inserted protease cleavage site in the ER or Golgi. This gives rise to a two-chain or heterodimeric rFVIII which is secreted in the extracellular space. These two chains are held together by divalent metal ions.

We have introduced the modified FVIII constructs shown in FIG. 3 and 4 into COS-1 cells, BHK cells, and HepG2 cells, and the modified constructs (rFVIII-R4, rFVIII-RKR2, FVIII(Δ721-1689), and FVIII(Δ731-1689)) have enhanced activity compared to full length FVIII and rFVIII-SQ (FIGS. 5-7 and Table I). This may result from either 1) enhanced secretion; 2) partial cofactor activity upon secretion; or 3) enhanced processing by thrombin or other protease (since two out of the three thrombin cleavage sites have been eliminated or are essentially already processed). These novel rFVIII constructs only need to be cleaved by thrombin at Arg372 to become fully processed to FVIIIa; in contrast full length FVIII and FVIII-SQ need to be processed at 372, 740, and 1689 to yield the active cofactor FVIIIa.

Transient transfection data of rFVIII-SQ, rFVIII-R4, and rFVIII-RKR2 are shown in FIG. 5. Results are based on a one-stage FVIII-specific APTT clotting assay and values are expressed as ng/mL FVIII/day for each construct, where 150 ng/mL is equal to 1 IU/mL FVIII activity.

Stable cell lines were also made in BHK cells for rFVIII-SQ and rFVIII-RKR2. In this experiment, each of the FVIII constructs were introduced into BHK cells using a Neomycin gene as a selectable marker. Each stable cell line was placed in a T25 flask and when cells reached 95% confluency, the media was changed and FVIII activity and antigen levels were measured 24 hr later. Specific activities were calculated based on a one-stage APTT clotting assay and a FVIII-specific ELISA using ReFacto as a standard in both assays (FIG. 6). Based on these data, rFVIII-RKR2 has ˜7-8-fold increase in specific activity (IU/mg) based on a one-stage FVIII specific APTT clotting assay.

FIG. 7 shows the transient transfection data for rFVIII-SQ, rFVIII-RKR2, FVIII(Δ721-1689), and FVIII(Δ731-1689). Cos-1 cells (100 mm plates) were transiently transfected with various FVIII constructs (25 μg) using Lipofectamine-2000. At selected time points, media from each well was removed and FVIII activity was assessed by a one-stage FVIII-specific APTT using FVIII-deficient plasma. FVIII antigen levels were determined by FVIII specific ELISA. The data are expressed as specific activity, where 1 unit of FVIII activity is equal to 200 ng/mL. The cells were cultured in OPTIMEN supplemented with insulin-transferrin-selenite, CaCl2, and Albumax. Table I is a summary of the results from FIG. 7.

TABLE I Summary of Results from FIG. 7 Clotting ELISA Assay Specific Act. (ng/ml) (IU/mg) (IU/mg) SD FVIII-SQ 68.70 0.27 3938.19 100.27 FVIII-RKR2 6.50 0.13 20380.22 3903.9 FVIII(721-1789) 12.95 0.41 29614.63 1442.6 FVIII(731-1689) 7.08 0.37 1861.87 4551.6

Additional exemplary constructs of the invention include those set forth are described in Table II:

TABLE II Variant FVIII Polypeptides Heavy Chain and B-domain Deletions: 1-740 *(Δ 741-1689) 1690-2332 1-730 *(Δ 731-1689) 1690-2332 1-720 *(Δ 721-1689) 1690-2332 1-710 *(Δ 711-1689) 1690-2332 1-700 *(Δ 701-1689) 1690-2332 B-domain and Light Chain Deletions: 1-740 *(Δ 741-1699) 1700-2332 1-740 *(Δ 741-1709) 1710-2332 1-740 *(Δ 741-1719) 1720-2332 1-740 *(Δ 741-1729) 1730-2332 Heavy and Light Chain Deletions: 1-730 *(Δ 731-1699) 1700-2332 1-730 *(Δ 731-1709) 1710-2332 1-730 *(Δ 731-1719) 1720-2332 1-730 *(Δ 731-1729) 1730-2332 1-720 *(Δ 721-1699) 1700-2332 1-720 *(Δ 721-1709) 1710-2332 1-720 *(Δ 721-1719) 1720-2332 1-720 *(Δ 721-1729) 1730-2332 1-710 *(Δ 711-1699) 1700-2332 1-710 *(Δ 711-1709) 1710-2332 1-710 *(Δ 711-1719) 1720-2332 1-710 *(Δ 711-1729) 1730-2332 1-700 *(Δ 701-1699) 1700-2332 1-700 *(Δ 701-1709) 1710-2332 1-700 *(Δ 701-1719) 1720-2332 1-700 *(Δ 701-1729) 1730-2332
* Note in all constructs, the amino acid numbers are correspond to SEQ ID NO: 2. Between the asterisk indicates the presence of at least one PACE-furin or PACE furin-like cleavage site. Exemplary PACE-furin or PACE furin-like cleavage sites include, but are not limited to, RRRR (SEQ ID NO: 4) or RKRRKR (SEQ ID NO: 5).

While certain of the preferred embodiments of the present invention have been described and specifically exemplified above, it is not intended that the invention be limited to such embodiments. Various modifications may be made thereto without departing from the scope and spirit of the present invention, as set forth in the following claims.

Claims

1-10. (canceled)

11. A method of treating hemophilia in a patient in need thereof, comprising:

a) providing a vector comprising a nucleic acid sequence encoding a biologically active recombinant human factor VIII (FVIII), operably linked to expression control elements; and
b) administering said vector to said patient under conditions that result in the expression of a modified human FVIII protein at a level that provides a therapeutic effect in said patient.

12. The method of claim 11, wherein said vector is selected from the group consisting of adenoviral-based vectors, adeno-associated viral vectors, retroviral vectors, and transposon-transposase vector systems.

13. (canceled)

14. (canceled)

15. The method of claim 11, wherein said FVIII variant comprises two segments connected by at least one PACE-furin cleavage site, wherein said segments are selected from the group consisting of:

a) a first segment consisting of the sequence of amino acids 1 to 700 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1690 to 2332 of SEQ ID NO: 2;
b) a first segment consisting of the sequence of amino acids 1 to 710 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1690 to 2332 of SEQ ID NO: 2;
c) a first segment consisting of the sequence of amino acids 1 to 720 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1690 to 2332 of SEQ ID NO: 2;
d) a first segment consisting of the sequence of amino acids 1 to 730 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1690 to 2332 of SEQ ID NO: 2;
e) a first segment consisting of the sequence of amino acids 1 to 740 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1700 to 2332 of SEQ ID NO: 2;
f) a first segment consisting of the sequence of amino acids 1 to 740 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1710 to 2332 of SEQ ID NO: 2;
g) a first segment consisting of the sequence of amino acids 1 to 740 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1720 to 2332 of SEQ ID NO: 2;
h) a first segment consisting of the sequence of amino acids 1 to 740 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1730 to 2332 of SEQ ID NO: 2;
i) a first segment consisting of the sequence of amino acids 1 to 730 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1700 to 2332 of SEQ ID NO: 2;
j) a first segment consisting of the sequence of amino acids 1 to 730 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1710 to 2332 of SEQ ID NO: 2;
k) a first segment consisting of the sequence of amino acids 1 to 730 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1720 to 2332 of SEQ ID NO: 2;
l) a first segment consisting of the sequence of amino acids 1 to 730 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1730 to 2332 of SEQ ID NO: 2;
m) a first segment consisting of the sequence of amino acids 1 to 720 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1700 to 2332 of SEQ ID NO: 2;
n) a first segment consisting of the sequence of amino acids 1 to 720 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1710 to 2332 of SEQ ID NO: 2;
o) a first segment consisting of the sequence of amino acids 1 to 720 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1720 to 2332 of SEQ ID NO: 2;
p) a first segment consisting of the sequence of amino acids 1 to 720 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1730 to 2332 of SEQ ID NO: 2;
q) a first segment consisting of the sequence of amino acids 1 to 710 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1700 to 2332 of SEQ ID NO: 2;
r) a first segment consisting of the sequence of amino acids 1 to 710 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1710 to 2332 of SEQ ID NO: 2;
s) a first segment consisting of the sequence of amino acids 1 to 710 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1720 to 2332 of SEQ ID NO: 2;
t) a first segment consisting of the sequence of amino acids 1 to 710 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1730 to 2332 of SEQ ID NO: 2;
u) a first segment consisting of the sequence of amino acids 1 to 700 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1700 to 2332 of SEQ ID NO: 2;
v) a first segment consisting of the sequence of amino acids 1 to 700 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1710 to 2332 of SEQ ID NO: 2;
w) a first segment consisting of the sequence of amino acids 1 to 700 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1720 to 2332 of SEQ ID NO: 2; and
x) a first segment consisting of the sequence of amino acids 1 to 700 of SEQ ID NO: 2 and a second segment consisting of the sequence of amino acids 1730 to 2332 of SEQ ID NO: 2;
wherein said FVIII variant lacks amino acids 1648 to 1689 of SEQ ID NO: 2.
Patent History
Publication number: 20070166283
Type: Application
Filed: Mar 14, 2007
Publication Date: Jul 19, 2007
Inventors: Katherine High (Merion, PA), Rodney Camire (Voorhees, NJ)
Application Number: 11/686,162
Classifications
Current U.S. Class: 424/93.200; 514/44.000; 536/23.200; 530/383.000; 435/320.100
International Classification: A61K 48/00 (20060101); C07H 21/04 (20060101); C07K 14/755 (20060101);